SBMLBioModels: S - S

S


Shibeko2012 - Model of IIa generation: MODEL1808150001v0.0.1

Mathematical model of blood coagulation investigating the effects of varied rFVIIa and TF concentration.

Details

Recombinant factor VIIa (rFVIIa) is used for treatment of hemophilia patients with inhibitors, as well for off-label treatment of severe bleeding in trauma and surgery. Effective bleeding control requires supraphysiological doses of rFVIIa, posing both high expense and uncertain thrombotic risk. Two major competing theories offer different explanations for the supraphysiological rFVIIa dosing requirement: (1) the need to overcome competition between FVIIa and FVII zymogen for tissue factor (TF) binding, and (2) a high-dose-requiring phospholipid-related pathway of FVIIa action. In the present study, we found experimental conditions in which both mechanisms contribute simultaneously and independently to rFVIIa-driven thrombin generation in FVII-deficient human plasma. From mathematical simulations of our model of FX activation, which were confirmed by thrombin-generation experiments, we conclude that the action of rFVIIa at pharmacologic doses is dominated by the TF-dependent pathway with a minor contribution from a phospholipid-dependent mechanism. We established a dose-response curve for rFVIIa that is useful to explain dosing strategies. In the present study, we present a pathway to reconcile the 2 major mechanisms of rFVIIa action, a necessary step to understanding future dose optimization and evaluation of new rFVIIa analogs currently under development. link: http://identifiers.org/pubmed/22563088

Shimoni2009 - Escherichia Coli SOS: MODEL2937159804v0.0.1

Shimoni2009 - Escherichia Coli SOS Simple model, involving only the basic components of the circuit, sufficient to expl…

Details

BACKGROUND: DNA damage in Escherichia coli evokes a response mechanism called the SOS response. The genetic circuit of this mechanism includes the genes recA and lexA, which regulate each other via a mixed feedback loop involving transcriptional regulation and protein-protein interaction. Under normal conditions, recA is transcriptionally repressed by LexA, which also functions as an auto-repressor. In presence of DNA damage, RecA proteins recognize stalled replication forks and participate in the DNA repair process. Under these conditions, RecA marks LexA for fast degradation. Generally, such mixed feedback loops are known to exhibit either bi-stability or a single steady state. However, when the dynamics of the SOS system following DNA damage was recently studied in single cells, ordered peaks were observed in the promoter activity of both genes (Friedman et al., 2005, PLoS Biol. 3(7):e238). This surprising phenomenon was masked in previous studies of cell populations. Previous attempts to explain these results harnessed additional genes to the system and deployed complex deterministic mathematical models that were only partially successful in explaining the results. METHODOLOGY/PRINCIPAL FINDINGS: Here we apply stochastic methods, which are better suited for dynamic simulations of single cells. We show that a simple model, involving only the basic components of the circuit, is sufficient to explain the peaks in the promoter activities of recA and lexA. Notably, deterministic simulations of the same model do not produce peaks in the promoter activities. CONCLUSION/SIGNIFICANCE: We conclude that the double negative mixed feedback loop with auto-repression accounts for the experimentally observed peaks in the promoter activities. In addition to explaining the experimental results, this result shows that including additional regulations in a mixed feedback loop may dramatically change the dynamic functionality of this regulatory module. Furthermore, our results suggests that stochastic fluctuations strongly affect the qualitative behavior of important regulatory modules even under biologically relevant conditions, thus emphasizing the importance of stochastic analysis of regulatory circuits. link: http://identifiers.org/pubmed/19424504

Shin2016 - Unveiling Hidden Dynamics of Hippo Signalling: BIOMD0000000832v0.0.1

This is a mathematical model describing Hippo signalling pathway activity. It includes descriptions of crosstalk with th…

Details

The Hippo signalling pathway has recently emerged as an important regulator of cell apoptosis and proliferation with significant implications in human diseases. In mammals, the pathway contains the core kinases MST1/2, which phosphorylate and activate LATS1/2 kinases. The pro-apoptotic function of the MST/LATS signalling axis was previously linked to the Akt and ERK MAPK pathways, demonstrating that the Hippo pathway does not act alone but crosstalks with other signalling pathways to coordinate network dynamics and cellular outcomes. These crosstalks were characterised by a multitude of complex regulatory mechanisms involving competitive protein-protein interactions and phosphorylation mediated feedback loops. However, how these different mechanisms interplay in different cellular contexts to drive the context-specific network dynamics of Hippo-ERK signalling remains elusive. Using mathematical modelling and computational analysis, we uncovered that the Hippo-ERK network can generate highly diverse dynamical profiles that can be clustered into distinct dose-response patterns. For each pattern, we offered mechanistic explanation that defines when and how the observed phenomenon can arise. We demonstrated that Akt displays opposing, dose-dependent functions towards ERK, which are mediated by the balance between the Raf-1/MST2 protein interaction module and the LATS1 mediated feedback regulation. Moreover, Ras displays a multi-functional role and drives biphasic responses of both MST2 and ERK activities; which are critically governed by the competitive protein interaction between MST2 and Raf-1. Our study represents the first in-depth and systematic analysis of the Hippo-ERK network dynamics and provides a concrete foundation for future studies. link: http://identifiers.org/pubmed/27527217

Parameters:

NameDescription
Km_93 = 0.9015; kc_92 = 0.9203Reaction: iRaf1 => Raf1; RasGTP, Rate Law: compartment*kc_92*iRaf1*RasGTP/(Km_93+iRaf1)
Km_122 = 297.2; V_121 = 1027.0Reaction: RasGTP => RasGDP, Rate Law: compartment*V_121*RasGTP/(Km_122+RasGTP)
ka_41 = 0.4237; kd_41 = 1.226Reaction: aMST2 + RASSF1A => aMST2uRASSF1A, Rate Law: compartment*(ka_41*aMST2*RASSF1A-kd_41*aMST2uRASSF1A)
kd_31 = 0.6117Reaction: dMST2 => aMST2, Rate Law: compartment*kd_31*dMST2
Km_91 = 0.8821; V_91 = 2.071Reaction: Raf1 => iRaf1, Rate Law: compartment*V_91*Raf1/(Km_91+Raf1)
V_22 = 7511.0; Km_22 = 816.2Reaction: iMST2 => MST2, Rate Law: compartment*V_22*iMST2/(Km_22+iMST2)
V_81 = 2261.0; Km_81 = 0.08503Reaction: aLATS1 => LATS1, Rate Law: compartment*V_81*aLATS1/(Km_81+aLATS1)
Km_92 = 10.68; kc_91 = 0.1177Reaction: Raf1 => iRaf1; aLATS1, Rate Law: compartment*kc_91*aLATS1*Raf1/(Km_92+Raf1)
aEGFR = 500.0; Km_11 = 51.21; kc_11 = 0.001149Reaction: Akt => pAkt, Rate Law: compartment*kc_11*aEGFR*Akt/(Km_11+Akt)
Km_13 = 0.744; kc_12 = 0.717Reaction: Akt => pAkt; RasGTP, Rate Law: compartment*kc_12*Akt*RasGTP/(Km_13+Akt)
Km_101 = 457.5; V_101 = 994.8Reaction: aRaf1 => Raf1, Rate Law: compartment*V_101*aRaf1/(Km_101+aRaf1)
V_11 = 0.08687; Km_12 = 0.01497Reaction: pAkt => Akt, Rate Law: compartment*V_11*pAkt/(Km_12+pAkt)
Km_21 = 427.3; V_21 = 1414.0Reaction: aMST2 => MST2, Rate Law: compartment*V_21*aMST2/(Km_21+aMST2)
V_131 = 995.3; Km_132 = 151.0Reaction: ppERK => ERK, Rate Law: compartment*V_131*ppERK/(Km_132+ppERK)
ka_22 = 0.0684; kd_21 = 0.113Reaction: MST2 + RASSF1A => MST2uRASSF1A, Rate Law: compartment*(ka_22*MST2*RASSF1A-kd_21*MST2uRASSF1A)
Km_111 = 0.07678; V_111 = 254.7Reaction: ipRaf1 => aRaf1, Rate Law: compartment*V_111*ipRaf1/(Km_111+ipRaf1)
ka_71 = 28.12; kd_71 = 4.886E-4Reaction: iMST2 + iRaf1 => iRaf1uiMST2, Rate Law: compartment*(ka_71*iMST2*iRaf1-kd_71*iRaf1uiMST2)
V_102 = 317.3; Km_102 = 3.197Reaction: Raf1 => aRaf1, Rate Law: compartment*V_102*Raf1/(Km_102+Raf1)
kc_112 = 0.002742; Km_112 = 207.1Reaction: aRaf1 => ipRaf1; ppERK, Rate Law: compartment*kc_112*aRaf1*ppERK/(Km_112+aRaf1)
kc_131 = 5.342; Km_131 = 0.03676Reaction: ERK => ppERK; aRaf1, Rate Law: compartment*kc_131*aRaf1*ERK/(Km_131+ERK)
kc_21 = 6684.0; Km_23 = 8.313E-4Reaction: MST2 => iMST2; pAkt, Rate Law: compartment*kc_21*MST2*pAkt/(Km_23+MST2)
kc_82 = 2.93E-4; Km_83 = 22.26Reaction: LATS1 => aLATS1; aMST2uRASSF1A, Rate Law: compartment*kc_82*aMST2uRASSF1A*LATS1/(Km_83+LATS1)
aEGFR = 500.0; kc_121 = 0.2061; Km_121 = 120.5Reaction: RasGDP => RasGTP, Rate Law: compartment*kc_121*aEGFR*RasGDP/(Km_121+RasGDP)
ka_21 = 4472.0Reaction: MST2 => dMST2, Rate Law: compartment*ka_21*MST2^2
Km_51 = 6.708; V_51 = 5.688E-4Reaction: MST2uRASSF1A => aMST2uRASSF1A, Rate Law: compartment*V_51*MST2uRASSF1A/(Km_51+MST2uRASSF1A)
kc_81 = 6189.0; Km_82 = 3961.0Reaction: LATS1 => aLATS1; aMST2, Rate Law: compartment*kc_81*aMST2*LATS1/(Km_82+LATS1)

States:

NameDescription
MST2uRASSF1A[Ras Association Domain-Containing Protein 1; STE20-Like Serine/Threonine-Protein Kinase]
aMST2uRASSF1A[STE20-Like Serine/Threonine-Protein Kinase; Ras Association Domain-Containing Protein 1]
MST2[STE20-Like Serine/Threonine-Protein Kinase]
aMST2[STE20-Like Serine/Threonine-Protein Kinase]
Akt[AKT kinase]
iRaf1[RAF proto-oncogene serine/threonine-protein kinase]
ipRaf1[RAF proto-oncogene serine/threonine-protein kinase]
Raf1[RAF proto-oncogene serine/threonine-protein kinase]
aLATS1[serine/threonine-protein kinase LATS1]
LATS1[serine/threonine-protein kinase LATS1]
RasGDP[RAS Family Gene]
ppERK[Mitogen-activated protein kinase 3]
pAkt[AKT kinase]
dMST2[STE20-Like Serine/Threonine-Protein Kinase]
aRaf1[RAF proto-oncogene serine/threonine-protein kinase]
RasGTP[RAS Family Gene]
RASSF1A[Ras Association Domain-Containing Protein 1]
iMST2[STE20-Like Serine/Threonine-Protein Kinase]
ERK[Mitogen-activated protein kinase 3]
iRaf1uiMST2[RAF proto-oncogene serine/threonine-protein kinase; STE20-Like Serine/Threonine-Protein Kinase]

Shin2019- Regulation of nuclear factor of activated T-cells (NFAT): MODEL2003200003v0.0.1

A properly functioning immune system is vital for an organism's wellbeing. Immune tolerance is a critical feature of the…

Details

Dendritic cells are a promising immunotherapy tool for boosting an individual's antigen-specific immune response to cancer. We develop a mathematical model using differential and delay-differential equations to describe the interactions between dendritic cells, effector-immune cells, and tumor cells. We account for the trafficking of immune cells between lymph, blood, and tumor compartments. Our model reflects experimental results both for dendritic cell trafficking and for immune suppression of tumor growth in mice. In addition, in silico experiments suggest more effective immunotherapy treatment protocols can be achieved by modifying dose location and schedule. A sensitivity analysis of the model reveals which patient-specific parameters have the greatest impact on treatment efficacy. link: http://identifiers.org/pubmed/23516248

Shin2019- Regulation of nuclear factor of activated T-cells (NFAT): MODEL2003200002v0.0.1

A properly functioning immune system is vital for an organism's wellbeing. Immune tolerance is a critical feature of the…

Details

A properly functioning immune system is vital for an organism's wellbeing. Immune tolerance is a critical feature of the immune system that allows immune cells to mount effective responses against exogenous pathogens such as viruses and bacteria, while preventing attack to self-tissues. Activation-induced cell death (AICD) in T lymphocytes, in which repeated stimulations of the T-cell receptor (TCR) lead to activation and then apoptosis of T cells, is a major mechanism for T cell homeostasis and helps maintain peripheral immune tolerance. Defects in AICD can lead to development of autoimmune diseases. Despite its importance, the regulatory mechanisms that underlie AICD remain poorly understood, particularly at an integrative network level. Here, we develop a dynamic multi-pathway model of the integrated TCR signalling network and perform model-based analysis to characterize the network-level properties of AICD. Model simulation and analysis show that amplified activation of the transcriptional factor NFAT in response to repeated TCR stimulations, a phenomenon central to AICD, is tightly modulated by a coupled positive-negative feedback mechanism. NFAT amplification is predominantly enabled by a positive feedback self-regulated by NFAT, while opposed by a NFAT-induced negative feedback via Carabin. Furthermore, model analysis predicts an optimal therapeutic window for drugs that help minimize proliferation while maximize AICD of T cells. Overall, our study provides a comprehensive mathematical model of TCR signalling and model-based analysis offers new network-level insights into the regulation of activation-induced cell death in T cells. link: http://identifiers.org/pubmed/31337782

Shin_2018_EGFR-PYK2-c-Met interaction network_model: BIOMD0000000826v0.0.1

Systems modelling of the EGFR-PYK2-c-Met interaction network predicted and prioritized synergistic drug combinations for…

Details

Prediction of drug combinations that effectively target cancer cells is a critical challenge for cancer therapy, in particular for triple-negative breast cancer (TNBC), a highly aggressive breast cancer subtype with no effective targeted treatment. As signalling pathway networks critically control cancer cell behaviour, analysis of signalling network activity and crosstalk can help predict potent drug combinations and rational stratification of patients, thus bringing therapeutic and prognostic values. We have previously showed that the non-receptor tyrosine kinase PYK2 is a downstream effector of EGFR and c-Met and demonstrated their crosstalk signalling in basal-like TNBC. Here we applied a systems modelling approach and developed a mechanistic model of the integrated EGFR-PYK2-c-Met signalling network to identify and prioritize potent drug combinations for TNBC. Model predictions validated by experimental data revealed that among six potential combinations of drug pairs targeting the central nodes of the network, including EGFR, c-Met, PYK2 and STAT3, co-targeting of EGFR and PYK2 and to a lesser extent of EGFR and c-Met yielded strongest synergistic effect. Importantly, the synergy in co-targeting EGFR and PYK2 was linked to switch-like cell proliferation-associated responses. Moreover, simulations of patient-specific models using public gene expression data of TNBC patients led to predictive stratification of patients into subgroups displaying distinct susceptibility to specific drug combinations. These results suggest that mechanistic systems modelling is a powerful approach for the rational design, prediction and prioritization of potent combination therapies for individual patients, thus providing a concrete step towards personalized treatment for TNBC and other tumour types. link: http://identifiers.org/pubmed/29920512

Parameters:

NameDescription
PF396 = 0.0; kc11 = 0.321366; STAT3tot = 144.212; Ki3b = 1.0; Km11 = 20.6063Reaction: => pSTAT3; STAT3uStattic, pPYK2, Rate Law: rootCompartment*kc11*pPYK2*rootCompartment/(1+PF396/Ki3b)*((STAT3tot-pSTAT3*rootCompartment)-STAT3uStattic*rootCompartment)/(Km11+((STAT3tot-pSTAT3*rootCompartment)-STAT3uStattic*rootCompartment))/rootCompartment
Vmax24 = 4.39542E9; Km24 = 0.156675Reaction: pERK =>, Rate Law: rootCompartment*Vmax24*pERK*rootCompartment/(Km24+pERK*rootCompartment)/rootCompartment
Km17 = 9.81748; HGF = 0.0; kc17 = 8.10961E-4; caHGF = 0.0090365Reaction: cMET => pcMET, Rate Law: rootCompartment*(kc17*HGF+caHGF)*cMET*rootCompartment/(Km17+cMET*rootCompartment)/rootCompartment
Vmax22 = 0.034914; Km22 = 46.4515Reaction: aPTP =>, Rate Law: rootCompartment*Vmax22*aPTP*rootCompartment/(Km22+aPTP*rootCompartment)/rootCompartment
Stattictot = 0.0; ka25 = 127.35; STAT3tot = 144.212; kd25 = 11.749Reaction: => STAT3uStattic; pSTAT3, Rate Law: rootCompartment*(ka25*((STAT3tot-pSTAT3*rootCompartment)-STAT3uStattic*rootCompartment)*(Stattictot-STAT3uStattic*rootCompartment)-kd25*STAT3uStattic*rootCompartment)/rootCompartment
Vs13 = 0.0937562; Vmax13 = 0.354813; Km13 = 38.7258Reaction: => cMETm; pSTAT3, Rate Law: rootCompartment*(Vs13+Vmax13*pSTAT3*rootCompartment/(Km13+pSTAT3*rootCompartment))/rootCompartment
kdeg6 = 53.5797Reaction: PYK2m =>, Rate Law: rootCompartment*kdeg6*PYK2m*rootCompartment/rootCompartment
kdeg14 = 4.56037Reaction: cMETm =>, Rate Law: rootCompartment*kdeg14*cMETm*rootCompartment/rootCompartment
kc10 = 0.00610942; Vmax10 = 0.530884; Km10 = 9.14113Reaction: pPYK2 => PYK2; aPTP, Rate Law: rootCompartment*(Vmax10+kc10*aPTP*rootCompartment)*pPYK2*rootCompartment/(Km10+pPYK2*rootCompartment)/rootCompartment
EGF = 10.0; caEGF = 0.0891251; kc1 = 413.048; Km1 = 248.886; Gefitinib = 0.0; EGFRtot = 398.107; Ki1 = 1.0Reaction: => pEGFR; EGFRub, Rate Law: rootCompartment*kc1*(EGF/(1+Gefitinib/Ki1)+caEGF)*((EGFRtot-pEGFR*rootCompartment)-EGFRub*rootCompartment)/(Km1+((EGFRtot-pEGFR*rootCompartment)-EGFRub*rootCompartment))/rootCompartment
Vs5 = 26.5461; Km5 = 4.74242; Vmax5 = 34.0408Reaction: => PYK2m; pSTAT3, Rate Law: rootCompartment*(Vs5+Vmax5*pSTAT3*rootCompartment/(Km5+pSTAT3*rootCompartment))/rootCompartment
Vmax4 = 11.1173; Km4 = 90.7821Reaction: EGFRub =>, Rate Law: rootCompartment*Vmax4*EGFRub*rootCompartment/(Km4+EGFRub*rootCompartment)/rootCompartment
Km20 = 24.322; Vmax20 = 0.0483059; kc20 = 35.6451Reaction: pCbl => ; aPTP, Rate Law: rootCompartment*(Vmax20+kc20*aPTP*rootCompartment)*pCbl*rootCompartment/(Km20+pCbl*rootCompartment)/rootCompartment
Km7 = 3.33426; Vmax7 = 3.34965Reaction: => PYK2; PYK2m, Rate Law: rootCompartment*Vmax7*PYK2m*rootCompartment/(Km7+PYK2m*rootCompartment)/rootCompartment
EMD = 0.0; Km9 = 34.914; kc9a = 0.463447; kc9b = 0.988553; Ki9 = 1.65577Reaction: PYK2 => pPYK2; pEGFR, pcMET, Rate Law: rootCompartment*(kc9a*pEGFR*rootCompartment+kc9b*pcMET*rootCompartment/(1+EMD/Ki9))*PYK2*rootCompartment/(Km9+PYK2*rootCompartment)/rootCompartment
Km2 = 3.80189; Vmax2 = 112.202; kc2 = 1406.05Reaction: pEGFR => ; aPTP, Rate Law: rootCompartment*(Vmax2+kc2*aPTP*rootCompartment)*pEGFR*rootCompartment/(Km2+pEGFR*rootCompartment)/rootCompartment
kc16 = 1.1749; Km16 = 528.445; kdeg16 = 24.4906Reaction: cMET => ; pCbl, Rate Law: rootCompartment*(kdeg16+kc16*pCbl*rootCompartment)*cMET*rootCompartment/(Km16+cMET*rootCompartment)/rootCompartment
Km18 = 9.95405; Vmax18 = 0.0606736Reaction: pcMET => cMET, Rate Law: rootCompartment*Vmax18*pcMET*rootCompartment/(Km18+pcMET*rootCompartment)/rootCompartment
kc19 = 52.723; Km19 = 13.3045; Cbltot = 174.985Reaction: => pCbl; pEGFR, Rate Law: rootCompartment*kc19*pEGFR*rootCompartment*(Cbltot-pCbl*rootCompartment)/(Km19+(Cbltot-pCbl*rootCompartment))/rootCompartment
Km12 = 11.5878; kc12 = 2.89734E-4; Vmax12 = 7.63836Reaction: pSTAT3 => ; aPTP, Rate Law: rootCompartment*(Vmax12+kc12*aPTP*rootCompartment)*pSTAT3*rootCompartment/(Km12+pSTAT3*rootCompartment)/rootCompartment
kdeg8 = 0.0566239Reaction: PYK2 =>, Rate Law: rootCompartment*kdeg8*PYK2*rootCompartment/rootCompartment
EMD = 0.0; ERKtot = 166.725; kc23b = 8.43335E8; kc23a = 7.03072E9; Km23 = 2.83139; Ki23 = 13.4896Reaction: => pERK; pEGFR, pcMET, Rate Law: rootCompartment*(kc23a*pcMET*rootCompartment/(1+EMD/Ki23)+kc23b*pEGFR*rootCompartment)*(ERKtot-pERK*rootCompartment)/(Km23+(ERKtot-pERK*rootCompartment))/rootCompartment
Vmax15 = 91.4113; Km15 = 6.45654Reaction: => cMET; cMETm, Rate Law: rootCompartment*Vmax15*cMETm*rootCompartment/(Km15+cMETm*rootCompartment)/rootCompartment
kc3 = 10.7895; PF396 = 0.0; Ki3a = 0.0835603; Ki3b = 1.0; Vmax3 = 1.03753E-4; EGFRtot = 398.107; Km3 = 2.2856Reaction: => EGFRub; PYK2, pCbl, pEGFR, pPYK2, Rate Law: rootCompartment*(Vmax3+kc3*pCbl*rootCompartment)*((EGFRtot-pEGFR*rootCompartment)-EGFRub*rootCompartment)/(Km3+((EGFRtot-pEGFR*rootCompartment)-EGFRub*rootCompartment))*Ki3a/(Ki3a+(PYK2*rootCompartment+pPYK2*rootCompartment)/(1+PF396/Ki3b))/rootCompartment
kc21 = 0.00397192; PTPtot = 296.483; Km21 = 52.723Reaction: => aPTP; pEGFR, Rate Law: rootCompartment*kc21*pEGFR*rootCompartment*(PTPtot-aPTP*rootCompartment)/(Km21+(PTPtot-aPTP*rootCompartment))/rootCompartment

States:

NameDescription
pcMET[PR:P08581]
STAT3uStattic[signal transducer and activator of transcription 3; stattic]
pCbl[E3 Ubiquitin-Protein Ligase CBL]
pPYK2[Protein Tyrosine Kinase]
aPTP[Protein Tyrosine Phosphatase]
pSTAT3[signal transducer and activator of transcription 3]
cMET[PR:P08581]
PYK2m[Protein Tyrosine Kinase; messenger RNA]
PYK2[Protein Tyrosine Kinase]
pERK[mitogen-activated protein kinase]
pEGFR[epidermal growth factor receptor]
cMETm[PR:P08581; messenger RNA]
EGFRub[epidermal growth factor receptor; ubiquinated]

Shlomi2011 - Warburg effect, metabolic model: MODEL1105100000v0.0.1

Shlomi2011 - Warburg effect, metabolic modelUsing a genome-scale human metabolic network model accounting for stoichiome…

Details

The Warburg effect–a classical hallmark of cancer metabolism–is a counter-intuitive phenomenon in which rapidly proliferating cancer cells resort to inefficient ATP production via glycolysis leading to lactate secretion, instead of relying primarily on more efficient energy production through mitochondrial oxidative phosphorylation, as most normal cells do. The causes for the Warburg effect have remained a subject of considerable controversy since its discovery over 80 years ago, with several competing hypotheses. Here, utilizing a genome-scale human metabolic network model accounting for stoichiometric and enzyme solvent capacity considerations, we show that the Warburg effect is a direct consequence of the metabolic adaptation of cancer cells to increase biomass production rate. The analysis is shown to accurately capture a three phase metabolic behavior that is observed experimentally during oncogenic progression, as well as a prominent characteristic of cancer cells involving their preference for glutamine uptake over other amino acids. link: http://identifiers.org/pubmed/21423717

Shorten2007_SkeletalMuscleFatigue: MODEL0912160004v0.0.1

This a model from the article: A mathematical model of fatigue in skeletal muscle force contraction. Shorten PR, O'C…

Details

The ability for muscle to repeatedly generate force is limited by fatigue. The cellular mechanisms behind muscle fatigue are complex and potentially include breakdown at many points along the excitation-contraction pathway. In this paper we construct a mathematical model of the skeletal muscle excitation-contraction pathway based on the cellular biochemical events that link excitation to contraction. The model includes descriptions of membrane voltage, calcium cycling and crossbridge dynamics and was parameterised and validated using the response characteristics of mouse skeletal muscle to a range of electrical stimuli. This model was used to uncover the complexities of skeletal muscle fatigue. We also parameterised our model to describe force kinetics in fast and slow twitch fibre types, which have a number of biochemical and biophysical differences. How these differences interact to generate different force/fatigue responses in fast- and slow- twitch fibres is not well understood and we used our modelling approach to bring new insights to this relationship. link: http://identifiers.org/pubmed/18080210

Shrestha2010_HyperCalcemia_PTHresponse: BIOMD0000000277v0.0.1

This a model from the article: A mathematical model of parathyroid hormone response to acute changes in plasma ioniz…

Details

A complex bio-mechanism, commonly referred to as calcium homeostasis, regulates plasma ionized calcium (Ca(2+)) concentration in the human body within a narrow range which is crucial for maintaining normal physiology and metabolism. Taking a step towards creating a complete mathematical model of calcium homeostasis, we focus on the short-term dynamics of calcium homeostasis and consider the response of the parathyroid glands to acute changes in plasma Ca(2+) concentration. We review available models, discuss their limitations, then present a two-pool, linear, time-varying model to describe the dynamics of this calcium homeostasis subsystem, the Ca-PTH axis. We propose that plasma PTH concentration and plasma Ca(2+) concentration bear an asymmetric reverse sigmoid relation. The parameters of our model are successfully estimated based on clinical data corresponding to three healthy subjects that have undergone induced hypocalcemic clamp tests. In the first validation of this kind, with parameters estimated separately for each subject we test the model's ability to predict the same subject's induced hypercalcemic clamp test responses. Our results demonstrate that a two-pool, linear, time-varying model with an asymmetric reverse sigmoid relation characterizes the short-term dynamics of the Ca-PTH axis. link: http://identifiers.org/pubmed/20406649

Parameters:

NameDescription
alpha = 0.0569; t0 = 575.0; Ca0 = 1.22; Ca1 = 0.2624Reaction: Ca = piecewise(Ca0, time < t0, Ca0+Ca1*(1-exp((-alpha)*(time-t0)))), Rate Law: missing
lambda_Ca = 170.0; k = 9.8436755; lambda_1 = 0.0125Reaction: x1 = (k-lambda_Ca*x1)-lambda_1*x1, Rate Law: (k-lambda_Ca*x1)-lambda_1*x1
lambda_Ca = 170.0; lambda_2 = 0.5595Reaction: x2 = lambda_Ca*x1-lambda_2*x2, Rate Law: lambda_Ca*x1-lambda_2*x2

States:

NameDescription
x1[Parathyroid hormone]
x2[Parathyroid hormone]
Ca[calcium(2+); Calcium cation]

Shrestha2010_HypoCalcemia_PTHresponse: BIOMD0000000276v0.0.1

This a model from the article: A mathematical model of parathyroid hormone response to acute changes in plasma ioniz…

Details

A complex bio-mechanism, commonly referred to as calcium homeostasis, regulates plasma ionized calcium (Ca(2+)) concentration in the human body within a narrow range which is crucial for maintaining normal physiology and metabolism. Taking a step towards creating a complete mathematical model of calcium homeostasis, we focus on the short-term dynamics of calcium homeostasis and consider the response of the parathyroid glands to acute changes in plasma Ca(2+) concentration. We review available models, discuss their limitations, then present a two-pool, linear, time-varying model to describe the dynamics of this calcium homeostasis subsystem, the Ca-PTH axis. We propose that plasma PTH concentration and plasma Ca(2+) concentration bear an asymmetric reverse sigmoid relation. The parameters of our model are successfully estimated based on clinical data corresponding to three healthy subjects that have undergone induced hypocalcemic clamp tests. In the first validation of this kind, with parameters estimated separately for each subject we test the model's ability to predict the same subject's induced hypercalcemic clamp test responses. Our results demonstrate that a two-pool, linear, time-varying model with an asymmetric reverse sigmoid relation characterizes the short-term dynamics of the Ca-PTH axis. link: http://identifiers.org/pubmed/20406649

Parameters:

NameDescription
Ca1 = 0.1817; t0 = 575.0; Ca0 = 1.255; alpha = 0.0442Reaction: Ca = piecewise(Ca0, time < t0, Ca0-Ca1*(1-exp((-alpha)*(time-t0)))), Rate Law: missing
lambda_Ca = 170.0; k = 9.8436755; lambda_1 = 0.0125Reaction: x1 = (k-lambda_Ca*x1)-lambda_1*x1, Rate Law: (k-lambda_Ca*x1)-lambda_1*x1
lambda_Ca = 170.0; lambda_2 = 0.5595Reaction: x2 = lambda_Ca*x1-lambda_2*x2, Rate Law: lambda_Ca*x1-lambda_2*x2

States:

NameDescription
x1[Parathyroid hormone]
x2[Parathyroid hormone]
Ca[calcium(2+); Calcium cation]

Sible2007 - Mitotic cell cycle mecanism in Xenopus Laevis: BIOMD0000000942v0.0.1

Although not a traditional experimental "method," mathematical modeling can provide a powerful approach for investigatin…

Details

Although not a traditional experimental "method," mathematical modeling can provide a powerful approach for investigating complex cell signaling networks, such as those that regulate the eukaryotic cell division cycle. We describe here one modeling approach based on expressing the rates of biochemical reactions in terms of nonlinear ordinary differential equations. We discuss the steps and challenges in assigning numerical values to model parameters and the importance of experimental testing of a mathematical model. We illustrate this approach throughout with the simple and well-characterized example of mitotic cell cycles in frog egg extracts. To facilitate new modeling efforts, we describe several publicly available modeling environments, each with a collection of integrated programs for mathematical modeling. This review is intended to justify the place of mathematical modeling as a standard method for studying molecular regulatory networks and to guide the non-expert to initiate modeling projects in order to gain a systems-level perspective for complex control systems. link: http://identifiers.org/pubmed/17189866

Parameters:

NameDescription
KKa = 0.1; ka = 0.02Reaction: => Cdc25_phosphorylated; Cyclin_Cdk1_MPF, Cdc25_total, Rate Law: nuclear*ka*Cyclin_Cdk1_MPF*(Cdc25_total-Cdc25_phosphorylated)/((KKa+Cdc25_total)-Cdc25_phosphorylated)
k2 = 0.25Reaction: Cyclin_Cdk1_MPF =>, Rate Law: nuclear*k2*Cyclin_Cdk1_MPF
kh = 0.15; KKh = 0.01Reaction: IE_phosphorylated => ; ppase, Rate Law: nuclear*kh*ppase*IE_phosphorylated/(KKh+IE_phosphorylated)
kd = 0.13; KKd = 1.0Reaction: APC_active => ; ppase, Rate Law: nuclear*kd*ppase*APC_active/(KKd+APC_active)
KKc = 0.01; kc = 0.13Reaction: => APC_active; IE_phosphorylated, APC_total, Rate Law: nuclear*kc*IE_phosphorylated*(APC_total-APC_active)/((KKc+APC_total)-APC_active)
kwee = 1.0Reaction: Cyclin_Cdk1_MPF => Cyclin_Cdk1_preMPF; Wee1, Rate Law: nuclear*kwee*Cyclin_Cdk1_MPF
k3 = 0.005Reaction: Cyclin => Cyclin_Cdk1_MPF; Cdk1, Rate Law: nuclear*k3*Cdk1*Cyclin
k1 = 1.0Reaction: => Cyclin, Rate Law: nuclear*k1
k25 = 0.017Reaction: Cyclin_Cdk1_preMPF => Cyclin_Cdk1_MPF; Cdc25_phosphorylated, Rate Law: nuclear*k25*Cyclin_Cdk1_preMPF
kb = 0.1; KKb = 1.0Reaction: Cdc25_phosphorylated => ; ppase, Rate Law: nuclear*kb*ppase*Cdc25_phosphorylated/(KKb+Cdc25_phosphorylated)
ke = 0.02; KKe = 0.1Reaction: => Wee1_phosphorylated; Cyclin_Cdk1_MPF, Wee1_total, Rate Law: nuclear*ke*Cyclin_Cdk1_MPF*(Wee1_total-Wee1_phosphorylated)/((KKe+Wee1_total)-Wee1_phosphorylated)
KKf = 1.0; kf = 0.1Reaction: Wee1_phosphorylated => ; ppase, Rate Law: nuclear*kf*ppase*Wee1_phosphorylated/(KKf+Wee1_phosphorylated)
KKg = 0.01; kg = 0.02Reaction: => IE_phosphorylated; Cyclin_Cdk1_MPF, IE_total, Rate Law: nuclear*kg*Cyclin_Cdk1_MPF*(IE_total-IE_phosphorylated)/((KKg+IE_total)-IE_phosphorylated)

States:

NameDescription
Wee1[Wee1-like protein kinase 1-B]
APC active[Adenomatous polyposis coli homolog; active]
Cyclin Cdk1 MPF[G2/mitotic-specific cyclin-B1; Cyclin-dependent kinase 1-A]
Wee1 phosphorylated[Wee1-like protein kinase 1-B; phosphorylated]
IEIE
Cdc25[M-phase inducer phosphatase 1-B]
Cdc25 phosphorylated[M-phase inducer phosphatase 1-B; phosphorylated]
Cyclin[G2/mitotic-specific cyclin-B1]
Cyclin total[G2/mitotic-specific cyclin-B1]
IE phosphorylated[phosphorylated]
Cdk1[Cyclin-dependent kinase 1-A]
Cyclin Cdk1 preMPF[G2/mitotic-specific cyclin-B1; Cyclin-dependent kinase 1-A; phosphorylated]

Siebert2008_MuscleContraction_CC: MODEL1006230119v0.0.1

This a model from the article: Nonlinearities make a difference: comparison of two common Hill-type models with real m…

Details

Compared to complex structural Huxley-type models, Hill-type models phenomenologically describe muscle contraction using only few state variables. The Hill-type models dominate in the ever expanding field of musculoskeletal simulations for simplicity and low computational cost. Reasonable parameters are required to gain insight into mechanics of movement. The two most common Hill-type muscle models used contain three components. The series elastic component is connected in series to the contractile component. A parallel elastic component is either connected in parallel to both the contractile and the series elastic component (model [CC+SEC]), or is connected in parallel only with the contractile component (model [CC]). As soon as at least one of the components exhibits substantial nonlinearities, as, e.g., the contractile component by the ability to turn on and off, the two models are mechanically different. We tested which model ([CC+SEC] or [CC]) represents the cat soleus better. Ramp experiments consisting of an isometric and an isokinetic part were performed with an in situ cat soleus preparation using supramaximal nerve stimulation. Hill-type models containing force-length and force-velocity relationship, excitation-contraction coupling and series and parallel elastic force-elongation relations were fitted to the data. To test which model might represent the muscle better, the obtained parameters were compared with experimentally determined parameters. Determined in situations with negligible passive force, the force-velocity relation and the series elastic component relation are independent of the chosen model. In contrast to model [CC+SEC], these relations predicted by model [CC] were in accordance with experimental relations. In conclusion model [CC] seemed to better represent the cat soleus contraction dynamics and should be preferred in the nonlinear regression of muscle parameters and in musculoskeletal modeling. link: http://identifiers.org/pubmed/18049823

Siebert2008_MuscleContraction_CCSEC: MODEL1006230120v0.0.1

This a model from the article: Nonlinearities make a difference: comparison of two common Hill-type models with real m…

Details

Compared to complex structural Huxley-type models, Hill-type models phenomenologically describe muscle contraction using only few state variables. The Hill-type models dominate in the ever expanding field of musculoskeletal simulations for simplicity and low computational cost. Reasonable parameters are required to gain insight into mechanics of movement. The two most common Hill-type muscle models used contain three components. The series elastic component is connected in series to the contractile component. A parallel elastic component is either connected in parallel to both the contractile and the series elastic component (model [CC+SEC]), or is connected in parallel only with the contractile component (model [CC]). As soon as at least one of the components exhibits substantial nonlinearities, as, e.g., the contractile component by the ability to turn on and off, the two models are mechanically different. We tested which model ([CC+SEC] or [CC]) represents the cat soleus better. Ramp experiments consisting of an isometric and an isokinetic part were performed with an in situ cat soleus preparation using supramaximal nerve stimulation. Hill-type models containing force-length and force-velocity relationship, excitation-contraction coupling and series and parallel elastic force-elongation relations were fitted to the data. To test which model might represent the muscle better, the obtained parameters were compared with experimentally determined parameters. Determined in situations with negligible passive force, the force-velocity relation and the series elastic component relation are independent of the chosen model. In contrast to model [CC+SEC], these relations predicted by model [CC] were in accordance with experimental relations. In conclusion model [CC] seemed to better represent the cat soleus contraction dynamics and should be preferred in the nonlinear regression of muscle parameters and in musculoskeletal modeling. link: http://identifiers.org/pubmed/18049823

Sier2017_E2_combined: MODEL1711210002v0.0.1

Using scaling from PhysB model Blood flow in L/hr Compartments in Kg Baseline as ~0.003nM Free E2 in Blood_venous E2 bi…

Details

Estrogen is a vital hormone that regulates many biological functions within the body. These include roles in the development of the secondary sexual organs in both sexes, plus uterine angiogenesis and proliferation during the menstrual cycle and pregnancy in women. The varied biological roles of estrogens in human health also make them a therapeutic target for contraception, mitigation of the adverse effects of the menopause, and treatment of estrogen-responsive tumours. In addition, endogenous (e.g. genetic variation) and external (e.g. exposure to estrogen-like chemicals) factors are known to impact estrogen biology. To understand how these multiple factors interact to determine an individual's response to therapy is complex, and may be best approached through a systems approach.We present a physiologically-based pharmacokinetic model (PBPK) of estradiol, and validate it against plasma kinetics in humans following intravenous and oral exposure. We extend this model by replacing the intrinsic clearance term with: a detailed kinetic model of estrogen metabolism in the liver; or, a genome-scale model of liver metabolism. Both models were validated by their ability to reproduce clinical data on estradiol exposure. We hypothesise that the enhanced mechanistic information contained within these models will lead to more robust predictions of the biological phenotype that emerges from the complex interactions between estrogens and the body.To demonstrate the utility of these models we examine the known drug-drug interactions between phenytoin and oral estradiol. We are able to reproduce the approximate 50% reduction in area under the concentration-time curve for estradiol associated with this interaction. Importantly, the inclusion of a genome-scale metabolic model allows the prediction of this interaction without directly specifying it within the model. In addition, we predict that PXR activation by drugs results in an enhanced ability of the liver to excrete glucose. This has important implications for the relationship between drug treatment and metabolic syndrome.We demonstrate how the novel coupling of PBPK models with genome-scale metabolic networks has the potential to aid prediction of drug action, including both drug-drug interactions and changes to the metabolic landscape that may predispose an individual to disease development. link: http://identifiers.org/pubmed/29246152

Sier2017_estrogen_PBPK_GSMN: MODEL1711210003v0.0.1

Physiologically-based Pharmacokinetic (PBPK) model of estradiol disposition in humans. Based on Sier et al_2017_estroge…

Details

Estrogen is a vital hormone that regulates many biological functions within the body. These include roles in the development of the secondary sexual organs in both sexes, plus uterine angiogenesis and proliferation during the menstrual cycle and pregnancy in women. The varied biological roles of estrogens in human health also make them a therapeutic target for contraception, mitigation of the adverse effects of the menopause, and treatment of estrogen-responsive tumours. In addition, endogenous (e.g. genetic variation) and external (e.g. exposure to estrogen-like chemicals) factors are known to impact estrogen biology. To understand how these multiple factors interact to determine an individual's response to therapy is complex, and may be best approached through a systems approach.We present a physiologically-based pharmacokinetic model (PBPK) of estradiol, and validate it against plasma kinetics in humans following intravenous and oral exposure. We extend this model by replacing the intrinsic clearance term with: a detailed kinetic model of estrogen metabolism in the liver; or, a genome-scale model of liver metabolism. Both models were validated by their ability to reproduce clinical data on estradiol exposure. We hypothesise that the enhanced mechanistic information contained within these models will lead to more robust predictions of the biological phenotype that emerges from the complex interactions between estrogens and the body.To demonstrate the utility of these models we examine the known drug-drug interactions between phenytoin and oral estradiol. We are able to reproduce the approximate 50% reduction in area under the concentration-time curve for estradiol associated with this interaction. Importantly, the inclusion of a genome-scale metabolic model allows the prediction of this interaction without directly specifying it within the model. In addition, we predict that PXR activation by drugs results in an enhanced ability of the liver to excrete glucose. This has important implications for the relationship between drug treatment and metabolic syndrome.We demonstrate how the novel coupling of PBPK models with genome-scale metabolic networks has the potential to aid prediction of drug action, including both drug-drug interactions and changes to the metabolic landscape that may predispose an individual to disease development. link: http://identifiers.org/pubmed/29246152

Sigal2019 - Mathematical modelling of cancer stem cell-targeted immunotherapy: MODEL1911270001v0.0.1

This is a mathematical model studies how specific immune system components, namely dendritic cells and cytotoxic T-cells…

Details

The cancer stem cell hypothesis states that tumors are heterogeneous and comprised of several different cell types that have a range of reproductive potentials. Cancer stem cells (CSCs), represent one class of cells that has both reproductive potential and the ability to differentiate. These cells are thought to drive the progression of aggressive and recurring cancers since they give rise to all other constituent cells within a tumor. With the development of immunotherapy in the last decade, the specific targeting of CSCs has become feasible and presents a novel therapeutic approach. In this paper, we construct a mathematical model to study how specific components of the immune system, namely dendritic cells and cytotoxic T-cells interact with different cancer cell types (CSCs and non-CSCs). Using a system of ordinary differential equations, we model the effects of immunotherapy, specifically dendritic cell vaccines and T-cell adoptive therapy, on tumor growth, with and without chemotherapy. The model reproduces several results observed in the literature, including temporal measurements of tumor size from in vivo experiments, and it is used to predict the optimal treatment schedule when combining different treatment modalities. Importantly, the model also demonstrates that chemotherapy increases tumorigenicity whereas CSC-targeted immunotherapy decreases it. link: http://identifiers.org/pubmed/31622595

Sigurdsson2010 - Genome-scale metabolic model of Mus Musculus (iMM1415): MODEL1507180055v0.0.1

Sigurdsson2010 - Genome-scale metabolic model of Mus Musculus (iMM1415)This model is described in the article: [A detai…

Details

BACKGROUND: Well-curated and validated network reconstructions are extremely valuable tools in systems biology. Detailed metabolic reconstructions of mammals have recently emerged, including human reconstructions. They raise the question if the various successful applications of microbial reconstructions can be replicated in complex organisms. RESULTS: We mapped the published, detailed reconstruction of human metabolism (Recon 1) to other mammals. By searching for genes homologous to Recon 1 genes within mammalian genomes, we were able to create draft metabolic reconstructions of five mammals, including the mouse. Each draft reconstruction was created in compartmentalized and non-compartmentalized version via two different approaches. Using gap-filling algorithms, we were able to produce all cellular components with three out of four versions of the mouse metabolic reconstruction. We finalized a functional model by iterative testing until it passed a predefined set of 260 validation tests. The reconstruction is the largest, most comprehensive mouse reconstruction to-date, accounting for 1,415 genes coding for 2,212 gene-associated reactions and 1,514 non-gene-associated reactions.We tested the mouse model for phenotype prediction capabilities. The majority of predicted essential genes were also essential in vivo. However, our non-tissue specific model was unable to predict gene essentiality for many of the metabolic genes shown to be essential in vivo. Our knockout simulation of the lipoprotein lipase gene correlated well with experimental results, suggesting that softer phenotypes can also be simulated. CONCLUSIONS: We have created a high-quality mouse genome-scale metabolic reconstruction, iMM1415 (Mus Musculus, 1415 genes). We demonstrate that the mouse model can be used to perform phenotype simulations, similar to models of microbe metabolism. Since the mouse is an important experimental organism, this model should become an essential tool for studying metabolic phenotypes in mice, including outcomes from drug screening. link: http://identifiers.org/pubmed/20959003

Silber2007_IntravenousGlucose_IntegratedGlucoseInsulinModel: MODEL1112110004v0.0.1

This a model from the article: An integrated model for glucose and insulin regulation in healthy volunteers and type 2…

Details

An integrated model for the regulation of glucose and insulin concentrations following intravenous glucose provocations in healthy volunteers and type 2 diabetic patients was developed. Data from 72 individuals were included. Total glucose, labeled glucose, and insulin concentrations were determined. Simultaneous analysis of all data by nonlinear mixed effect modeling was performed in NONMEM. Integrated models for glucose, labeled glucose, and insulin were developed. Control mechanisms for regulation of glucose production, insulin secretion, and glucose uptake were incorporated. Physiologically relevant differences between healthy volunteers and patients were identified in the regulation of glucose production, elimination rate of glucose, and secretion of insulin. The model was able to describe the insulin and glucose profiles well and also showed a good ability to simulate data. The features of the present model are likely to be of interest for analysis of data collected in antidiabetic drug development and for optimization of study design. link: http://identifiers.org/pubmed/17766701

Simon2019 - NIK-dependent p100 processing into p52 and IkBd degradation, mass action, SBML 2v4: BIOMD0000000870v0.0.1

This model represents NIK-dependent p100 processing into p52 and NIK-dependent IkBd degradation with mass action kinetic…

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=0.05Reaction: p100_NIK => p52 + NIK, Rate Law: compartment*k1*p100_NIK
k1=1.6E-5; k2=2.4E-4Reaction: p100 => IkBd, Rate Law: compartment*(k1*p100^2-k2*IkBd)
k1=3.8E-4Reaction: p52 =>, Rate Law: compartment*k1*p52
k1=0.005; k2=2.4E-4Reaction: p100 + NIK => p100_NIK, Rate Law: compartment*(k1*p100*NIK-k2*p100_NIK)
k1=0.2Reaction: p100t => p100, Rate Law: compartment*k1*p100t

States:

NameDescription
IkBd[Nuclear factor NF-kappa-B p100 subunit]
p100t[ENSG00000077150]
p52[Nuclear factor NF-kappa-B p100 subunit]
NIK[Mitogen-activated protein kinase kinase kinase 14]
IkBd NIK[Mitogen-activated protein kinase kinase kinase 14; Nuclear factor NF-kappa-B p100 subunit]
p100 NIK[Mitogen-activated protein kinase kinase kinase 14; Nuclear factor NF-kappa-B p100 subunit]
p100[Nuclear factor NF-kappa-B p100 subunit]

Simon2019 - NIK-dependent p100 processing into p52 and IkBd degradation, Michaelis-Menten, SBML 2v4: BIOMD0000000869v0.0.1

This model represents NIK-dependent p100 processing into p52 and NIK-dependent IkBd degradation with Michaelis-Menten ki…

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=1.6E-5; k2=2.4E-4Reaction: p100 => IkBd, Rate Law: compartment*(k1*p100^2-k2*IkBd)
Km=10.0; kcat=0.05Reaction: IkBd => ; NIK, Rate Law: compartment*NIK*kcat*IkBd/(Km+IkBd)
k1=3.8E-4Reaction: p100 =>, Rate Law: compartment*k1*p100
k1=0.2Reaction: p100t => p100, Rate Law: compartment*k1*p100t

States:

NameDescription
IkBd[Nuclear factor NF-kappa-B p100 subunit]
p100t[ENSG00000077150]
p52[Nuclear factor NF-kappa-B p100 subunit]
p100[Nuclear factor NF-kappa-B p100 subunit]

Simon2019 - NIK-dependent p100 processing into p52, Mass Action, SBML 2v4: BIOMD0000000868v0.0.1

This model represents NIK-dependent p100 processing into p52 with mass action kinetics. While this model shows identical…

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=0.05Reaction: p100_NIK => p52 + NIK, Rate Law: compartment*k1*p100_NIK
k1=0.005; k2=2.4E-4Reaction: p100 + NIK => p100_NIK, Rate Law: compartment*(k1*p100*NIK-k2*p100_NIK)
k1=3.8E-4Reaction: p100 =>, Rate Law: compartment*k1*p100
k1=0.2Reaction: p100t => p100, Rate Law: compartment*k1*p100t

States:

NameDescription
p100 NIK[Mitogen-activated protein kinase kinase kinase 14; Nuclear factor NF-kappa-B p100 subunit]
NIK[Mitogen-activated protein kinase kinase kinase 14]
p52[Nuclear factor NF-kappa-B p100 subunit]
p100t[ENSG00000077150]
p100[Nuclear factor NF-kappa-B p100 subunit]

Simon2019 - NIK-dependent p100 processing into p52, Michaelis-Menten, SBML 2v4: BIOMD0000000866v0.0.1

This model represents NIK-dependent p100 processing into p52 with Michaelis-Menten kinetics. While this model shows iden…

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=3.8E-4Reaction: p100 =>, Rate Law: compartment*k1*p100
Km=10.0; kcat=0.05Reaction: p100 => p52; NIK, Rate Law: compartment*NIK*kcat*p100/(Km+p100)
k1=0.2Reaction: p100t => p100, Rate Law: compartment*k1*p100t

States:

NameDescription
p100t[ENSG00000077150]
p52[Nuclear factor NF-kappa-B p100 subunit]
p100[Nuclear factor NF-kappa-B p100 subunit]

Singh2006_IL6_Signal_Transduction: BIOMD0000000151v0.0.1

Cytokines like interleukin-6 (IL-6) play an important role in triggering the acute phase response of the body to injury…

Details

The model reproduces Fig 2, Fig3A and Fig 3B of the paper. The ODE for x1(gp180) and x3 (gp 130) is wrong and the authors have communicated to the curator that the species ought to have a constant value. There are a few other differences from the paper and these were made in consultation with the authors. Model was successfully tested on MathSBML

To the extent possible under law, all copyright and related or neighbouring rights to this encoded model have been dedicated to the public domain worldwide. Please refer to CC0 Public Domain Dedication for more information.

In summary, you are entitled to use this encoded model in absolutely any manner you deem suitable, verbatim, or with modification, alone or embedded it in a larger context, redistribute it, commercially or not, in a restricted way or not.

To cite BioModels Database, please use:

Li C, Donizelli M, Rodriguez N, Dharuri H, Endler L, Chelliah V, Li L, He E, Henry A, Stefan MI, Snoep JL, Hucka M, Le Novère N, Laibe C (2010) BioModels Database: An enhanced, curated and annotated resource for published quantitative kinetic models. BMC Syst Biol., 4:92.

Parameters:

NameDescription
kf13 = 2.0E-7; kr13 = 0.2Reaction: x10 + x9 => x14, Rate Law: cytosol*(kf13*x9*x10-kr13*x14)
k49 = 0.058Reaction: x58 => x59 + x55, Rate Law: cytosol*k49*x58
kr46 = 0.001833; kf46 = 0.011Reaction: x55 + x51 => x56, Rate Law: cytosol*(kf46*x55*x51-kr46*x56)
kr44 = 0.001833; kf44 = 0.011Reaction: x53 + x51 => x54, Rate Law: cytosol*(kf44*x51*x53-kr44*x54)
kf39 = 0.3; kr39 = 9.0E-4Reaction: x40 => x45 + x8, Rate Law: cytosol*(kf39*x40-kr39*x45*x8)
k23 = 5.0E-4Reaction: x32 => x33 + x29, Rate Law: cytosol*k23*x32
kr11 = 0.2; kf11 = 0.001Reaction: x17 + x10 => x18, Rate Law: cytosol*(kf11*x10*x17-kr11*x18)
k6 = 0.4Reaction: x11 => x10 + x8, Rate Law: cytosol*k6*x11
kr29 = 7.0E-4; kf29 = 1.0Reaction: x48 => x51 + x49, Rate Law: cytosol*(kf29*x48-kr29*x49*x51)
kf34 = 6.0; kr34 = 0.06Reaction: x16 => x39, Rate Law: cytosol*(kf34*x16-kr34*x39)
kr42 = 0.2; kf42 = 0.0717Reaction: x51 + x50 => x52, Rate Law: cytosol*(kf42*x50*x51-kr42*x52)
kf25 = 0.01; kr25 = 0.0214Reaction: x40 + x35 => x41, Rate Law: cytosol*(kf25*x35*x40-kr25*x41)
kr38 = 0.55; kf38 = 0.01Reaction: x46 + x34 => x45, Rate Law: cytosol*(kf38*x34*x46-kr38*x45)
k43 = 1.0Reaction: x52 => x47 + x50, Rate Law: cytosol*k43*x52
kf40 = 0.03; kr40 = 0.064Reaction: x45 + x35 => x44, Rate Law: cytosol*(kf40*x35*x45-kr40*x44)
kf2 = 0.02; kr2 = 0.02Reaction: x6 => x5 + x2, Rate Law: cytosol*(kr2*x6-kf2*x2*x5)
kr52 = 0.033; kf52 = 1.1E-4Reaction: x61 + x57 => x62, Rate Law: cytosol*(kf52*x57*x61-kr52*x62)
kf56 = 0.014; kr56 = 0.6Reaction: x66 + x65 => x67, Rate Law: cytosol*(kf56*x65*x66-kr56*x67)
k53 = 16.0Reaction: x62 => x63 + x57, Rate Law: cytosol*k53*x62
Km = 340.0; Vm = 1.7Reaction: x46 => x15, Rate Law: cytosol*Vm*x46/(Km+x46)
kf50 = 2.5E-4; kr50 = 0.5Reaction: x59 + x55 => x60, Rate Law: cytosol*(kf50*x55*x59-kr50*x60)
kr24 = 0.55; kf24 = 0.01Reaction: x39 + x34 => x40, Rate Law: cytosol*(kf24*x39*x34-kr24*x40)
k51 = 0.058Reaction: x60 => x59 + x53, Rate Law: cytosol*k51*x60
kf3 = 0.04Reaction: x6 => x7, Rate Law: cytosol*kf3*x6^2
k12 = 0.003Reaction: x18 => x17 + x9, Rate Law: cytosol*k12*x18
kr8 = 0.1; kf8 = 0.02Reaction: x10 => x13, Rate Law: cytosol*(2*kf8*x10^2-2*kr8*x13)
k45 = 3.5Reaction: x54 => x55 + x51, Rate Law: cytosol*k45*x54
kr26 = 1.3; kf26 = 0.015Reaction: x41 + x36 => x42, Rate Law: cytosol*(kf26*x36*x41-kr26*x42)
kf48 = 0.0143; kr48 = 0.8Reaction: x59 + x57 => x58, Rate Law: cytosol*(kf48*x57*x59-kr48*x58)
kf54 = 1.1E-4; kr54 = 0.033Reaction: x64 => x63 + x57, Rate Law: cytosol*(kr54*x64-kf54*x57*x63)
k14 = 0.005Reaction: x13 => x20, Rate Law: cytosol*k14*x13
k20 = 0.01Reaction: => x29; x26, Rate Law: nucleus*k20*x26
kf32 = 0.1; kr32 = 2.45E-4Reaction: x41 => x44 + x8, Rate Law: cytosol*(kf32*x41-kr32*x44*x8)
kr1 = 0.05; kf1 = 0.1Reaction: x4 => x5; x3, Rate Law: cytosol*(kf1*x3*x4-kr1*x5)
kf37 = 0.3; kr37 = 9.0E-4Reaction: x39 => x46 + x8, Rate Law: cytosol*(kf37*x39-kr37*x8*x46)
kf58 = 0.005; kr58 = 0.5Reaction: x66 + x63 => x68, Rate Law: cytosol*(kf58*x63*x66-kr58*x68)
kr3 = 0.2Reaction: x7 => x6, Rate Law: cytosol*kr3*x7
k4 = 0.005Reaction: x7 => x8, Rate Law: cytosol*k4*x7
k10 = 0.003Reaction: x32 => x29 + x15 + x9 + x7, Rate Law: cytosol*k10*x32
kf7 = 0.005; kr7 = 0.5Reaction: x10 + x8 => x12, Rate Law: cytosol*(kf7*x8*x10-kr7*x12)
kf21 = 0.02; kr21 = 0.1Reaction: x29 + x8 => x30, Rate Law: cytosol*(kf21*x29*x8-kr21*x30)
kf9 = 0.001; kr9 = 0.2Reaction: x15 + x8 => x16, Rate Law: cytosol*(kf9*x8*x15-kr9*x16)
kr5 = 0.8; kf5 = 0.008Reaction: x30 + x9 => x31, Rate Law: cytosol*(kf5*x9*x30-kr5*x31)
k17 = 0.05Reaction: x22 => x9, Rate Law: nucleus*k17*x22
k47 = 2.9Reaction: x56 => x51 + x57, Rate Law: cytosol*k47*x56
k55 = 6.7Reaction: x64 => x65 + x57, Rate Law: cytosol*k55*x64
kf15 = 0.001; kr15 = 0.2Reaction: x23 + x20 => x27, Rate Law: nucleus*(kf15*x23*x20-kr15*x27)
kr33 = 0.021; kf33 = 0.3Reaction: x44 => x46 + x38, Rate Law: cytosol*(kf33*x44-kr33*x38*x46)
k59 = 0.3Reaction: x68 => x66 + x61, Rate Law: cytosol*k59*x68

States:

NameDescription
x16[Tyrosine-protein phosphatase non-receptor type 11; Tyrosine-protein kinase JAK1; Interleukin 6 signal transducer; Interleukin-6; Interleukin-6 receptor subunit alpha]
x54[Mitogen activated protein kinase kinase 1Putative uncharacterized protein; RAF proto-oncogene serine/threonine-protein kinase]
x32[Suppressor of cytokine signaling 3; Signal transducer and activator of transcription 3; Tyrosine-protein phosphatase non-receptor type 11; Tyrosine-protein kinase JAK1; Interleukin 6 signal transducer; Interleukin-6; Interleukin-6 receptor subunit alpha]
x60[Mitogen activated protein kinase kinase 1Putative uncharacterized protein]
x62[Mitogen-activated protein kinase 1; Mitogen activated protein kinase kinase 1Putative uncharacterized protein]
x38[Son of sevenless homolog 1; Growth factor receptor-bound protein 2]
x45[Growth factor receptor-bound protein 2; Tyrosine-protein phosphatase non-receptor type 11]
x23PP2
x66Phosp3
x51[RAF proto-oncogene serine/threonine-protein kinase]
x46[Tyrosine-protein phosphatase non-receptor type 11]
x11[Signal transducer and activator of transcription 3; Tyrosine-protein kinase JAK1; Interleukin 6 signal transducer; Interleukin-6; Interleukin-6 receptor subunit alpha]
x29[Suppressor of cytokine signaling 3]
x53[Mitogen activated protein kinase kinase 1Putative uncharacterized protein]
x59Phosp2
x12[Signal transducer and activator of transcription 3; Tyrosine-protein kinase JAK1; Interleukin 6 signal transducer; Interleukin-6; Interleukin-6 receptor subunit alpha]
x5[Interleukin 6 signal transducer; Tyrosine-protein kinase JAK1]
x40[Growth factor receptor-bound protein 2; Tyrosine-protein phosphatase non-receptor type 11; Tyrosine-protein kinase JAK1; Interleukin 6 signal transducer; Interleukin-6; Interleukin-6 receptor subunit alpha]
x19[Signal transducer and activator of transcription 3]
x63[Mitogen-activated protein kinase 1]
x57[Mitogen activated protein kinase kinase 1Putative uncharacterized protein]
x55[Mitogen activated protein kinase kinase 1Putative uncharacterized protein]
x20[Signal transducer and activator of transcription 3]
x41[Growth factor receptor-bound protein 2; Tyrosine-protein phosphatase non-receptor type 11; Tyrosine-protein kinase JAK1; Interleukin 6 signal transducer; Interleukin-6; Interleukin-6 receptor subunit alpha]
x39[Tyrosine-protein phosphatase non-receptor type 11; Tyrosine-protein kinase JAK1; Interleukin 6 signal transducer; Interleukin-6; Interleukin-6 receptor subunit alpha]
x50Phosp1
x61[Mitogen-activated protein kinase 1]
x9[Signal transducer and activator of transcription 3]
x15[Tyrosine-protein phosphatase non-receptor type 11]
x8[Tyrosine-protein kinase JAK1; Interleukin 6 signal transducer; Interleukin-6; Interleukin-6 receptor subunit alpha]
x7[Tyrosine-protein kinase JAK1; Interleukin 6 signal transducer; Interleukin-6; Interleukin-6 receptor subunit alpha]
x13[Signal transducer and activator of transcription 3]
x52[RAF proto-oncogene serine/threonine-protein kinase]
x21[Signal transducer and activator of transcription 3]
x10[Signal transducer and activator of transcription 3]
x64[Mitogen-activated protein kinase 1; Mitogen activated protein kinase kinase 1Putative uncharacterized protein]

Singh2006_TCA_Ecoli_acetate: BIOMD0000000221v0.0.1

This a model from the article: Kinetic modeling of tricarboxylic acid cycle and glyoxylate bypass in Mycobacterium tub…

Details

BACKGROUND: Targeting persistent tubercule bacilli has become an important challenge in the development of anti-tuberculous drugs. As the glyoxylate bypass is essential for persistent bacilli, interference with it holds the potential for designing new antibacterial drugs. We have developed kinetic models of the tricarboxylic acid cycle and glyoxylate bypass in Escherichia coli and Mycobacterium tuberculosis, and studied the effects of inhibition of various enzymes in the M. tuberculosis model. RESULTS: We used E. coli to validate the pathway-modeling protocol and showed that changes in metabolic flux can be estimated from gene expression data. The M. tuberculosis model reproduced the observation that deletion of one of the two isocitrate lyase genes has little effect on bacterial growth in macrophages, but deletion of both genes leads to the elimination of the bacilli from the lungs. It also substantiated the inhibition of isocitrate lyases by 3-nitropropionate. On the basis of our simulation studies, we propose that: (i) fractional inactivation of both isocitrate dehydrogenase 1 and isocitrate dehydrogenase 2 is required for a flux through the glyoxylate bypass in persistent mycobacteria; and (ii) increasing the amount of active isocitrate dehydrogenases can stop the flux through the glyoxylate bypass, so the kinase that inactivates isocitrate dehydrogenase 1 and/or the proposed inactivator of isocitrate dehydrogenase 2 is a potential target for drugs against persistent mycobacteria. In addition, competitive inhibition of isocitrate lyases along with a reduction in the inactivation of isocitrate dehydrogenases appears to be a feasible strategy for targeting persistent mycobacteria. CONCLUSION: We used kinetic modeling of biochemical pathways to assess various potential anti-tuberculous drug targets that interfere with the glyoxylate bypass flux, and indicated the type of inhibition needed to eliminate the pathogen. The advantage of such an approach to the assessment of drug targets is that it facilitates the study of systemic effect(s) of the modulation of the target enzyme(s) in the cellular environment. link: http://identifiers.org/pubmed/16887020

Parameters:

NameDescription
Kgly_ms=2.0 mmol*l^(-1); Vr_ms=0.285 mmol*l^(-1)*(60*s)^(-1); Kmal_ms=1.0 mmol*l^(-1); Kaca_ms=0.01 mmol*l^(-1); Vf_ms=28.5 mmol*l^(-1)*(60*s)^(-1); Kcoa_ms=0.1 mmol*l^(-1)Reaction: gly + aca => mal + coa, Rate Law: cell*(Vf_ms*gly/Kgly_ms*aca/Kaca_ms-Vr_ms*mal/Kmal_ms*coa/Kcoa_ms)/((1+gly/Kgly_ms+mal/Kmal_ms)*(1+aca/Kaca_ms+coa/Kcoa_ms))
Vr_icl=0.285 mmol*l^(-1)*(60*s)^(-1); Vf_icl=28.5 mmol*l^(-1)*(60*s)^(-1); Kicit_icl=0.604 mmol*l^(-1); Kgly_icl=0.13 mmol*l^(-1); Ksuc_icl=0.59 mmol*l^(-1)Reaction: icit => suc + gly, Rate Law: cell*(Vf_icl*icit/Kicit_icl-Vr_icl*suc/Ksuc_icl*gly/Kgly_icl)/(1+icit/Kicit_icl+suc/Ksuc_icl+gly/Kgly_icl+icit/Kicit_icl*suc/Ksuc_icl+suc/Ksuc_icl*gly/Kgly_icl)
Vr_fum=144.67 mmol*l^(-1)*(60*s)^(-1); Kmal_fum=0.04 mmol*l^(-1); Vf_fum=156.24 mmol*l^(-1)*(60*s)^(-1); Kfa_fum=0.15 mmol*l^(-1)Reaction: fa => mal, Rate Law: cell*(Vf_fum*fa/Kfa_fum-Vr_fum*mal/Kmal_fum)/(1+fa/Kfa_fum+mal/Kmal_fum)
Vf_scas=8.96 mmol*l^(-1)*(60*s)^(-1); Ksca_scas=0.02 mmol*l^(-1); Ksuc_scas=5.0 mmol*l^(-1); Vr_scas=0.0896 mmol*l^(-1)*(60*s)^(-1)Reaction: sca => suc, Rate Law: cell*(Vf_scas*sca/Ksca_scas-Vr_scas*suc/Ksuc_scas)/(1+sca/Ksca_scas+suc/Ksuc_scas)
Kaca_cs=0.03 mmol*l^(-1); Kcit_cs=0.7 mmol*l^(-1); Vf_cs=446.88 mmol*l^(-1)*(60*s)^(-1); Koaa_cs=0.07 mmol*l^(-1); Vr_cs=4.4688 mmol*l^(-1)*(60*s)^(-1); Kcoa_cs=0.3 mmol*l^(-1)Reaction: aca + oaa => coa + cit, Rate Law: cell*(Vf_cs*aca/Kaca_cs*oaa/Koaa_cs-Vr_cs*coa/Kcoa_cs*cit/Kcit_cs)/((1+aca/Kaca_cs+coa/Kcoa_cs)*(1+oaa/Koaa_cs+cit/Kcit_cs))
Vr_acn=6.2928 mmol*l^(-1)*(60*s)^(-1); Kcit_acn=1.7 mmol*l^(-1); Kicit_acn=3.33 mmol*l^(-1); Vf_acn=629.28 mmol*l^(-1)*(60*s)^(-1)Reaction: cit => icit, Rate Law: cell*(Vf_acn*cit/Kcit_acn-Vr_acn*icit/Kicit_acn)/(1+cit/Kcit_acn+icit/Kicit_acn)
Ksca_kdh=1.0 mmol*l^(-1); Kakg_kdh=0.1 mmol*l^(-1); Vr_kdh=0.57344 mmol*l^(-1)*(60*s)^(-1); Vf_kdh=57.344 mmol*l^(-1)*(60*s)^(-1)Reaction: akg => sca, Rate Law: cell*(Vf_kdh*akg/Kakg_kdh-Vr_kdh*sca/Ksca_kdh)/(1+akg/Kakg_kdh+sca/Ksca_kdh)
Vf_icd=6.625 mmol*l^(-1)*(60*s)^(-1); Vr_icd=0.06625 mmol*l^(-1)*(60*s)^(-1); Kakg_icd=0.13 mmol*l^(-1); Kicit_icd=0.008 mmol*l^(-1)Reaction: icit => akg, Rate Law: cell*(Vf_icd*icit/Kicit_icd-Vr_icd*akg/Kakg_icd)/(1+icit/Kicit_icd+akg/Kakg_icd)
Vf_mdh=1390.9 mmol*l^(-1)*(60*s)^(-1); Koaa_mdh=0.04 mmol*l^(-1); Kmal_mdh=2.6 mmol*l^(-1); Vr_mdh=1276.06 mmol*l^(-1)*(60*s)^(-1)Reaction: mal => oaa, Rate Law: cell*(Vf_mdh*mal/Kmal_mdh-Vr_mdh*oaa/Koaa_mdh)/(1+mal/Kmal_mdh+oaa/Koaa_mdh)
Vr_sdh=16.24 mmol*l^(-1)*(60*s)^(-1); Vf_sdh=17.7 mmol*l^(-1)*(60*s)^(-1); Ksuc_sdh=0.02 mmol*l^(-1); Kfa_sdh=0.4 mmol*l^(-1)Reaction: suc => fa, Rate Law: cell*(Vf_sdh*suc/Ksuc_sdh-Vr_sdh*fa/Kfa_sdh)/(1+suc/Ksuc_sdh+fa/Kfa_sdh)

States:

NameDescription
gly[glyoxylic acid; Glyoxylate]
icit[isocitric acid; Isocitrate]
coa[coenzyme A; CoA]
mal[malic acid; Malate]
akg[2-oxoglutaric acid; 2-Oxoglutarate]
aca[acetyl-CoA; Acetyl-CoA]
cit[citric acid; Citrate]
oaa[oxaloacetic acid; Oxaloacetate]
biosynbiosyn
fa[fumaric acid; Fumarate]
suc[succinic acid; Succinate]
sca[succinyl-CoA; Succinyl-CoA]

Singh2006_TCA_Ecoli_glucose: BIOMD0000000222v0.0.1

This a model from the article: Kinetic modeling of tricarboxylic acid cycle and glyoxylate bypass in Mycobacterium tub…

Details

BACKGROUND: Targeting persistent tubercule bacilli has become an important challenge in the development of anti-tuberculous drugs. As the glyoxylate bypass is essential for persistent bacilli, interference with it holds the potential for designing new antibacterial drugs. We have developed kinetic models of the tricarboxylic acid cycle and glyoxylate bypass in Escherichia coli and Mycobacterium tuberculosis, and studied the effects of inhibition of various enzymes in the M. tuberculosis model. RESULTS: We used E. coli to validate the pathway-modeling protocol and showed that changes in metabolic flux can be estimated from gene expression data. The M. tuberculosis model reproduced the observation that deletion of one of the two isocitrate lyase genes has little effect on bacterial growth in macrophages, but deletion of both genes leads to the elimination of the bacilli from the lungs. It also substantiated the inhibition of isocitrate lyases by 3-nitropropionate. On the basis of our simulation studies, we propose that: (i) fractional inactivation of both isocitrate dehydrogenase 1 and isocitrate dehydrogenase 2 is required for a flux through the glyoxylate bypass in persistent mycobacteria; and (ii) increasing the amount of active isocitrate dehydrogenases can stop the flux through the glyoxylate bypass, so the kinase that inactivates isocitrate dehydrogenase 1 and/or the proposed inactivator of isocitrate dehydrogenase 2 is a potential target for drugs against persistent mycobacteria. In addition, competitive inhibition of isocitrate lyases along with a reduction in the inactivation of isocitrate dehydrogenases appears to be a feasible strategy for targeting persistent mycobacteria. CONCLUSION: We used kinetic modeling of biochemical pathways to assess various potential anti-tuberculous drug targets that interfere with the glyoxylate bypass flux, and indicated the type of inhibition needed to eliminate the pathogen. The advantage of such an approach to the assessment of drug targets is that it facilitates the study of systemic effect(s) of the modulation of the target enzyme(s) in the cellular environment. link: http://identifiers.org/pubmed/16887020

Parameters:

NameDescription
Vf_scas=3.5 mM_per_min; Ksca_scas=0.02 mM; Vr_scas=0.035 mM_per_min; Ksuc_scas=5.0 mMReaction: sca => suc, Rate Law: cell*(Vf_scas*sca/Ksca_scas-Vr_scas*suc/Ksuc_scas)/(1+sca/Ksca_scas+suc/Ksuc_scas)
Kaca_cs=0.03 mM; Kcit_cs=0.7 mM; Vf_cs=91.2 mM_per_min; Koaa_cs=0.07 mM; Kcoa_cs=0.3 mM; Vr_cs=0.912 mM_per_minReaction: aca + oaa => coa + cit, Rate Law: cell*(Vf_cs*aca/Kaca_cs*oaa/Koaa_cs-Vr_cs*coa/Kcoa_cs*cit/Kcit_cs)/((1+aca/Kaca_cs+coa/Kcoa_cs)*(1+oaa/Koaa_cs+cit/Kcit_cs))
Vr_sdh=7.31 mM_per_min; Vf_sdh=7.38 mM_per_min; Ksuc_sdh=0.02 mM; Kfa_sdh=0.4 mMReaction: suc => fa, Rate Law: cell*(Vf_sdh*suc/Ksuc_sdh-Vr_sdh*fa/Kfa_sdh)/(1+suc/Ksuc_sdh+fa/Kfa_sdh)
Koaa_mdh=0.04 mM; Kmal_mdh=2.6 mM; Vr_mdh=353.11 mM_per_min; Vf_mdh=356.64 mM_per_minReaction: mal => oaa, Rate Law: cell*(Vf_mdh*mal/Kmal_mdh-Vr_mdh*oaa/Koaa_mdh)/(1+mal/Kmal_mdh+oaa/Koaa_mdh)
Ksuc_icl=0.59 mM; Kgly_icl=0.13 mM; Vf_icl=1.9 mM_per_min; Vr_icl=0.019 mM_per_min; Kicit_icl=0.604 mMReaction: icit => suc + gly, Rate Law: cell*(Vf_icl*icit/Kicit_icl-Vr_icl*suc/Ksuc_icl*gly/Kgly_icl)/(1+icit/Kicit_icl+suc/Ksuc_icl+gly/Kgly_icl+icit/Kicit_icl*suc/Ksuc_icl+suc/Ksuc_icl*gly/Kgly_icl)
Vf_fum=44.64 mM_per_min; Vr_fum=37.2 mM_per_min; Kmal_fum=0.04 mM; Kfa_fum=0.15 mMReaction: fa => mal, Rate Law: cell*(Vf_fum*fa/Kfa_fum-Vr_fum*mal/Kmal_fum)/(1+fa/Kfa_fum+mal/Kmal_fum)
Kicit_acn=3.33 mM; Vr_acn=0.912 mM_per_min; Kcit_acn=1.7 mM; Vf_acn=91.2 mM_per_minReaction: cit => icit, Rate Law: cell*(Vf_acn*cit/Kcit_acn-Vr_acn*icit/Kicit_acn)/(1+cit/Kcit_acn+icit/Kicit_acn)
Kakg_kdh=0.1 mM; Vr_kdh=0.3584 mM_per_min; Vf_kdh=35.84 mM_per_min; Ksca_kdh=1.0 mMReaction: akg => sca, Rate Law: cell*(Vf_kdh*akg/Kakg_kdh-Vr_kdh*sca/Ksca_kdh)/(1+akg/Kakg_kdh+sca/Ksca_kdh)
Kakg_icd=0.13 mM; Kicit_icd=0.008 mM; Vr_icd=0.1472 mM_per_min; Vf_icd=14.72 mM_per_minReaction: icit => akg, Rate Law: cell*(Vf_icd*icit/Kicit_icd-Vr_icd*akg/Kakg_icd)/(1+icit/Kicit_icd+akg/Kakg_icd)
Kmal_ms=1.0 mM; Vf_ms=1.9 mM_per_min; Vr_ms=0.019 mM_per_min; Kgly_ms=2.0 mM; Kcoa_ms=0.1 mM; Kaca_ms=0.01 mMReaction: gly + aca => mal + coa, Rate Law: cell*(Vf_ms*gly/Kgly_ms*aca/Kaca_ms-Vr_ms*mal/Kmal_ms*coa/Kcoa_ms)/((1+gly/Kgly_ms+mal/Kmal_ms)*(1+aca/Kaca_ms+coa/Kcoa_ms))

States:

NameDescription
gly[glyoxylic acid; Glyoxylate]
icit[isocitric acid; Isocitrate]
coa[coenzyme A; C000010]
mal[malic acid; Malate]
akg[2-oxoglutaric acid; 2-Oxoglutarate]
aca[acetyl-CoA; Acetyl-CoA]
cit[citric acid; Citrate]
fa[fumaric acid; Fumarate]
biosynbiosyn
oaa[oxaloacetic acid; Oxaloacetate]
suc[succinic acid; Succinate]
sca[succinyl-CoA; Succinyl-CoA]

Singh2006_TCA_mtu_model1: BIOMD0000000219v0.0.1

This a model from the article: Kinetic modeling of tricarboxylic acid cycle and glyoxylate bypass in Mycobacterium tub…

Details

BACKGROUND: Targeting persistent tubercule bacilli has become an important challenge in the development of anti-tuberculous drugs. As the glyoxylate bypass is essential for persistent bacilli, interference with it holds the potential for designing new antibacterial drugs. We have developed kinetic models of the tricarboxylic acid cycle and glyoxylate bypass in Escherichia coli and Mycobacterium tuberculosis, and studied the effects of inhibition of various enzymes in the M. tuberculosis model. RESULTS: We used E. coli to validate the pathway-modeling protocol and showed that changes in metabolic flux can be estimated from gene expression data. The M. tuberculosis model reproduced the observation that deletion of one of the two isocitrate lyase genes has little effect on bacterial growth in macrophages, but deletion of both genes leads to the elimination of the bacilli from the lungs. It also substantiated the inhibition of isocitrate lyases by 3-nitropropionate. On the basis of our simulation studies, we propose that: (i) fractional inactivation of both isocitrate dehydrogenase 1 and isocitrate dehydrogenase 2 is required for a flux through the glyoxylate bypass in persistent mycobacteria; and (ii) increasing the amount of active isocitrate dehydrogenases can stop the flux through the glyoxylate bypass, so the kinase that inactivates isocitrate dehydrogenase 1 and/or the proposed inactivator of isocitrate dehydrogenase 2 is a potential target for drugs against persistent mycobacteria. In addition, competitive inhibition of isocitrate lyases along with a reduction in the inactivation of isocitrate dehydrogenases appears to be a feasible strategy for targeting persistent mycobacteria. CONCLUSION: We used kinetic modeling of biochemical pathways to assess various potential anti-tuberculous drug targets that interfere with the glyoxylate bypass flux, and indicated the type of inhibition needed to eliminate the pathogen. The advantage of such an approach to the assessment of drug targets is that it facilitates the study of systemic effect(s) of the modulation of the target enzyme(s) in the cellular environment. link: http://identifiers.org/pubmed/16887020

Parameters:

NameDescription
Ksuc_ssadh=0.15 mM; Vr_ssadh=0.0651 mM_per_min; Vf_ssadh=6.51 mM_per_min; Kssa_ssadh=0.015 mMReaction: ssa => suc, Rate Law: cell*(Vf_ssadh*ssa/Kssa_ssadh-Vr_ssadh*suc/Ksuc_ssadh)/(1+ssa/Kssa_ssadh+suc/Ksuc_ssadh)
Vr_scas=0.012 mM_per_min; Ksca_scas=0.02 mM; Vf_scas=1.2 mM_per_min; Ksuc_scas=5.0 mMReaction: sca => suc, Rate Law: cell*(Vf_scas*sca/Ksca_scas-Vr_scas*suc/Ksuc_scas)/(1+sca/Ksca_scas+suc/Ksuc_scas)
Kakg_kgd=0.48 mM; Vr_kgd=0.483 mM_per_min; Vf_kgd=48.3 mM_per_min; Kssa_kgd=4.8 mMReaction: akg => ssa, Rate Law: cell*(Vf_kgd*akg/Kakg_kgd-Vr_kgd*ssa/Kssa_kgd)/(1+akg/Kakg_kgd+ssa/Kssa_kgd)
Vr_icl1=0.01172 mM_per_min; Kicit_icl1=0.145 mM; Ksuc_icl1=0.59 mM; Vf_icl1=1.172 mM_per_min; Kgly_icl1=0.13 mMReaction: icit => suc + gly, Rate Law: cell*(Vf_icl1*icit/Kicit_icl1-Vr_icl1*suc/Ksuc_icl1*gly/Kgly_icl1)/(1+icit/Kicit_icl1+suc/Ksuc_icl1+gly/Kgly_icl1+icit/Kicit_icl1*suc/Ksuc_icl1+suc/Ksuc_icl1*gly/Kgly_icl1)
Kfa_sdh=0.15 mM; Ksuc_sdh=0.12 mM; Vf_sdh=1.02 mM_per_min; Vr_sdh=1.02 mM_per_minReaction: suc => fa, Rate Law: cell*(Vf_sdh*suc/Ksuc_sdh-Vr_sdh*fa/Kfa_sdh)/(1+suc/Ksuc_sdh+fa/Kfa_sdh)
Kakg_icd1=0.3 mM; Vf_icd1=10.2 mM_per_min; Vr_icd1=0.102 mM_per_min; Kicit_icd1=0.03 mMReaction: icit => akg, Rate Law: cell*(Vf_icd1*icit/Kicit_icd1-Vr_icd1*akg/Kakg_icd1)/(1+icit/Kicit_icd1+akg/Kakg_icd1)
Vr_icd2=0.09965 mM_per_min; Kakg_icd2=0.6 mM; Vf_icd2=9.965 mM_per_min; Kicit_icd2=0.06 mMReaction: icit => akg, Rate Law: cell*(Vf_icd2*icit/Kicit_icd2-Vr_icd2*akg/Kakg_icd2)/(1+icit/Kicit_icd2+akg/Kakg_icd2)
Kakg_kdh=0.1 mM; Ksca_kdh=1.0 mM; Vr_kdh=0.57344 mM_per_min; Vf_kdh=57.344 mM_per_minReaction: akg => sca, Rate Law: cell*(Vf_kdh*akg/Kakg_kdh-Vr_kdh*sca/Ksca_kdh)/(1+akg/Kakg_kdh+sca/Ksca_kdh)
Kakg_icd1=0.3 mM; Vf_icd1=10.2 mM_per_min; Vr_icd2=0.09965 mM_per_min; Kakg_icd2=0.6 mM; Vr_icd1=0.102 mM_per_min; Vf_icd2=9.965 mM_per_min; Kicit_icd1=0.03 mM; Kicit_icd2=0.06 mMReaction: akg => biosyn; icit, Rate Law: cell*0.0341*((Vf_icd1*icit/Kicit_icd1-Vr_icd1*akg/Kakg_icd1)/(1+icit/Kicit_icd1+akg/Kakg_icd1)+(Vf_icd2*icit/Kicit_icd2-Vr_icd2*akg/Kakg_icd2)/(1+icit/Kicit_icd2+akg/Kakg_icd2))
Vr_cs=0.648 mM_per_min; Kaca_cs=0.05 mM; Kcit_cs=0.12 mM; Kcoa_cs=0.5 mM; Vf_cs=64.8 mM_per_min; Koaa_cs=0.012 mMReaction: aca + oaa => coa + cit, Rate Law: cell*(Vf_cs*aca/Kaca_cs*oaa/Koaa_cs-Vr_cs*coa/Kcoa_cs*cit/Kcit_cs)/((1+aca/Kaca_cs+coa/Kcoa_cs)*(1+oaa/Koaa_cs+cit/Kcit_cs))
Vr_acn=0.312 mM_per_min; Kicit_acn=0.7 mM; Vf_acn=31.2 mM_per_min; Kcit_acn=1.7 mMReaction: cit => icit, Rate Law: cell*(Vf_acn*cit/Kcit_acn-Vr_acn*icit/Kicit_acn)/(1+cit/Kcit_acn+icit/Kicit_acn)
Kmal_fum=2.38 mM; Kfa_fum=0.25 mM; Vr_fum=87.7 mM_per_min; Vf_fum=87.7 mM_per_minReaction: fa => mal, Rate Law: cell*(Vf_fum*fa/Kfa_fum-Vr_fum*mal/Kmal_fum)/(1+fa/Kfa_fum+mal/Kmal_fum)
Kicit_icl2=1.3 mM; Kgly_icl2=1.3 mM; Vr_icl2=0.00391 mM_per_min; Ksuc_icl2=5.9 mM; Vf_icl2=0.391 mM_per_minReaction: icit => suc + gly, Rate Law: cell*(Vf_icl2*icit/Kicit_icl2-Vr_icl2*suc/Ksuc_icl2*gly/Kgly_icl2)/(1+icit/Kicit_icl2+suc/Ksuc_icl2+gly/Kgly_icl2+icit/Kicit_icl2*suc/Ksuc_icl2+suc/Ksuc_icl2*gly/Kgly_icl2)
Koaa_mdh=0.0443 mM; Vr_mdh=184.0 mM_per_min; Kmal_mdh=0.833 mM; Vf_mdh=184.0 mM_per_minReaction: mal => oaa, Rate Law: cell*(Vf_mdh*mal/Kmal_mdh-Vr_mdh*oaa/Koaa_mdh)/(1+mal/Kmal_mdh+oaa/Koaa_mdh)
Kmal_ms=1.0 mM; Vf_ms=20.0 mM_per_min; Kgly_ms=0.057 mM; Kaca_ms=0.03 mM; Vr_ms=0.2 mM_per_min; Kcoa_ms=0.1 mMReaction: gly + aca => mal + coa, Rate Law: cell*(Vf_ms*gly/Kgly_ms*aca/Kaca_ms-Vr_ms*mal/Kmal_ms*coa/Kcoa_ms)/((1+gly/Kgly_ms+mal/Kmal_ms)*(1+aca/Kaca_ms+coa/Kcoa_ms))

States:

NameDescription
gly[glyoxylic acid; Glyoxylate]
icit[isocitric acid; Isocitrate]
coa[coenzyme A; CoA]
mal[malic acid; Malate]
ssa[succinic semialdehyde; Succinate semialdehyde]
akg[2-oxoglutaric acid; 2-Oxoglutarate]
aca[acetyl-CoA; Acetyl-CoA]
cit[citric acid; Citrate]
oaa[oxaloacetic acid; Oxaloacetate]
biosynbiosyn
fa[fumaric acid; Fumarate]
suc[succinic acid; Succinate]
sca[succinyl-CoA; Succinyl-CoA]

Singh2006_TCA_mtu_model2: BIOMD0000000218v0.0.1

This a model from the article: Kinetic modeling of tricarboxylic acid cycle and glyoxylate bypass in Mycobacterium tub…

Details

BACKGROUND: Targeting persistent tubercule bacilli has become an important challenge in the development of anti-tuberculous drugs. As the glyoxylate bypass is essential for persistent bacilli, interference with it holds the potential for designing new antibacterial drugs. We have developed kinetic models of the tricarboxylic acid cycle and glyoxylate bypass in Escherichia coli and Mycobacterium tuberculosis, and studied the effects of inhibition of various enzymes in the M. tuberculosis model. RESULTS: We used E. coli to validate the pathway-modeling protocol and showed that changes in metabolic flux can be estimated from gene expression data. The M. tuberculosis model reproduced the observation that deletion of one of the two isocitrate lyase genes has little effect on bacterial growth in macrophages, but deletion of both genes leads to the elimination of the bacilli from the lungs. It also substantiated the inhibition of isocitrate lyases by 3-nitropropionate. On the basis of our simulation studies, we propose that: (i) fractional inactivation of both isocitrate dehydrogenase 1 and isocitrate dehydrogenase 2 is required for a flux through the glyoxylate bypass in persistent mycobacteria; and (ii) increasing the amount of active isocitrate dehydrogenases can stop the flux through the glyoxylate bypass, so the kinase that inactivates isocitrate dehydrogenase 1 and/or the proposed inactivator of isocitrate dehydrogenase 2 is a potential target for drugs against persistent mycobacteria. In addition, competitive inhibition of isocitrate lyases along with a reduction in the inactivation of isocitrate dehydrogenases appears to be a feasible strategy for targeting persistent mycobacteria. CONCLUSION: We used kinetic modeling of biochemical pathways to assess various potential anti-tuberculous drug targets that interfere with the glyoxylate bypass flux, and indicated the type of inhibition needed to eliminate the pathogen. The advantage of such an approach to the assessment of drug targets is that it facilitates the study of systemic effect(s) of the modulation of the target enzyme(s) in the cellular environment. link: http://identifiers.org/pubmed/16887020

Parameters:

NameDescription
Ksuc_ssadh=0.15 mM; Vr_ssadh=0.0651 mM_per_min; Vf_ssadh=6.51 mM_per_min; Kssa_ssadh=0.015 mMReaction: ssa => suc, Rate Law: cell*(Vf_ssadh*ssa/Kssa_ssadh-Vr_ssadh*suc/Ksuc_ssadh)/(1+ssa/Kssa_ssadh+suc/Ksuc_ssadh)
Vr_scas=0.012 mM_per_min; Ksca_scas=0.02 mM; Vf_scas=1.2 mM_per_min; Ksuc_scas=5.0 mMReaction: sca => suc, Rate Law: cell*(Vf_scas*sca/Ksca_scas-Vr_scas*suc/Ksuc_scas)/(1+sca/Ksca_scas+suc/Ksuc_scas)
Kakg_kgd=0.48 mM; Vr_kgd=0.483 mM_per_min; Vf_kgd=48.3 mM_per_min; Kssa_kgd=4.8 mMReaction: akg => ssa, Rate Law: cell*(Vf_kgd*akg/Kakg_kgd-Vr_kgd*ssa/Kssa_kgd)/(1+akg/Kakg_kgd+ssa/Kssa_kgd)
Vr_icl1=0.01172 mM_per_min; Kicit_icl1=0.145 mM; Ksuc_icl1=0.59 mM; Vf_icl1=1.172 mM_per_min; Kgly_icl1=0.13 mMReaction: icit => suc + gly, Rate Law: cell*(Vf_icl1*icit/Kicit_icl1-Vr_icl1*suc/Ksuc_icl1*gly/Kgly_icl1)/(1+icit/Kicit_icl1+suc/Ksuc_icl1+gly/Kgly_icl1+icit/Kicit_icl1*suc/Ksuc_icl1+suc/Ksuc_icl1*gly/Kgly_icl1)
Kfa_sdh=0.15 mM; Ksuc_sdh=0.12 mM; Vf_sdh=1.02 mM_per_min; Vr_sdh=1.02 mM_per_minReaction: suc => fa, Rate Law: cell*(Vf_sdh*suc/Ksuc_sdh-Vr_sdh*fa/Kfa_sdh)/(1+suc/Ksuc_sdh+fa/Kfa_sdh)
Kakg_icd1=0.3 mM; Vf_icd1=10.2 mM_per_min; Vr_icd1=0.102 mM_per_min; Kicit_icd1=0.03 mMReaction: icit => akg, Rate Law: cell*(Vf_icd1*icit/Kicit_icd1-Vr_icd1*akg/Kakg_icd1)/(1+icit/Kicit_icd1+akg/Kakg_icd1)
Vr_icd2=0.09965 mM_per_min; Kakg_icd2=0.6 mM; Vf_icd2=9.965 mM_per_min; Kicit_icd2=0.06 mMReaction: icit => akg, Rate Law: cell*(Vf_icd2*icit/Kicit_icd2-Vr_icd2*akg/Kakg_icd2)/(1+icit/Kicit_icd2+akg/Kakg_icd2)
Kakg_icd1=0.3 mM; Vf_icd1=10.2 mM_per_min; Vr_icd2=0.09965 mM_per_min; Kakg_icd2=0.6 mM; Vr_icd1=0.102 mM_per_min; Vf_icd2=9.965 mM_per_min; Kicit_icd1=0.03 mM; Kicit_icd2=0.06 mMReaction: akg => biosyn; icit, Rate Law: cell*0.0341*((Vf_icd1*icit/Kicit_icd1-Vr_icd1*akg/Kakg_icd1)/(1+icit/Kicit_icd1+akg/Kakg_icd1)+(Vf_icd2*icit/Kicit_icd2-Vr_icd2*akg/Kakg_icd2)/(1+icit/Kicit_icd2+akg/Kakg_icd2))
Vr_cs=0.648 mM_per_min; Kaca_cs=0.05 mM; Kcit_cs=0.12 mM; Kcoa_cs=0.5 mM; Vf_cs=64.8 mM_per_min; Koaa_cs=0.012 mMReaction: aca + oaa => coa + cit, Rate Law: cell*(Vf_cs*aca/Kaca_cs*oaa/Koaa_cs-Vr_cs*coa/Kcoa_cs*cit/Kcit_cs)/((1+aca/Kaca_cs+coa/Kcoa_cs)*(1+oaa/Koaa_cs+cit/Kcit_cs))
Vr_acn=0.312 mM_per_min; Kicit_acn=0.7 mM; Vf_acn=31.2 mM_per_min; Kcit_acn=1.7 mMReaction: cit => icit, Rate Law: cell*(Vf_acn*cit/Kcit_acn-Vr_acn*icit/Kicit_acn)/(1+cit/Kcit_acn+icit/Kicit_acn)
Kmal_fum=2.38 mM; Kfa_fum=0.25 mM; Vr_fum=87.7 mM_per_min; Vf_fum=87.7 mM_per_minReaction: fa => mal, Rate Law: cell*(Vf_fum*fa/Kfa_fum-Vr_fum*mal/Kmal_fum)/(1+fa/Kfa_fum+mal/Kmal_fum)
Kicit_icl2=1.3 mM; Kgly_icl2=1.3 mM; Vr_icl2=0.00391 mM_per_min; Ksuc_icl2=5.9 mM; Vf_icl2=0.391 mM_per_minReaction: icit => suc + gly, Rate Law: cell*(Vf_icl2*icit/Kicit_icl2-Vr_icl2*suc/Ksuc_icl2*gly/Kgly_icl2)/(1+icit/Kicit_icl2+suc/Ksuc_icl2+gly/Kgly_icl2+icit/Kicit_icl2*suc/Ksuc_icl2+suc/Ksuc_icl2*gly/Kgly_icl2)
Koaa_mdh=0.0443 mM; Vr_mdh=184.0 mM_per_min; Kmal_mdh=0.833 mM; Vf_mdh=184.0 mM_per_minReaction: mal => oaa, Rate Law: cell*(Vf_mdh*mal/Kmal_mdh-Vr_mdh*oaa/Koaa_mdh)/(1+mal/Kmal_mdh+oaa/Koaa_mdh)
Kmal_ms=1.0 mM; Vf_ms=20.0 mM_per_min; Kgly_ms=0.057 mM; Kaca_ms=0.03 mM; Vr_ms=0.2 mM_per_min; Kcoa_ms=0.1 mMReaction: gly + aca => mal + coa, Rate Law: cell*(Vf_ms*gly/Kgly_ms*aca/Kaca_ms-Vr_ms*mal/Kmal_ms*coa/Kcoa_ms)/((1+gly/Kgly_ms+mal/Kmal_ms)*(1+aca/Kaca_ms+coa/Kcoa_ms))

States:

NameDescription
gly[glyoxylic acid; Glyoxylate]
icit[isocitric acid; Isocitrate]
coa[coenzyme A; CoA]
mal[malic acid; Malate]
ssa[succinic semialdehyde; Succinate semialdehyde]
akg[2-oxoglutaric acid; 2-Oxoglutarate]
aca[acetyl-CoA; Acetyl-CoA]
cit[citric acid; Citrate]
oaa[oxaloacetic acid; Oxaloacetate]
biosynbiosyn
fa[fumaric acid; Fumarate]
suc[succinic acid; Succinate]
sca[succinyl-CoA; Succinyl-CoA]

Sips2015 - Glucose and non-esterified fatty acids (NEFA) metabolism: MODEL1806080001v0.0.1

This a model from the article: Model-Based Quantification of the Systemic Interplay between Glucose and Fatty Acids…

Details

In metabolic diseases such as Type 2 Diabetes and Non-Alcoholic Fatty Liver Disease, the systemic regulation of postprandial metabolite concentrations is disturbed. To understand this dysregulation, a quantitative and temporal understanding of systemic postprandial metabolite handling is needed. Of particular interest is the intertwined regulation of glucose and non-esterified fatty acids (NEFA), due to the association between disturbed NEFA metabolism and insulin resistance. However, postprandial glucose metabolism is characterized by a dynamic interplay of simultaneously responding regulatory mechanisms, which have proven difficult to measure directly. Therefore, we propose a mathematical modelling approach to untangle the systemic interplay between glucose and NEFA in the postprandial period. The developed model integrates data of both the perturbation of glucose metabolism by NEFA as measured under clamp conditions, and postprandial time-series of glucose, insulin, and NEFA. The model can describe independent data not used for fitting, and perturbations of NEFA metabolism result in an increased insulin, but not glucose, response, demonstrating that glucose homeostasis is maintained. Finally, the model is used to show that NEFA may mediate up to 30-45% of the postprandial increase in insulin-dependent glucose uptake at two hours after a glucose meal. In conclusion, the presented model can quantify the systemic interactions of glucose and NEFA in the postprandial state, and may therefore provide a new method to evaluate the disturbance of this interplay in metabolic disease. link: http://identifiers.org/pubmed/26356502

Sivakumar2011 - EGF Receptor Signaling Pathway: BIOMD0000000394v0.0.1

Sivakumar2011 - EGF Receptor Signaling Pathway EGFR belongs to the human epidermal receptor (HER) family of receptor ty…

Details

The Notch, Sonic Hedgehog (Shh), Wnt, and EGF pathways have long been known to influence cell fate specification in the developing nervous system. Here we attempted to evaluate the contemporary knowledge about neural stem cell differentiation promoted by various drug-based regulations through a systems biology approach. Our model showed the phenomenon of DAPT-mediated antagonism of Enhancer of split [E(spl)] genes and enhancement of Shh target genes by a SAG agonist that were effectively demonstrated computationally and were consistent with experimental studies. However, in the case of model simulation of Wnt and EGF pathways, the model network did not supply any concurrent results with experimental data despite the fact that drugs were added at the appropriate positions. This paves insight into the potential of crosstalks between pathways considered in our study. Therefore, we manually developed a map of signaling crosstalk, which included the species connected by representatives from Notch, Shh, Wnt, and EGF pathways and highlighted the regulation of a single target gene, Hes-1, based on drug-induced simulations. These simulations provided results that matched with experimental studies. Therefore, these signaling crosstalk models complement as a tool toward the discovery of novel regulatory processes involved in neural stem cell maintenance, proliferation, and differentiation during mammalian central nervous system development. To our knowledge, this is the first report of a simple crosstalk map that highlights the differential regulation of neural stem cell differentiation and underscores the flow of positive and negative regulatory signals modulated by drugs. link: http://identifiers.org/pubmed/21978399

Parameters:

NameDescription
kdiss_r6_s144 = 1.0; kass_r6_s144 = 1.0Reaction: s127 => s127; s144, Rate Law: s144*(kass_r6_s144*s127-kdiss_r6_s144*s127)
kM_r14_s27 = 0.038; kM_r14_s28 = 1.65; kcatn_r14 = 0.725; kcatp_r14 = 0.558Reaction: s27 => s28; s26, Rate Law: s26*(kcatp_r14/kM_r14_s27*s27-kcatn_r14/kM_r14_s28*s28)/(1+s27/kM_r14_s27+s28/kM_r14_s28)
kcatp_r8_s31 = 0.727; kM_r8_s124_s23 = 0.47; kcatn_r8_s31 = 0.636; kM_r8_s31_s23 = 0.614; kI_r8_s29 = 1.219; kM_r8_s31_s24 = 1.367; kI_r8_s22 = 0.583; kcatp_r8_s124 = 0.511; kI_r8_s33 = 0.293; kcatn_r8_s124 = 1.083; kM_r8_s124_s24 = 0.786Reaction: s23 => s24; s22, s29, s124, s33, s31, Rate Law: kI_r8_s22/(kI_r8_s22+s22)*kI_r8_s29/(kI_r8_s29+s29)*kI_r8_s33/(kI_r8_s33+s33)*(s124*(kcatp_r8_s124/kM_r8_s124_s23*s23-kcatn_r8_s124/kM_r8_s124_s24*s24)/(1+s23/kM_r8_s124_s23+s24/kM_r8_s124_s24)+s31*(kcatp_r8_s31/kM_r8_s31_s23*s23-kcatn_r8_s31/kM_r8_s31_s24*s24)/(1+s23/kM_r8_s31_s23+s24/kM_r8_s31_s24))
kcatp_r9 = 2.0; kcatn_r9 = 0.693; kM_r9_s25 = 0.626; kM_r9_s26 = 0.463Reaction: s25 => s26; s24, Rate Law: s24*(kcatp_r9/kM_r9_s25*s25-kcatn_r9/kM_r9_s26*s26)/(1+s25/kM_r9_s25+s26/kM_r9_s26)
kass_r15 = 2.0; kdiss_r15 = 0.074Reaction: s28 => s34, Rate Law: kass_r15*s28-kdiss_r15*s34
kdiss_r4_s144 = 1.0; kass_r4_s144 = 1.0Reaction: s124 + s125 => s124 + s126; s144, Rate Law: s144*(kass_r4_s144*s124*s125-kdiss_r4_s144*s124*s126)
kI_re11_s142 = 1.0; kM_re11_s129 = 1.0; Vp_re11 = 1.0; ki_re11_s129 = 1.0; kM_re11_s147 = 1.0Reaction: s129 + s147 => s144; s142, Rate Law: kI_re11_s142/(kI_re11_s142+s142)*Vp_re11*s129*s147/(ki_re11_s129*kM_re11_s147+kM_re11_s147*s129+kM_re11_s129*s147+s129*s147)
kcatn_r11 = 0.566; kM_r11_s30 = 1.021; kM_r11_s29 = 1.459; kcatp_r11 = 0.787Reaction: s29 => s30; s127, Rate Law: s127*(kcatp_r11/kM_r11_s29*s29-kcatn_r11/kM_r11_s30*s30)/(1+s29/kM_r11_s29+s30/kM_r11_s30)
kass_r17_s3 = 0.73; kdiss_r17_s3 = 1.13Reaction: s123 => s129; s3, Rate Law: s3*(kass_r17_s3*s123^2-kdiss_r17_s3*s129)
kass_r7_s144 = 1.0; kdiss_r7_s144 = 1.0Reaction: s21 => s22; s144, Rate Law: s144*(kass_r7_s144*s21-kdiss_r7_s144*s22)

States:

NameDescription
s29[Phosphatidylethanolamine-binding protein 1]
s27[MAP kinase activity]
s23[RAF proto-oncogene serine/threonine-protein kinase]
s124[Ras-like protein 1]
s24[RAF proto-oncogene serine/threonine-protein kinase]
s25[Dual specificity mitogen-activated protein kinase kinase 1]
s34Mitogenesis_br_Differentiation
s30[Phosphatidylethanolamine-binding protein 1]
s147[protein complex]
s123[Receptor protein-tyrosine kinase]
s26[Dual specificity mitogen-activated protein kinase kinase 1]
s21[RAC-alpha serine/threonine-protein kinase]
s22[RAC-alpha serine/threonine-protein kinase]
s28[MAP kinase activity]
s127[Protein kinase C alpha type]
s129[Receptor protein-tyrosine kinase]
s144[protein complex]
s126[GTP]
s125[GDP]

Sivakumar2011 - Hedgehog Signaling Pathway: BIOMD0000000395v0.0.1

Sivakumar2011 - Hedgehog Signaling Pathway This is the current model for the Hedgehog signaling pathway. The best data…

Details

The Notch, Sonic Hedgehog (Shh), Wnt, and EGF pathways have long been known to influence cell fate specification in the developing nervous system. Here we attempted to evaluate the contemporary knowledge about neural stem cell differentiation promoted by various drug-based regulations through a systems biology approach. Our model showed the phenomenon of DAPT-mediated antagonism of Enhancer of split [E(spl)] genes and enhancement of Shh target genes by a SAG agonist that were effectively demonstrated computationally and were consistent with experimental studies. However, in the case of model simulation of Wnt and EGF pathways, the model network did not supply any concurrent results with experimental data despite the fact that drugs were added at the appropriate positions. This paves insight into the potential of crosstalks between pathways considered in our study. Therefore, we manually developed a map of signaling crosstalk, which included the species connected by representatives from Notch, Shh, Wnt, and EGF pathways and highlighted the regulation of a single target gene, Hes-1, based on drug-induced simulations. These simulations provided results that matched with experimental studies. Therefore, these signaling crosstalk models complement as a tool toward the discovery of novel regulatory processes involved in neural stem cell maintenance, proliferation, and differentiation during mammalian central nervous system development. To our knowledge, this is the first report of a simple crosstalk map that highlights the differential regulation of neural stem cell differentiation and underscores the flow of positive and negative regulatory signals modulated by drugs. link: http://identifiers.org/pubmed/21978399

Parameters:

NameDescription
kass_r55 = 1.56Reaction: s158 => s75, Rate Law: kass_r55*s158
kdiss_r25 = 0.73; kass_r25 = 1.27Reaction: s160 => s161 + s69, Rate Law: kass_r25*s160-kdiss_r25*s161*s69
kdiss_r23_s21 = 1.0; kass_r23_s21 = 1.0Reaction: s159 => s68 + s160; s21, Rate Law: s21*(kass_r23_s21*s159-kdiss_r23_s21*s68*s160)
kass_r52 = 0.6; kdiss_r52 = 1.67Reaction: s140 => s75, Rate Law: kass_r52*s140-kdiss_r52*s75
kass_re24_s157 = 1.0Reaction: s148 + s150 => s159; s157, Rate Law: s157*kass_re24_s157*s148*s150
kcatp_r53 = 1.29; kM_r53_s70 = 0.79; kcatn_r53 = 1.62Reaction: s70 => s70; s48, Rate Law: s48*(kcatp_r53/kM_r53_s70*s70-kcatn_r53/kM_r53_s70*s70)/(1+s70/kM_r53_s70+s70/kM_r53_s70)
kass_r54 = 1.28; kdiss_r54 = 0.71Reaction: s70 + s71 => s158, Rate Law: kass_r54*s70*s71-kdiss_r54*s158
kass_r7 = 1.13; kdiss_r7 = 1.122Reaction: s7 + s1 => s21, Rate Law: kass_r7*s7*s1-kdiss_r7*s21
kass_r51 = 1.23; kdiss_r51 = 0.46Reaction: s135 + s128 => s140, Rate Law: kass_r51*s135*s128-kdiss_r51*s140
kass_r15_s21 = 1.53; kdiss_r15_s21 = 0.89Reaction: s46 + s9 => s48 + s10; s21, Rate Law: s21*(kass_r15_s21*s46*s9-kdiss_r15_s21*s48*s10)
kass_r26 = 1.33; kdiss_r26 = 0.61Reaction: s161 => s70, Rate Law: kass_r26*s161-kdiss_r26*s70
kM_r14_s46 = 0.215; kcatp_r14 = 1.146; kcatn_r14 = 1.75; kM_r14_s69 = 1.03Reaction: s69 => s46; s21, Rate Law: s21*(kcatp_r14/kM_r14_s69*s69-kcatn_r14/kM_r14_s46*s46)/(1+s69/kM_r14_s69+s46/kM_r14_s46)

States:

NameDescription
s150[protein complex]
s1[Protein patched homolog 1]
s48[protein complex]
s159[protein complex]
s135[Sin3-associated polypeptide 18]
s7[Sonic hedgehog protein]
s68[microtubule]
s75[positive regulation of hh target transcription factor activity]
s71[CREB-binding protein]
s128[protein complex]
s9[ATP]
s161Complex_br_(Su(fu)/Cubitus)
s10[ADP]
s21[protein complex]
s148smoothened
s46[protein complex]
s70[Cubitus interruptusCubitus interruptus, isoform A]
s140[protein complex]
s160[protein complex]
s69[protein complex]
s158[protein complex]

Sivakumar2011 - Notch Signaling Pathway: BIOMD0000000396v0.0.1

Sivakumar2011 - Notch Signaling Pathway Notch is a transmembrane receptor that mediates local cell-cell communication a…

Details

The Notch, Sonic Hedgehog (Shh), Wnt, and EGF pathways have long been known to influence cell fate specification in the developing nervous system. Here we attempted to evaluate the contemporary knowledge about neural stem cell differentiation promoted by various drug-based regulations through a systems biology approach. Our model showed the phenomenon of DAPT-mediated antagonism of Enhancer of split [E(spl)] genes and enhancement of Shh target genes by a SAG agonist that were effectively demonstrated computationally and were consistent with experimental studies. However, in the case of model simulation of Wnt and EGF pathways, the model network did not supply any concurrent results with experimental data despite the fact that drugs were added at the appropriate positions. This paves insight into the potential of crosstalks between pathways considered in our study. Therefore, we manually developed a map of signaling crosstalk, which included the species connected by representatives from Notch, Shh, Wnt, and EGF pathways and highlighted the regulation of a single target gene, Hes-1, based on drug-induced simulations. These simulations provided results that matched with experimental studies. Therefore, these signaling crosstalk models complement as a tool toward the discovery of novel regulatory processes involved in neural stem cell maintenance, proliferation, and differentiation during mammalian central nervous system development. To our knowledge, this is the first report of a simple crosstalk map that highlights the differential regulation of neural stem cell differentiation and underscores the flow of positive and negative regulatory signals modulated by drugs. link: http://identifiers.org/pubmed/21978399

Parameters:

NameDescription
kdiss_r13 = 2.0; kass_r13 = 0.5Reaction: s24 + s26 + s27 + s29 => s35, Rate Law: kass_r13*s24*s26*s27*s29-kdiss_r13*s35
kcatn_r16 = 1.0; kcatp_r16 = 1.0; kM_r16_s39 = 1.0; ki_r16_s39 = 1.0Reaction: s24 + s39 => s37; s38, Rate Law: (kcatp_r16/(ki_r16_s39*kM_r16_s39)*s38*s24*s39-kcatn_r16/kM_r16_s39*s38*s37)/(1+s24/ki_r16_s39+s39/ki_r16_s39+s24*s39/(ki_r16_s39*kM_r16_s39)+s37/kM_r16_s39)
kcatp_r9 = 1.5; kM_r9_s22 = 0.05; kcatn_r9 = 0.04; kM_r9_s7 = 1.0Reaction: s7 => s22; s23, Rate Law: s23*(kcatp_r9/kM_r9_s7*s7-kcatn_r9/kM_r9_s22*s22)/(1+s7/kM_r9_s7+s22/kM_r9_s22)
kM_r18_s4 = 1.0; ki_r18_s4 = 1.5; kcatn_r18 = 1.5; kcatp_r18 = 1.0Reaction: s1 + s4 => s41; s42, Rate Law: (kcatp_r18/(ki_r18_s4*kM_r18_s4)*s42*s1*s4-kcatn_r18/kM_r18_s4*s42*s41)/(1+s1/ki_r18_s4+s4/ki_r18_s4+s1*s4/(ki_r18_s4*kM_r18_s4)+s41/kM_r18_s4)
kcatp_r28 = 1.71; ki_r28_s41 = 1.28; kcatn_r28 = 1.48; kM_r28_s41 = 1.64Reaction: s7 + s41 => s67; s2, Rate Law: (kcatp_r28/(ki_r28_s41*kM_r28_s41)*s2*s7*s41-kcatn_r28/kM_r28_s41*s2*s67)/(1+s7/ki_r28_s41+s41/ki_r28_s41+s7*s41/(ki_r28_s41*kM_r28_s41)+s67/kM_r28_s41)
kI_r21_s2 = 1.5; kass_r21 = 1.5; kdiss_r21 = 1.5Reaction: s41 + s48 => s53; s2, Rate Law: kI_r21_s2/(kI_r21_s2+s2)*(kass_r21*s41*s48-kdiss_r21*s53)
kM_r26_s25 = 1.7; kM_r26_s64 = 1.61; kcatn_r26 = 1.0; kcatp_r26 = 0.5Reaction: s25 => s64; s65, Rate Law: s65*(kcatp_r26/kM_r26_s25*s25-kcatn_r26/kM_r26_s64*s64)/(1+s25/kM_r26_s25+s64/kM_r26_s64)
kM_r25_s15 = 1.5; kM_r25_s53 = 1.5; kcatn_r25 = 1.5; kM_r25_s60 = 1.25; kcatp_r25 = 1.0Reaction: s53 => s60 + s15; s21, Rate Law: s21*(kcatp_r25*s53/kM_r25_s53-kcatn_r25*s60/kM_r25_s60*s15/kM_r25_s15)/(s53/kM_r25_s53+(1+s60/kM_r25_s60)*(1+s15/kM_r25_s15))
kcatp_r29 = 1.86; kM_r29_s67 = 1.61; kM_r29_s18 = 0.15; kM_r29_s15 = 1.87; kcatn_r29 = 1.78Reaction: s67 => s18 + s15; s21, Rate Law: s21*(kcatp_r29*s67/kM_r29_s67-kcatn_r29*s18/kM_r29_s18*s15/kM_r29_s15)/(s67/kM_r29_s67+(1+s18/kM_r29_s18)*(1+s15/kM_r29_s15))
kM_r8_s63 = 1.5; kcatp_r8 = 0.5; kcatn_r8 = 1.5; kM_r8_s15 = 1.0; kM_r8_s19 = 2.0Reaction: s15 => s19 + s63; s82, Rate Law: s82*(kcatp_r8*s15/kM_r8_s15-kcatn_r8*s19/kM_r8_s19*s63/kM_r8_s63)/(s15/kM_r8_s15+(1+s19/kM_r8_s19)*(1+s63/kM_r8_s63))
kI_re16_s81 = 0.00594; kass_re16 = 0.004; kdiss_re16 = 2.0Reaction: s76 + s77 => s82; s81, Rate Law: kI_re16_s81/(kI_re16_s81+s81)*(kass_re16*s76*s77-kdiss_re16*s82)
kass_r31 = 0.055; kdiss_r31 = 2.0Reaction: s35 => s75, Rate Law: kass_r31*s35-kdiss_r31*s75
kdiss_r10 = 0.01; kI_r10_s25 = 1.0; kass_r10 = 2.0Reaction: s63 => s24; s25, Rate Law: kI_r10_s25/(kI_r10_s25+s25)*(kass_r10*s63-kdiss_r10*s24)
kass_r30 = 1.95Reaction: s32 => s75, Rate Law: kass_r30*s32
kcatp_r11 = 0.5; kM_r11_s32 = 1.0; kcatn_r11 = 0.5; kM_r11_s26 = 1.5; kM_r11_s28 = 1.0Reaction: s32 => s26 + s28; s24, Rate Law: s24*(kcatp_r11*s32/kM_r11_s32-kcatn_r11*s26/kM_r11_s26*s28/kM_r11_s28)/(s32/kM_r11_s32+(1+s26/kM_r11_s26)*(1+s28/kM_r11_s28))
kass_r17 = 1.5; kdiss_r17 = 1.5Reaction: s37 => s40, Rate Law: kass_r17*s37-kdiss_r17*s40

States:

NameDescription
s76[CCO:U0000005]
s7[Delta-like protein 1]
s24[protein complex]
s35[protein complex]
s18[protein complex]
s37[protein complex]
s40[protein]
s53[protein complex]
s19[protein complex]
s32[protein complex]
s22[protein]
s77[CCO:U0000005]
s15[protein complex]
s1[NOTCH1 protein]
s48[Serrate]
s67[protein complex]
s63[protein complex]
s41[NOTCH1 protein]
s25[Protein numb homolog]
s75[Basic helix-loop-helix transcription factorE(Spl)]
s4[L-fucose]
s82[Gamma-secretase subunit PEN-2; Gamma-secretase subunit APH-1A]
s26[Suppressor of hairless protein]
s64a25_degraded
s28CoR
s39[CCO:F0004655]
s60[protein complex]
s29[protein]
s27[Mastermind-like protein 1]

Sivakumar2011_NeuralStemCellDifferentiation_Crosstalk: BIOMD0000000398v0.0.1

Sivakumar2011_NeuralStemCellDifferentiation_CrosstalkThis model is generated by integrating [BIOMD0000000394](http://ww…

Details

The Notch, Sonic Hedgehog (Shh), Wnt, and EGF pathways have long been known to influence cell fate specification in the developing nervous system. Here we attempted to evaluate the contemporary knowledge about neural stem cell differentiation promoted by various drug-based regulations through a systems biology approach. Our model showed the phenomenon of DAPT-mediated antagonism of Enhancer of split [E(spl)] genes and enhancement of Shh target genes by a SAG agonist that were effectively demonstrated computationally and were consistent with experimental studies. However, in the case of model simulation of Wnt and EGF pathways, the model network did not supply any concurrent results with experimental data despite the fact that drugs were added at the appropriate positions. This paves insight into the potential of crosstalks between pathways considered in our study. Therefore, we manually developed a map of signaling crosstalk, which included the species connected by representatives from Notch, Shh, Wnt, and EGF pathways and highlighted the regulation of a single target gene, Hes-1, based on drug-induced simulations. These simulations provided results that matched with experimental studies. Therefore, these signaling crosstalk models complement as a tool toward the discovery of novel regulatory processes involved in neural stem cell maintenance, proliferation, and differentiation during mammalian central nervous system development. To our knowledge, this is the first report of a simple crosstalk map that highlights the differential regulation of neural stem cell differentiation and underscores the flow of positive and negative regulatory signals modulated by drugs. link: http://identifiers.org/pubmed/21978399

Parameters:

NameDescription
kass_re35_s89 = 1.0; kdiss_re35_s89 = 1.0Reaction: s88 => s73; s89, Rate Law: s89*(kass_re35_s89*s88-kdiss_re35_s89*s73)
kass_re36 = 1.0; kdiss_re36 = 1.0; kI_re36_s101 = 1.0Reaction: s96 + s98 => s100; s101, Rate Law: kI_re36_s101/(kI_re36_s101+s101)*(kass_re36*s96*s98-kdiss_re36*s100)
kdiss_re33 = 1.0; kass_re33 = 1.0Reaction: s81 + s83 => s85, Rate Law: kass_re33*s81*s83-kdiss_re33*s85
kdiss_re31 = 1.0; kass_re31 = 1.0Reaction: s53 + s68 => s72, Rate Law: kass_re31*s53*s68-kdiss_re31*s72
kcatn_re40 = 1.0; kcatp_re40 = 1.0; ki_re40_s124 = 1.0; kM_re40_s124 = 1.0Reaction: s122 + s124 => s135; s111, Rate Law: (kcatp_re40/(ki_re40_s124*kM_re40_s124)*s111*s122*s124-kcatn_re40/kM_re40_s124*s111*s135)/(1+s122/ki_re40_s124+s124/ki_re40_s124+s122*s124/(ki_re40_s124*kM_re40_s124)+s135/kM_re40_s124)
kass_re34_s85 = 1.0; kass_re34_s89 = 1.0; kdiss_re34_s89 = 1.0; kdiss_re34_s85 = 1.0Reaction: s88 => s88; s85, s89, Rate Law: s85*(kass_re34_s85*s88-kdiss_re34_s85*s88)+s89*(kass_re34_s89*s88-kdiss_re34_s89*s88)
kM_re29_s60_s58 = 1.0; kV_re29_s60 = 1.0; kG_s57 = 1.0; kM_re29_s60_s53 = 1.0; kM_re29_s60_s57 = 1.0; kG_s58 = 1.0; kG_s53 = 1.0; kI_re29_s61 = 1.0Reaction: s57 => s53 + s58; s60, s61, Rate Law: kI_re29_s61/(kI_re29_s61+s61)*s60*kV_re29_s60*(s57/kM_re29_s60_s57*(kG_s57*kM_re29_s60_s57/(kG_s53*kM_re29_s60_s53*kG_s58*kM_re29_s60_s58))^(0.5)-s53/kM_re29_s60_s53*s58/kM_re29_s60_s58*(kG_s53*kM_re29_s60_s53*kG_s58*kM_re29_s60_s58/(kG_s57*kM_re29_s60_s57))^(0.5))/(s57/kM_re29_s60_s57+(1+s53/kM_re29_s60_s53)*(1+s58/kM_re29_s60_s58))
kass_re32 = 1.0; kdiss_re32 = 1.0Reaction: s72 => s73, Rate Law: kass_re32*s72-kdiss_re32*s73
kI_re42_s147 = 1.0; kdiss_re42 = 1.0; kI_re42_s135 = 1.0; kass_re42 = 1.0Reaction: s142 + s144 => s146; s147, s135, Rate Law: kI_re42_s147/(kI_re42_s147+s147)*kI_re42_s135/(kI_re42_s135+s135)*(kass_re42*s142*s144-kdiss_re42*s146)
kass_re37 = 1.0; kdiss_re37 = 1.0Reaction: s100 => s73, Rate Law: kass_re37*s100-kdiss_re37*s73
kass_re38 = 1.0; kdiss_re38 = 1.0Reaction: s107 + s109 => s111, Rate Law: kass_re38*s107*s109-kdiss_re38*s111
kass_re43 = 1.0; kdiss_re43 = 1.0Reaction: s144 => s73, Rate Law: kass_re43*s144-kdiss_re43*s73

States:

NameDescription
s146[protein complex]
s107[Protein Wnt-3a]
s111Complex Wnt-Frzzl
s124[protein complex]
s135[protein complex]
s142[Glycogen synthase kinase-3 beta]
s109[Frizzled]
s57[Neurogenic locus Notch protein]
s58[protein]
s53[Neurogenic locus notch homolog protein 1]
s122[Dishevelled, dsh homolog 1 (Drosophila)]
s100[protein complex]
s81[Sonic hedgehog protein]
s72[protein complex]
s96[Pro-epidermal growth factor]
s98[EGFR protein]
s144[Catenin beta-1]
s68[Recombining binding protein suppressor of hairless]
s73[139605]
s88[Smoothened homolog]
s85[protein complex]
s83[Protein patched homolog 1]

Sivakumar2011_WntSignalingPathway: BIOMD0000000397v0.0.1

Sivakumar2011_WntSignalingPathwayThe secreted protein Wnt activates the heptahelical receptor Frizzled on nieghboring ce…

Details

The Notch, Sonic Hedgehog (Shh), Wnt, and EGF pathways have long been known to influence cell fate specification in the developing nervous system. Here we attempted to evaluate the contemporary knowledge about neural stem cell differentiation promoted by various drug-based regulations through a systems biology approach. Our model showed the phenomenon of DAPT-mediated antagonism of Enhancer of split [E(spl)] genes and enhancement of Shh target genes by a SAG agonist that were effectively demonstrated computationally and were consistent with experimental studies. However, in the case of model simulation of Wnt and EGF pathways, the model network did not supply any concurrent results with experimental data despite the fact that drugs were added at the appropriate positions. This paves insight into the potential of crosstalks between pathways considered in our study. Therefore, we manually developed a map of signaling crosstalk, which included the species connected by representatives from Notch, Shh, Wnt, and EGF pathways and highlighted the regulation of a single target gene, Hes-1, based on drug-induced simulations. These simulations provided results that matched with experimental studies. Therefore, these signaling crosstalk models complement as a tool toward the discovery of novel regulatory processes involved in neural stem cell maintenance, proliferation, and differentiation during mammalian central nervous system development. To our knowledge, this is the first report of a simple crosstalk map that highlights the differential regulation of neural stem cell differentiation and underscores the flow of positive and negative regulatory signals modulated by drugs. link: http://identifiers.org/pubmed/21978399

Parameters:

NameDescription
kass_r107 = 0.91; kdiss_r107 = 1.056Reaction: s239 => s5, Rate Law: kass_r107*s239-kdiss_r107*s5
kass_r67 = 0.86; kdiss_r67 = 0.7Reaction: s188 + s172 => s305, Rate Law: kass_r67*s188*s172-kdiss_r67*s305
kass_r66 = 1.99; kdiss_r66 = 0.036Reaction: s183 + s173 => s188, Rate Law: kass_r66*s183*s173-kdiss_r66*s188
kass_re65 = 1.68Reaction: s260 => s232, Rate Law: kass_re65*s260
kdiss_r105 = 1.62; kass_r105 = 0.48Reaction: s292 => s37, Rate Law: kass_r105*s292-kdiss_r105*s37
kass_r91 = 0.36; kdiss_r91 = 1.16Reaction: s266 => s155 + s267, Rate Law: kass_r91*s266-kdiss_r91*s155*s267
kass_r54 = 0.8; kdiss_r54 = 1.7Reaction: s123 + s75 => s159, Rate Law: kass_r54*s123*s75-kdiss_r54*s159
kass_r58 = 1.74; kdiss_r58 = 0.25Reaction: s36 => s232, Rate Law: kass_r58*s36-kdiss_r58*s232
kass_r48 = 0.85; kdiss_r48 = 1.36Reaction: s123 + s46 => s129, Rate Law: kass_r48*s123*s46-kdiss_r48*s129
kass_r1 = 0.784; kdiss_r1 = 0.82Reaction: s5 + s1 => s16, Rate Law: kass_r1*s5*s1-kdiss_r1*s16
kass_r103 = 0.45; kdiss_r103 = 1.277Reaction: s288 + s102 => s292, Rate Law: kass_r103*s288*s102-kdiss_r103*s292
kass_r98 = 1.97; kdiss_r98 = 1.09Reaction: s275 => s101 + s278, Rate Law: kass_r98*s275-kdiss_r98*s101*s278
kass_r63 = 1.77; kdiss_r63 = 0.61Reaction: s174 + s232 => s176, Rate Law: kass_r63*s174*s232-kdiss_r63*s176
kass_r64 = 1.29; kdiss_r64 = 0.72Reaction: s176 + s170 => s179, Rate Law: kass_r64*s176*s170-kdiss_r64*s179
kass_r68 = 2.0Reaction: s305 => s195, Rate Law: kass_r68*s305
kass_r65 = 1.8; kdiss_r65 = 0.004Reaction: s179 + s171 => s183, Rate Law: kass_r65*s179*s171-kdiss_r65*s183
kdiss_r5 = 0.92; kass_r5 = 1.15Reaction: s28 + s16 => s27, Rate Law: kass_r5*s28*s16-kdiss_r5*s27
kass_re64 = 0.83Reaction: s270 => s232, Rate Law: kass_re64*s270
kdiss_r47 = 0.81; kass_r47 = 1.31Reaction: s121 + s36 => s123, Rate Law: kass_r47*s121*s36-kdiss_r47*s123
kdiss_r106 = 1.13; kass_r106 = 0.05Reaction: s286 => s30, Rate Law: kass_r106*s286-kdiss_r106*s30
kass_r102 = 0.163; kdiss_r102 = 1.65Reaction: s286 + s31 => s288, Rate Law: kass_r102*s286*s31-kdiss_r102*s288
kdiss_r96 = 0.183; kass_r96 = 1.45Reaction: s159 + s268 => s275, Rate Law: kass_r96*s159*s268-kdiss_r96*s275
kass_r92 = 0.58; kdiss_r92 = 0.92Reaction: s267 => s61 + s260, Rate Law: kass_r92*s267-kdiss_r92*s61*s260
kdiss_r88 = 1.09; kass_r88 = 0.2Reaction: s252 + s61 => s259, Rate Law: kass_r88*s252*s61-kdiss_r88*s259
kass_r90 = 0.27; kdiss_r90 = 1.028Reaction: s259 + s268 => s266, Rate Law: kass_r90*s259*s268-kdiss_r90*s266
kass_r85_s30 = 0.7; kdiss_r85_s30 = 0.649Reaction: s129 + s32 => s245 + s33; s30, Rate Law: s30*(kass_r85_s30*s129*s32-kdiss_r85_s30*s245*s33)
kass_r104_s30 = 0.39; kdiss_r104_s30 = 1.278Reaction: s107 + s32 => s286 + s33; s27, s30, Rate Law: s30*(kass_r104_s30*s107*s32-kdiss_r104_s30*s286*s33)
kdiss_r99 = 0.854; kass_r99 = 0.51Reaction: s278 => s164 + s270, Rate Law: kass_r99*s278-kdiss_r99*s164*s270
kI_r86_s304 = 1.43; kass_r86_s37 = 0.87; kdiss_r86_s37 = 1.32Reaction: s245 + s32 + s32 + s32 => s252 + s33 + s33 + s33; s37, s304, Rate Law: kI_r86_s304/(kI_r86_s304+s304)*s37*(kass_r86_s37*s245*s32*s32*s32-kdiss_r86_s37*s252*s33*s33*s33)

States:

NameDescription
s107Complex_br_(Dishevelled/Beta-Arrestin/_br_Frodo)
s260[Catenin beta-1]
s159Complex_br_(Adenomatous Polyposis Coli/Axin/_br_PP2A/_Beta_-Catenin/_br_Siah-1/Ebi)
s172[CREB-binding protein]
s232[Catenin beta-1]
s5[Protein Wnt-3a]
s121Complex_br_(Axin/PP2A/_br_Adenomatous Polyposis Coli)
s278Complex_br_(Adenomatous Polyposis Coli/_Beta_-Catenin/_br_Axin/PP2A)
s61[Beta-TrCP]
s37[Glycogen synthase kinase-3 beta]
s36[Catenin beta-1]
s183Complex_br_(Bcl9/_Beta_-Catenin/_br_TCF/Smad4/_br_Pygo)
s31[Casein kinase II subunit beta; Casein kinase II subunit alpha]
s266Complex_br_(Adenomatous Polyposis Coli/Axin/_br_PP2A/Diversin/_br_Casein Kinase 1/_Beta_-Catenin/_br__beta_TrCP/Glycogen Synthase Kinase-3_Beta_)
s46[Diversin]
s268Ubiquitin
s129Complex_br_(Adenomatous Polyposis Coli/Axin/_br_Diversin/_Beta_-Catenin/_br_PP2A)
s1[Frizzled]
s292Complex_br_(Dishevelled/Casein Kinase 2/_br_Beta-Arrestin/Frodo/_br_FRAT)
s267Complex_br_(_beta_TrCP/_Beta_-Catenin)
s75Complex_br_(Ebi/Siah-1)
s33[ADP]
s101Complex_br_(Siah-1/Ebi)
s16Complex_br_(Wnt/Frizzled)
s179Complex_br_(TCF/_Beta_-Catenin/_br_Smad4/Bcl9)
s155Complex_br_(Adenomatous Polyposis Coli/Axin/_br_Diversin/Casein Kinase 1/_br_Glycogen Synthase Kinase-3_Beta_/PP2A)
s28[Low-density lipoprotein receptor-related protein 6; Low-density lipoprotein receptor-related protein 5]
s174Complex_br_(TCF/Smad4)
s270[Catenin beta-1]
s245Complex_br_(Adenomatous Polyposis Coli/_Beta_-Catenin/_br_Axin/PP2A/_br_Diversin/Casein Kinase 1)
s252Complex_br_(Adenomatous Polyposis Coli/_Beta_-Catenin/_br_Glycogen Synthase Kinase-3_Beta_/Axin/_br_PP2A/Diversin/_br_Casein Kinase 1)
s239[Wingless-type MMTV integration site family member 3a]
s32[ATP]
s170[B-cell CLL/lymphoma 9 protein]
s195Wnt Target Genes
s305Complex_br_(Bcl9/Pygo/../Smad4)
s275Complex_br_(Adenomatous Polyposis Coli/_Beta_-Catenin/_br_Siah-1/Ebi/_br_Axin/PP2A)
s164Complex_br_(Adenomatous Polyposis Coli/Axin/_br_PP2A)
s176Complex_br_(TCF/Smad4/_br__Beta_-Catenin)
s188Complex_br_(_Beta_-Catenin/TCF/_br_Smad4/Bcl9/_br_Pygo/SWI/_br_SNF)
s30[Casein kinase I isoform alpha]
s171[Protein pygopus]
s123Complex_br_(Adenomatous Polyposis Coli/Axin/_br__Beta_-Catenin/PP2A)
s288Complex_br_(Dishevelled/Beta-Arrestin/_br_Frodo/Casein Kinase 2)
s173[SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily A member 5]
s286Complex_br_(Dishevelled/Beta-Arrestin/_br_Frodo)
s259Complex_br_(Adenomatous Polyposis Coli/Axin/_br_PP2A/Diversin/_br_Casein Kinase 1/_Beta_-Catenin/_br__beta_TrCP/Glycogen Synthase Kinase-3_Beta_)
s102[Proto-oncogene FRAT1]
s27Complex_br_(Frizzled/Wnt/_br_LRP5/6)

Sizek2019 - PI3K_growth_CellCycle_Apoptosis: MODEL2006170002v0.0.1

This 89-node Boolean model of mammalian growth factor signaling can reproduce oscillations in PI3K signaling in cycling…

Details

The PI3K/AKT signaling pathway plays a role in most cellular functions linked to cancer progression, including cell growth, proliferation, cell survival, tissue invasion and angiogenesis. It is generally recognized that hyperactive PI3K/AKT1 are oncogenic due to their boost to cell survival, cell cycle entry and growth-promoting metabolism. That said, the dynamics of PI3K and AKT1 during cell cycle progression are highly nonlinear. In addition to negative feedback that curtails their activity, protein expression of PI3K subunits has been shown to oscillate in dividing cells. The low-PI3K/low-AKT1 phase of these oscillations is required for cytokinesis, indicating that oncogenic PI3K may directly contribute to genome duplication. To explore this, we construct a Boolean model of growth factor signaling that can reproduce PI3K oscillations and link them to cell cycle progression and apoptosis. The resulting modular model reproduces hyperactive PI3K-driven cytokinesis failure and genome duplication and predicts the molecular drivers responsible for these failures by linking hyperactive PI3K to mis-regulation of Polo-like kinase 1 (Plk1) expression late in G2. To do this, our model captures the role of Plk1 in cell cycle progression and accurately reproduces multiple effects of its loss: G2 arrest, mitotic catastrophe, chromosome mis-segregation / aneuploidy due to premature anaphase, and cytokinesis failure leading to genome duplication, depending on the timing of Plk1 inhibition along the cell cycle. Finally, we offer testable predictions on the molecular drivers of PI3K oscillations, the timing of these oscillations with respect to division, and the role of altered Plk1 and FoxO activity in genome-level defects caused by hyperactive PI3K. Our model is an important starting point for the predictive modeling of cell fate decisions that include AKT1-driven senescence, as well as the non-intuitive effects of drugs that interfere with mitosis. link: http://identifiers.org/pubmed/30875364

Sluka2016 - Acetaminophen PBPK: BIOMD0000000619v0.0.1

# Basic PBPK (Physiologically Based PharmacoKinetic) model of Acetaminophen. This is a basic model of Acetaminophen (APA…

Details

We describe a multi-scale, liver-centric in silico modeling framework for acetaminophen pharmacology and metabolism. We focus on a computational model to characterize whole body uptake and clearance, liver transport and phase I and phase II metabolism. We do this by incorporating sub-models that span three scales; Physiologically Based Pharmacokinetic (PBPK) modeling of acetaminophen uptake and distribution at the whole body level, cell and blood flow modeling at the tissue/organ level and metabolism at the sub-cellular level. We have used standard modeling modalities at each of the three scales. In particular, we have used the Systems Biology Markup Language (SBML) to create both the whole-body and sub-cellular scales. Our modeling approach allows us to run the individual sub-models separately and allows us to easily exchange models at a particular scale without the need to extensively rework the sub-models at other scales. In addition, the use of SBML greatly facilitates the inclusion of biological annotations directly in the model code. The model was calibrated using human in vivo data for acetaminophen and its sulfate and glucuronate metabolites. We then carried out extensive parameter sensitivity studies including the pairwise interaction of parameters. We also simulated population variation of exposure and sensitivity to acetaminophen. Our modeling framework can be extended to the prediction of liver toxicity following acetaminophen overdose, or used as a general purpose pharmacokinetic model for xenobiotics. link: http://identifiers.org/pubmed/27636091

Parameters:

NameDescription
QRest = 188.8 Volumetric_FlowReaction: CArt => CRest, Rate Law: QRest*CArt/VArt
kGutabs = 1.5 first_order_rate_constantReaction: AGutlumen => CGut, Rate Law: kGutabs*AGutlumen
Kkidney2plasma = 1.0 dimensionless; Qgfr = 0.9438 Volumetric_FlowReaction: CKidney => CTubules, Rate Law: Qgfr/VKidney*CKidney/Kkidney2plasma
QGut = 74.42 Volumetric_FlowReaction: CArt => CGut, Rate Law: QGut/VArt*CArt
CLmetabolism = 9.5 first_order_rate_constant; Kliver2plasma = 1.0 dimensionless; Fraction_unbound_plasma = 0.8 dimensionlessReaction: CLiver => CMetabolized, Rate Law: CLmetabolism*CLiver/(Kliver2plasma*Fraction_unbound_plasma)
Ratioblood2plasma = 1.09 dimensionless; Fraction_unbound_plasma = 0.8 dimensionless; KRest2plasma = 1.6 dimensionless; QRest = 188.8 Volumetric_FlowReaction: CRest => CVen, Rate Law: QRest/VRest*CRest*Ratioblood2plasma/(KRest2plasma*Fraction_unbound_plasma)
QGut = 74.42 Volumetric_Flow; Kliver2plasma = 1.0 dimensionless; QLiver = 19.42 Volumetric_Flow; Ratioblood2plasma = 1.09 dimensionless; Fraction_unbound_plasma = 0.8 dimensionlessReaction: CLiver => CVen, Rate Law: (QLiver+QGut)/VLiver*CLiver*Ratioblood2plasma/(Kliver2plasma*Fraction_unbound_plasma)
QCardiac = 363.0 Volumetric_FlowReaction: CLung => CArt, Rate Law: QCardiac/VLung*CLung
QLiver = 19.42 Volumetric_FlowReaction: CArt => CLiver, Rate Law: QLiver/VArt*CArt
QKidney = 80.37 Volumetric_FlowReaction: CArt => CKidney, Rate Law: QKidney/VArt*CArt
Kkidney2plasma = 1.0 dimensionless; QKidney = 80.37 Volumetric_Flow; Ratioblood2plasma = 1.09 dimensionless; Fraction_unbound_plasma = 0.8 dimensionlessReaction: CKidney => CVen, Rate Law: QKidney/VKidney*CKidney*Ratioblood2plasma/(Kkidney2plasma*Fraction_unbound_plasma)

States:

NameDescription
CVenCVen
CArtCArt
CKidneyCKidney
CMetabolizedCMetabolized
CGutCGut
AGutlumenAGutlumen
CTubulesCTubules
CLungCLung
CLiverCLiver
CRestCRest

Sluka2016 - Acetaminophen metabolism: BIOMD0000000624v0.0.1

Sluka2016 - Acetaminophen metabolism**Liver metabolism of Acetaminophen:** Acetaminophen (APAP) is metabolized in the li…

Details

We describe a multi-scale, liver-centric in silico modeling framework for acetaminophen pharmacology and metabolism. We focus on a computational model to characterize whole body uptake and clearance, liver transport and phase I and phase II metabolism. We do this by incorporating sub-models that span three scales; Physiologically Based Pharmacokinetic (PBPK) modeling of acetaminophen uptake and distribution at the whole body level, cell and blood flow modeling at the tissue/organ level and metabolism at the sub-cellular level. We have used standard modeling modalities at each of the three scales. In particular, we have used the Systems Biology Markup Language (SBML) to create both the whole-body and sub-cellular scales. Our modeling approach allows us to run the individual sub-models separately and allows us to easily exchange models at a particular scale without the need to extensively rework the sub-models at other scales. In addition, the use of SBML greatly facilitates the inclusion of biological annotations directly in the model code. The model was calibrated using human in vivo data for acetaminophen and its sulfate and glucuronate metabolites. We then carried out extensive parameter sensitivity studies including the pairwise interaction of parameters. We also simulated population variation of exposure and sensitivity to acetaminophen. Our modeling framework can be extended to the prediction of liver toxicity following acetaminophen overdose, or used as a general purpose pharmacokinetic model for xenobiotics. link: http://identifiers.org/pubmed/27636091

Parameters:

NameDescription
Vmax_2E1_APAP = 2.0E-5 flux; Km_2E1_APAP = 1.29 millimolarReaction: APAP => NAPQI, Rate Law: Vmax_2E1_APAP*APAP/(Km_2E1_APAP+APAP)
kGsh = 1.0E-4 first_order_rate_constant; GSHmax = 10.0 millimolarReaction: X1 => GSH, Rate Law: kGsh*(GSHmax-GSH)*compartment
Km_PhaseIIEnzGlu_APAP = 1.0 millimolar; Vmax_PhaseIIEnzGlu_APAP = 0.001 fluxReaction: APAP => APAPconj_Glu, Rate Law: Vmax_PhaseIIEnzGlu_APAP*APAP/(Km_PhaseIIEnzGlu_APAP+APAP)
kNapqiGsh = 0.1 second_order_rate_constantReaction: GSH + NAPQI => NAPQIGSH, Rate Law: kNapqiGsh*NAPQI*GSH*compartment*compartment
Km_PhaseIIEnzSul_APAP = 0.2 millimolar; Vmax_PhaseIIEnzSul_APAP = 1.75E-4 fluxReaction: APAP => APAPconj_Sul, Rate Law: Vmax_PhaseIIEnzSul_APAP*APAP/(Km_PhaseIIEnzSul_APAP+APAP)

States:

NameDescription
APAPconj Sul[paracetamol sulfate]
APAP[paracetamol]
NAPQI[N-acetyl-1,4-benzoquinone imine]
NAPQIGSH[acetaminophen glutathione conjugate]
APAPconj Glu[acetaminophen O-beta-D-glucosiduronic acid]
X1X1
GSH[glutathione]

Smallbone2009 - FBA model-Geometric Perspective: MODEL1105180000v0.0.1

This model is from the article: Flux balance analysis: a geometric perspective. Smallbone K, Simeonidis E. J Theor B…

Details

Advances in the field of bioinformatics have led to reconstruction of genome-scale networks for a number of key organisms. The application of physicochemical constraints to these stoichiometric networks allows researchers, through methods such as flux balance analysis, to highlight key sets of reactions necessary to achieve particular objectives. The key benefits of constraint-based analysis lie in the minimal knowledge required to infer systemic properties. However, network degeneracy leads to a large number of flux distributions that satisfy any objective; moreover, these distributions may be dominated by biologically irrelevant internal cycles. By examining the geometry underlying the problem, we define two methods for finding a unique solution within the space of all possible flux distributions; such a solution contains no internal cycles, and is representative of the space as a whole. The first method draws on typical geometric knowledge, but cannot be applied to large networks because of the high computational complexity of the problem. Thus a second method, an iteration of linear programs which scales easily to the genome scale, is defined. The algorithm is run on four recent genome-scale models, and unique flux solutions are found. The algorithm set out here will allow researchers in flux balance analysis to exchange typical solutions to their models in a reproducible format. Moreover, having found a single solution, statistical analyses such as correlations may be performed. link: http://identifiers.org/pubmed/19490860

Smallbone2010_Genome_Scale_Yeast_Kinetics: MODEL1001200000v0.0.1

This is the model described in the article: Towards a genome-scale kinetic model of cellular metabolism Smallbone K,…

Details

Advances in bioinformatic techniques and analyses have led to the availability of genome-scale metabolic reconstructions. The size and complexity of such networks often means that their potential behaviour can only be analysed with constraint-based methods. Whilst requiring minimal experimental data, such methods are unable to give insight into cellular substrate concentrations. Instead, the long-term goal of systems biology is to use kinetic modelling to characterize fully the mechanics of each enzymatic reaction, and to combine such knowledge to predict system behaviour.We describe a method for building a parameterized genome-scale kinetic model of a metabolic network. Simplified linlog kinetics are used and the parameters are extracted from a kinetic model repository. We demonstrate our methodology by applying it to yeast metabolism. The resultant model has 956 metabolic reactions involving 820 metabolites, and, whilst approximative, has considerably broader remit than any existing models of its type. Control analysis is used to identify key steps within the system.Our modelling framework may be considered a stepping-stone toward the long-term goal of a fully-parameterized model of yeast metabolism. The model is available in SBML format from the BioModels database (BioModels ID: MODEL1001200000) and at http://www.mcisb.org/resources/genomescale/. link: http://identifiers.org/pubmed/20109182

Smallbone2011_TrehaloseBiosynthesis: BIOMD0000000380v0.0.1

This model is from the article: Building a Kinetic Model of Trehalose Biosynthesis in Saccharomyces cerevisiae. Smal…

Details

In this chapter, we describe the steps needed to create a kinetic model of a metabolic pathway based on kinetic data from experimental measurements and literature review. Our methodology is presented by utilizing the example of trehalose metabolism in yeast. The biology of the trehalose cycle is briefly reviewed and discussed. link: http://identifiers.org/pubmed/21943906

Parameters:

NameDescription
Keq=0.3 dimensionless; Kf6p=0.29 mM; shock=1.0 dimensionless; heat = 0.0 dimensionless; Kg6p=1.4 mM; Vmax=1071.0 mM per minReaction: g6p => f6p, Rate Law: cell*shock^heat*Vmax/Kg6p*(g6p-f6p/Keq)/(1+g6p/Kg6p+f6p/Kf6p)
Kudg=0.886 mM; activity=1.0 dimensionless; heat = 0.0 dimensionless; Vmax=1.371 mM per min; shock=12.0 dimensionless; Kg6p=3.8 mMReaction: g6p + udg => t6p + udp + h, Rate Law: cell*activity*shock^heat*Vmax*g6p*udg/(Kg6p*Kudg)/((1+g6p/Kg6p)*(1+udg/Kudg))
Kg1p=0.023 mM; heat = 0.0 dimensionless; Vmax=0.3545 mM per min; Kg6p=0.05 mM; shock=16.0 dimensionless; Keq=0.1667 dimensionlessReaction: g6p => g1p, Rate Law: cell*shock^heat*Vmax/Kg6p*(g6p-g1p/Keq)/(1+g6p/Kg6p+g1p/Kg1p)
Kt6p=0.5 mM; heat = 0.0 dimensionless; Vmax=6.5 mM per min; shock=18.0 dimensionlessReaction: t6p + h2o => trh + pho, Rate Law: cell*shock^heat*Vmax*t6p/Kt6p/(1+t6p/Kt6p)
Kg6p=30.0 mM; Kglc=0.08 mM; Katp=0.15 mM; heat = 0.0 dimensionless; Keq=2000.0 dimensionless; shock=8.0 dimensionless; Vmax=289.6 mM per min; Kadp=0.23 mM; Kit6p=0.04 mMReaction: glc + atp => g6p + adp + h; t6p, Rate Law: cell*shock^heat*Vmax/(Kglc*Katp)*(glc*atp-g6p*adp/Keq)/((1+glc/Kglc+g6p/Kg6p+t6p/Kit6p)*(1+atp/Katp+adp/Kadp))
Vmax=15.2 mM per min; heat = 0.0 dimensionless; shock=6.0 dimensionless; Ktrh=2.99 mMReaction: trh + h2o => glc, Rate Law: cell*shock^heat*Vmax*trh/Ktrh/(1+trh/Ktrh)
heat = 0.0 dimensionless; Vmax=97.24 mM per min; shock=8.0 dimensionless; Ki=0.91 dimensionless; Kglc=1.1918 mMReaction: glx => glc, Rate Law: cell*shock^heat*Vmax*(glx-glc)/Kglc/(1+(glx+glc)/Kglc+Ki*glx*glc/Kglc^2)
Kiutp=0.11 mM; heat = 0.0 dimensionless; Kg1p=0.32 mM; Kutp=0.11 mM; Vmax=36.82 mM per min; shock=16.0 dimensionless; Kiudg=0.0035 mMReaction: g1p + utp + h => udg + ppi, Rate Law: cell*shock^heat*Vmax*utp*g1p/(Kutp*Kg1p)/(Kiutp/Kutp+utp/Kutp+g1p/Kg1p+utp*g1p/(Kutp*Kg1p)+Kiutp/Kutp*udg/Kiudg+g1p*udg/(Kg1p*Kiudg))

States:

NameDescription
ppi[diphosphate(4-)]
glx[alpha-D-glucose]
trh[alpha,alpha-trehalose]
pho[hydrogenphosphate]
glc[alpha-D-glucose]
h2o[water]
h[proton]
udp[UDP]
atp[ATP]
utp[UTP(4-)]
g6p[alpha-D-glucose 6-phosphate]
adp[ADP]
t6p[alpha,alpha-trehalose 6-phosphate]
f6p[beta-D-fructofuranose 6-phosphate]
udg[UDP-D-glucose]
g1p[D-glucopyranose 1-phosphate]

Smallbone2013 - Colon Crypt cycle - Version 0: BIOMD0000000520v0.0.1

Smallbone2013 - Colon Crypt cycle - Version 0This model is described in the article: [A mathematical model of the colon…

Details

Models of the development and early progression of colorectal cancer are based upon understanding the cycle of stem cell turnover, proliferation, differentiation and death. Existing crypt compartmental models feature a linear pathway of cell types, with little regulatory mechanism. Previous work has shown that there are perturbations in the enteroendocrine cell population of macroscopically normal crypts, a compartment not included in existing models. We show that existing models do not adequately recapitulate the dynamics of cell fate pathways in the crypt. We report the progressive development, iterative testing and fitting of a developed compartmental model with additional cell types, and which includes feedback mechanisms and cross-regulatory mechanisms between cell types. The fitting of the model to existing data sets suggests a need to invoke cross-talk between cell types as a feature of colon crypt cycle models. link: http://identifiers.org/pubmed/24354351

Parameters:

NameDescription
d2 = 1.83 per dayReaction: N2 => ; N2, Rate Law: d2*N2
d1 = 0.263 per dayReaction: N1 => ; N1, Rate Law: d1*N1
b1 = 0.547 per day; m1 = 29.2408052354609 cell; c1 = 1.0 per dayReaction: N1 => N1 + N2; N1, Rate Law: (b1+c1*N1/(N1+m1))*N1
a0 = 0.0999999999999998 per dayReaction: N0 => N0; N0, Rate Law: a0*N0
b0 = 0.218 per day; c0 = 1.0 per day; m0 = 2.92408052354609 cellReaction: N0 => N0 + N1; N0, Rate Law: (b0+c0*N0/(N0+m0))*N0
a1 = 0.239254806051979 per dayReaction: N1 => N1; N1, Rate Law: a1*N1
d0 = 0.1 per dayReaction: N0 => ; N0, Rate Law: d0*N0

States:

NameDescription
N1[stem cell]
N0[stem cell]
N2[stem cell]

Smallbone2013 - Colon Crypt cycle - Version 1: BIOMD0000000519v0.0.1

Smallbone2013 - Colon Crypt cycle - Version 1This model is described in the article: [A mathematical model of the colon…

Details

Models of the development and early progression of colorectal cancer are based upon understanding the cycle of stem cell turnover, proliferation, differentiation and death. Existing crypt compartmental models feature a linear pathway of cell types, with little regulatory mechanism. Previous work has shown that there are perturbations in the enteroendocrine cell population of macroscopically normal crypts, a compartment not included in existing models. We show that existing models do not adequately recapitulate the dynamics of cell fate pathways in the crypt. We report the progressive development, iterative testing and fitting of a developed compartmental model with additional cell types, and which includes feedback mechanisms and cross-regulatory mechanisms between cell types. The fitting of the model to existing data sets suggests a need to invoke cross-talk between cell types as a feature of colon crypt cycle models. link: http://identifiers.org/pubmed/24354351

Parameters:

NameDescription
p01 = 0.855699855699856 dimensionless; f0 = NaN cell per_dayReaction: N0 => N0 + N1, Rate Law: p01*f0
d1 = 0.420467092599869 per dayReaction: N1 => ; N1, Rate Law: d1*N1
p12 = 0.827377484810943 dimensionless; f1 = NaN cell per_dayReaction: N1 => N1 + N2, Rate Law: p12*f1
d2 = 1.10138534772246 per dayReaction: N2 => ; N2, Rate Law: d2*N2
d0 = 0.1 per dayReaction: N0 => ; N0, Rate Law: d0*N0
f0 = NaN cell per_day; p00 = NaN dimensionlessReaction: N0 => N0, Rate Law: p00*f0
f1 = NaN cell per_day; p11 = NaN dimensionlessReaction: N1 => N1, Rate Law: p11*f1

States:

NameDescription
N1[stem cell]
N0[stem cell]
N2[stem cell]

Smallbone2013 - Colon Crypt cycle - Version 2: BIOMD0000000518v0.0.1

Smallbone2013 - Colon Crypt cycle - Version 2This model is described in the article: [A mathematical model of the colon…

Details

Models of the development and early progression of colorectal cancer are based upon understanding the cycle of stem cell turnover, proliferation, differentiation and death. Existing crypt compartmental models feature a linear pathway of cell types, with little regulatory mechanism. Previous work has shown that there are perturbations in the enteroendocrine cell population of macroscopically normal crypts, a compartment not included in existing models. We show that existing models do not adequately recapitulate the dynamics of cell fate pathways in the crypt. We report the progressive development, iterative testing and fitting of a developed compartmental model with additional cell types, and which includes feedback mechanisms and cross-regulatory mechanisms between cell types. The fitting of the model to existing data sets suggests a need to invoke cross-talk between cell types as a feature of colon crypt cycle models. link: http://identifiers.org/pubmed/24354351

Parameters:

NameDescription
p01 = 0.815689334807208 dimensionless; f0 = NaN cell per_dayReaction: N0 => N0 + N1, Rate Law: p01*f0
p12 = 0.827377484810943 dimensionless; f1 = NaN cell per_dayReaction: N1 => N1 + N2, Rate Law: p12*f1
f0 = NaN cell per_day; p03 = NaN dimensionlessReaction: N0 => N0 + N3, Rate Law: p03*f0
d2 = 2.20277069544492 per day; K2X = 1.5709821429 cellReaction: N2 => ; N3, N2, N3, Rate Law: d2*N2*K2X/(N3+K2X)
K1X = 1.5709821429 cell; d1 = 0.840934185199738 per dayReaction: N1 => ; N3, N1, N3, Rate Law: d1*N1*K1X/(N3+K1X)
d3 = 0.0379622536021846 per dayReaction: N3 => ; N3, Rate Law: d3*N3
d0 = 0.2 per day; K0X = 1.5709821429 cellReaction: N0 => ; N3, N0, N3, Rate Law: d0*N0*K0X/(N3+K0X)
f1 = NaN cell per_day; p11 = NaN dimensionlessReaction: N1 => N1, Rate Law: p11*f1
f0 = NaN cell per_day; p00 = NaN dimensionlessReaction: N0 => N0, Rate Law: p00*f0

States:

NameDescription
N2[stem cell]
N1[stem cell]
N0[stem cell]
N3N3

Smallbone2013 - Colon Crypt cycle - Version 3: BIOMD0000000517v0.0.1

Smallbone2013 - Colon Crypt cycle - Version 3This model is described in the article: [A mathematical model of the colon…

Details

Models of the development and early progression of colorectal cancer are based upon understanding the cycle of stem cell turnover, proliferation, differentiation and death. Existing crypt compartmental models feature a linear pathway of cell types, with little regulatory mechanism. Previous work has shown that there are perturbations in the enteroendocrine cell population of macroscopically normal crypts, a compartment not included in existing models. We show that existing models do not adequately recapitulate the dynamics of cell fate pathways in the crypt. We report the progressive development, iterative testing and fitting of a developed compartmental model with additional cell types, and which includes feedback mechanisms and cross-regulatory mechanisms between cell types. The fitting of the model to existing data sets suggests a need to invoke cross-talk between cell types as a feature of colon crypt cycle models. link: http://identifiers.org/pubmed/24354351

Parameters:

NameDescription
d2 = 1.888676618 per day; K2X = 2.70405837954268 cellReaction: N2 => ; N3, N2, N3, Rate Law: d2*N2*K2X/(N3+K2X)
f0 = NaN cell per_day; p03 = NaN dimensionlessReaction: N0 => N0 + N3, Rate Law: p03*f0
d3 = 0.1677359306 per dayReaction: N3 => ; N3, Rate Law: d3*N3
d0 = 0.02 per day; K0X = 0.153646265911768 cellReaction: N0 => ; N3, N0, N3, Rate Law: d0*N0*K0X/(N3+K0X)
p12 = 0.8050459589 dimensionless; f1 = NaN cell per_dayReaction: N1 => N1 + N2, Rate Law: p12*f1
K1X = 15.3645644864404 cell; d1 = 0.5480597115 per dayReaction: N1 => ; N3, N1, N3, Rate Law: d1*N1*K1X/(N3+K1X)
p01 = 0.6313780928 dimensionless; f0 = NaN cell per_dayReaction: N0 => N0 + N1, Rate Law: p01*f0
f0 = NaN cell per_day; p00 = NaN dimensionlessReaction: N0 => N0, Rate Law: p00*f0
f1 = NaN cell per_day; p11 = NaN dimensionlessReaction: N1 => N1, Rate Law: p11*f1

States:

NameDescription
N3N3
N1[stem cell]
N0[stem cell]
N2[stem cell]

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 00: MODEL1303260000v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 00This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 01: MODEL1303260001v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 01This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 02: MODEL1303260002v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 02This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 03: MODEL1303260003v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 03This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 04: MODEL1303260004v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 04This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 05: MODEL1303260005v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 05This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 06: MODEL1303260006v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 06This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 07: MODEL1303260007v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 07This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 08: MODEL1303260008v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 08This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 09: MODEL1303260009v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 09This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 10: MODEL1303260010v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 10This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 11: MODEL1303260011v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 11This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 12: MODEL1303260012v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 12This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 13: MODEL1303260013v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 13This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 14: MODEL1303260014v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 14This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 15: MODEL1303260015v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 15This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 16: MODEL1303260016v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 16This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 17: MODEL1303260017v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 17This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 18: MODEL1303260018v0.0.1

Smallbone2013 - Glycolysis in S.cerevisiae - Iteration 18This model is described in the article: [A model of yeast glyc…

Details

We present an experimental and computational pipeline for the generation of kinetic models of metabolism, and demonstrate its application to glycolysis in Saccharomyces cerevisiae. Starting from an approximate mathematical model, we employ a "cycle of knowledge" strategy, identifying the steps with most control over flux. Kinetic parameters of the individual isoenzymes within these steps are measured experimentally under a standardised set of conditions. Experimental strategies are applied to establish a set of in vivo concentrations for isoenzymes and metabolites. The data are integrated into a mathematical model that is used to predict a new set of metabolite concentrations and reevaluate the control properties of the system. This bottom-up modelling study reveals that control over the metabolic network most directly involved in yeast glycolysis is more widely distributed than previously thought. link: http://identifiers.org/pubmed/23831062

Smallbone2013 - Human metabolism global reconstruction (recon 2.1): MODEL1311110000v0.0.1

Smallbone2013 - Human metabolism global reconstruction (recon 2.1)Recon 2.1. This model is described in the article: […

Details

Recon 2 is a highly curated reconstruction of the human metabolic network. Whilst the network is state of the art, it has shortcomings, including the presence of unbalanced reactions involving generic metabolites. By replacing these generic molecules with each of their specific instances, we can ensure full elemental balancing, in turn allowing constraint-based analyses to be performed. The resultant model, called Recon 2.1, is an order of magnitude larger than the original. link: http://arxiv.org/abs/1311.5696

Smallbone2013 - Human metabolism global reconstruction (recon 2.1x): MODEL1311110001v0.0.1

Smallbone2013 - Human metabolism global reconstruction (recon 2.1x)Recon 2.1x. This model is described in the article:…

Details

Recon 2 is a highly curated reconstruction of the human metabolic network. Whilst the network is state of the art, it has shortcomings, including the presence of unbalanced reactions involving generic metabolites. By replacing these generic molecules with each of their specific instances, we can ensure full elemental balancing, in turn allowing constraint-based analyses to be performed. The resultant model, called Recon 2.1, is an order of magnitude larger than the original. link: http://arxiv.org/abs/1311.5696

Smallbone2013 - Metabolic Control Analysis - Example 1: BIOMD0000000454v0.0.1

Smallbone2013 - Metabolic Control Analysis - Example 1Metabolic control analysis (MCA) is a biochemical formalism, defin…

Details

Metabolic control analysis is a biochemical formalism defined by Kacser and Burns in 1973, and given firm mathematical basis by Reder in 1988. The algorithm defined by Reder for calculating the control matrices is still used by software programs today, but is only valid for some biochemical models. We show that, with slight modification, the algorithm may be applied to all models. link: http://arxiv.org/pdf/1305.6449v1.pdf

Parameters:

NameDescription
p1=10.0 dimensionless; e1=1.0 dimensionlessReaction: y1 + x2 => x1 + x3; y1, x2, x1, x3, Rate Law: e1*(p1*y1*x2-x1*x3)/(1+y1+x2+x1+x3+y1*x2+x1*x3)
e3=1.0 dimensionless; p3=50.0 dimensionlessReaction: x1 => y2; x1, y2, Rate Law: e3*(p3*x1-y2)/(1+x1+y2)
e2=1.0 dimensionless; p2=10.0 dimensionlessReaction: y4 + x3 => y5 + x2; y4, x3, y5, x2, Rate Law: e2*(p2*y4*x3-y5*x2)/(1+x3+x2+y4+y5+x3*y4+x2*y5)
p4=10.0 dimensionless; e4=1.0 dimensionlessReaction: x1 => y3; x1, y3, Rate Law: e4*(p4*x1-y3)/(1+x1+y3)

States:

NameDescription
y3y3
x1x1
y4y4
y1y1
x2x2
y2y2
x3x3
y5y5

Smallbone2013 - Metabolic Control Analysis - Example 2: BIOMD0000000455v0.0.1

Smallbone2013 - Metabolic Control Analysis - Example 2Metabolic control analysis (MCA) is a biochemical formalism, defin…

Details

Metabolic control analysis is a biochemical formalism defined by Kacser and Burns in 1973, and given firm mathematical basis by Reder in 1988. The algorithm defined by Reder for calculating the control matrices is still used by software programs today, but is only valid for some biochemical models. We show that, with slight modification, the algorithm may be applied to all models. link: http://arxiv.org/pdf/1305.6449v1.pdf

Parameters:

NameDescription
p1=10.0 dimensionless; e1=1.0 dimensionlessReaction: y1 + x2 => x1 + x3; y1, x2, x1, x3, Rate Law: e1*(p1*y1*x2-x1*x3)/(1+y1+x2+x1+x3+y1*x2+x1*x3)
e3=1.0 dimensionless; p3=50.0 dimensionlessReaction: x1 => y2; x1, y2, Rate Law: e3*(p3*x1-y2)/(1+x1+y2)
p4=10.0 dimensionless; e4=1.0 dimensionlessReaction: x1 => y3; x1, y3, Rate Law: e4*(p4*x1-y3)/(1+x1+y3)
e2=1.0 dimensionless; p2=10.0 dimensionlessReaction: y4 + x3 => y5 + x2; y4, x3, y5, x2, Rate Law: e2*(p2*y4*x3-y5*x2)/(1+x3+x2+y4+y5+x3*y4+x2*y5)
e5=1.0 dimensionless; p5=0.0 dimensionlessReaction: x3 => y6; x3, Rate Law: e5*p5*x3

States:

NameDescription
y3y3
x1x1
y1y1
y4y4
x2x2
y2y2
y6y6
x3x3
y5y5

Smallbone2013 - Metabolic Control Analysis - Example 3: BIOMD0000000456v0.0.1

Smallbone2013 - Metabolic Control Analysis - Example 3Metabolic control analysis (MCA) is a biochemical formalism, defin…

Details

Metabolic control analysis is a biochemical formalism defined by Kacser and Burns in 1973, and given firm mathematical basis by Reder in 1988. The algorithm defined by Reder for calculating the control matrices is still used by software programs today, but is only valid for some biochemical models. We show that, with slight modification, the algorithm may be applied to all models. link: http://arxiv.org/pdf/1305.6449v1.pdf

Parameters:

NameDescription
e6=1.0 dimensionless; p6=1.0 dimensionlessReaction: y7 => x4; y7, Rate Law: e6*p6*y7
p1=10.0 dimensionless; e1=1.0 dimensionlessReaction: y1 + x2 => x1 + x3; y1, x2, x1, x3, Rate Law: e1*(p1*y1*x2-x1*x3)/(1+y1+x2+x1+x3+y1*x2+x1*x3)
e3=1.0 dimensionless; p3=50.0 dimensionlessReaction: x1 => y2; x1, y2, Rate Law: e3*(p3*x1-y2)/(1+x1+y2)
e2=1.0 dimensionless; p2=10.0 dimensionlessReaction: y4 + x3 => y5 + x2; y4, x3, y5, x2, Rate Law: e2*(p2*y4*x3-y5*x2)/(1+x3+x2+y4+y5+x3*y4+x2*y5)
p4=10.0 dimensionless; e4=1.0 dimensionlessReaction: x1 => y3; x1, y3, Rate Law: e4*(p4*x1-y3)/(1+x1+y3)
p7=1.0 dimensionless; e7=1.0 dimensionlessReaction: x4 => y8, Rate Law: e7*p7

States:

NameDescription
y3y3
x4x4
x2x2
x3x3
y8y8
x1x1
y4y4
y1y1
y7y7
y2y2
y5y5

Smallbone2013 - Serine biosynthesis: BIOMD0000000458v0.0.1

Smallbone2013 - Serine biosynthesisKinetic modelling of metabolic pathways in application to Serine biosynthesis. This…

Details

In this chapter, we describe the steps needed to create a kinetic model of a metabolic pathway using kinetic data from both experimental measurements and literature review. Our methodology is presented by using the example of serine biosynthesis in E. coli. link: http://identifiers.org/pubmed/23417802

Parameters:

NameDescription
KAphp=0.0032 mM; KiAser=0.0038 mM; kcatA=0.55 per s; KAp3g=1.2 mMReaction: p3g => php; serA, ser, serA, p3g, php, ser, Rate Law: cell*serA*kcatA*p3g/KAp3g/(1+p3g/KAp3g+php/KAphp)/(1+ser/KiAser)
kcatC=1.75 per s; KCpser=0.0017 mM; KCphp=0.0015 mMReaction: php => pser; serC, serC, php, pser, Rate Law: cell*serC*kcatC*php/KCphp/(1+php/KCphp+pser/KCpser)
KBpser=0.0015 mM; KBser=0.15 mM; kcatB=1.43 per sReaction: pser => ser; serB, serB, pser, ser, Rate Law: cell*serB*kcatB*pser/KBpser/(1+pser/KBpser+ser/KBser)

States:

NameDescription
p3g[3-phosphonato-D-glycerate(3-)]
ser[L-serine]
php[3-phosphonatooxypyruvate(3-)]
pser[O-phosphonato-L-serine(2-)]

Smith1980 - Hypothalamic Regulation: BIOMD0000000831v0.0.1

This a model from the article: Hypothalamic regulation of pituitary secretion of luteinizing hormone.II. Feedback cont…

Details

A general mathematical model describing the biochemical interactions of the hormones luteinizing hormone releasing hormone (LHRH), luteinizing hormone (LH) and testosterone (T) in the male is presented. The model structure consists of a negative feedback system of three ordinary differential equations, in which the quali]ative behavior is either a stable constant equilibrium solution or oscillatory solutions. A specific realization of the model is used to describe the experimental observations of pulsatile hormone release, its experimental suppression, the onset of puberty, the effects of castration, and several other qualitative and quantitative results. This model is presented as a first step in understanding the physi- cochemical interactions of the hypothalamic pituitary gonadal axis. link: http://identifiers.org/pubmed/6986927

Parameters:

NameDescription
g1 = 10.0 1/hReaction: => L; R, Rate Law: Compartment*g1*R
h = 12.0 1/h; H = 1.0 1; c = 100.0 ng/(l*h)Reaction: => R; T, Rate Law: Compartment*(c-h*T)*(1-H)
b1 = 1.29 1/hReaction: R =>, Rate Law: Compartment*b1*R
g2 = 0.7 1/hReaction: => T; L, Rate Law: Compartment*g2*L
b3 = 1.39 1/hReaction: T =>, Rate Law: Compartment*b3*T
b2 = 0.97 1/hReaction: L =>, Rate Law: Compartment*b2*L

States:

NameDescription
T[Thyroxine 5-deiodinase]
L[Luteinizing hormone]
R[Luteinizing hormone receptor]

Smith2004_CVS_human: MODEL1006230000v0.0.1

This a model from the article: Minimal haemodynamic system model including ventricular interaction and valve dynamics.…

Details

Characterising circulatory dysfunction and choosing a suitable treatment is often difficult and time consuming, and can result in a deterioration in patient condition, or unsuitable therapy choices. A stable minimal model of the human cardiovascular system (CVS) is developed with the ultimate specific aim of assisting medical staff for rapid, on site modelling to assist in diagnosis and treatment. Models found in the literature simulate specific areas of the CVS with limited direct usefulness to medical staff. Others model the full CVS as a closed loop system, but they were found to be very complex, difficult to solve, or unstable. This paper develops a model that uses a minimal number of governing equations with the primary goal of accurately capturing trends in the CVS dynamics in a simple, easily solved, robust model. The model is shown to have long term stability and consistency with non-specific initial conditions as a result. An "open on pressure close on flow" valve law is created to capture the effects of inertia and the resulting dynamics of blood flow through the cardiac valves. An accurate, stable solution is performed using a method that varies the number of states in the model depending on the specific phase of the cardiac cycle, better matching the real physiological conditions. Examples of results include a 9% drop in cardiac output when increasing the thoracic pressure from -4 to 0 mmHg, and an increase in blood pressure from 120/80 to 165/130 mmHg when the systemic resistance is doubled. These results show that the model adequately provides appropriate magnitudes and trends that are in agreement with existing data for a variety of physiologically verified test cases simulating human CVS function. link: http://identifiers.org/pubmed/15036180

Smith2009 - RGS mediated GTP hydrolysis: BIOMD0000000439v0.0.1

Smith2009 - RGS mediated GTP hydrolysisThis model is described in the article: [Dual positive and negative regulation o…

Details

G protein-coupled receptors (GPCRs) regulate a variety of intracellular pathways through their ability to promote the binding of GTP to heterotrimeric G proteins. Regulator of G protein signaling (RGS) proteins increases the intrinsic GTPase activity of Galpha-subunits and are widely regarded as negative regulators of G protein signaling. Using yeast we demonstrate that GTP hydrolysis is not only required for desensitization, but is essential for achieving a high maximal (saturated level) response. Thus RGS-mediated GTP hydrolysis acts as both a negative (low stimulation) and positive (high stimulation) regulator of signaling. To account for this we generated a new kinetic model of the G protein cycle where Galpha(GTP) enters an inactive GTP-bound state following effector activation. Furthermore, in vivo and in silico experimentation demonstrates that maximum signaling output first increases and then decreases with RGS concentration. This unimodal, non-monotone dependence on RGS concentration is novel. Analysis of the kinetic model has revealed a dynamic network motif that shows precisely how inclusion of the inactive GTP-bound state for the Galpha produces this unimodal relationship. link: http://identifiers.org/pubmed/19285552

Parameters:

NameDescription
k1=0.0025 1/(nM*hr)Reaction: R + L => RL; R, L, Rate Law: compartment*R*L*k1
k8=2.5 1/hrReaction: RGSGaGTP => GaGDPP + RGS; RGSGaGTP, Rate Law: compartment*RGSGaGTP*k8
k7=500.0 1/(nM*hr)Reaction: GaGTP + RGS => RGSGaGTP; GaGTP, RGS, Rate Law: compartment*GaGTP*RGS*k7
k9=0.005 1/hrReaction: GaGTP => GaGDPP; GaGTP, Rate Law: compartment*GaGTP*k9
k16=1000.0 1/(nM*hr)Reaction: GaGDP + Gbg => Gabg; GaGDP, Gbg, Rate Law: compartment*GaGDP*Gbg*k16
k15=1000.0 1/hrReaction: GaGDPP => GaGDP + P; GaGDPP, Rate Law: compartment*GaGDPP*k15
k4=0.005 1/(nM*hr)Reaction: RGabg + L => RGabgL; RGabg, L, Rate Law: compartment*RGabg*L*k4
k11=1.0 1/hrReaction: GaGTPEffector => inertGaGTP + Effector; GaGTPEffector, Rate Law: compartment*GaGTPEffector*k11
k5=50.0 1/hrReaction: RGabgL => RL + GaGTP + Gbg; RGabgL, Rate Law: compartment*RGabgL*k5
k17=10.0 1/hrReaction: P => ; P, Rate Law: compartment*P*k17
k13=0.3 1/hrReaction: RGSinertGaGTP => GaGDPP + RGS; RGSinertGaGTP, Rate Law: compartment*RGSinertGaGTP*k13
k6=0.2 1/hrReaction: Gabg => GaGTP + Gbg; Gabg, Rate Law: compartment*Gabg*k6
k12=50.0 1/(nM*hr)Reaction: inertGaGTP + RGS => RGSinertGaGTP; inertGaGTP, RGS, Rate Law: compartment*inertGaGTP*RGS*k12
k14=0.005 1/hrReaction: inertGaGTP => GaGDPP; inertGaGTP, Rate Law: compartment*inertGaGTP*k14
k3=0.02 1/(nM*hr)Reaction: RL + Gabg => RGabgL; RL, Gabg, Rate Law: compartment*RL*Gabg*k3
k2=0.005 1/(nM*hr)Reaction: R + Gabg => RGabg; R, Gabg, Rate Law: compartment*R*Gabg*k2
k10=10.0 1/(nM*hr)Reaction: Effector + GaGTP => GaGTPEffector; Effector, GaGTP, Rate Law: compartment*Effector*GaGTP*k10
ka = 1.5 1/hrReaction: => z1; GaGTPEffector, GaGTPEffector, Rate Law: compartment*GaGTPEffector*ka

States:

NameDescription
RGabg[IPR000276; heterotrimeric G-protein complex]
GaGTP[GTP; Guanine nucleotide-binding protein G(t) subunit alpha-1]
GaGDPP[GDP; Guanine nucleotide-binding protein G(t) subunit alpha-1]
inertGaGTP[GTP; Guanine nucleotide-binding protein G(t) subunit alpha-1; inactive]
RGS[IPR000342]
P[phosphate(3-)]
z1[SBO:0000347]
RL[receptor complex]
LL
Gabg[heterotrimeric G-protein complex]
Gbg[Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Guanine nucleotide-binding protein G(T) subunit gamma-T1]
z3[SBO:0000347]
GaGDP[GDP; Guanine nucleotide-binding protein G(t) subunit alpha-1]
GaGTPEffector[GTP; Guanine nucleotide-binding protein G(t) subunit alpha-1; SBO:0000459]
RGSGaGTP[GTP; Guanine nucleotide-binding protein G(t) subunit alpha-1; IPR000342]
RGSinertGaGTP[GTP; Guanine nucleotide-binding protein G(t) subunit alpha-1; IPR000342; inactive]
Effector[SBO:0000459]
RGabgL[IPR000276; heterotrimeric G-protein complex; SBO:0000280]
z2[SBO:0000347]
R[IPR000276]

Smith2010_Foxo_PTMs_AgeingRelatedSignallingPathway_C: MODEL1112260002v0.0.1

This a model from the article: Modelling the response of FOXO transcription factors to multiple post-translational mod…

Details

FOXO transcription factors are an important, conserved family of regulators of cellular processes including metabolism, cell-cycle progression, apoptosis and stress resistance. They are required for the efficacy of several of the genetic interventions that modulate lifespan. FOXO activity is regulated by multiple post-translational modifications (PTMs) that affect its subcellular localization, half-life, DNA binding and transcriptional activity. Here, we show how a mathematical modelling approach can be used to simulate the effects, singly and in combination, of these PTMs. Our model is implemented using the Systems Biology Markup Language (SBML), generated by an ancillary program and simulated in a stochastic framework. The use of the ancillary program to generate the SBML is necessary because the possibility that many regulatory PTMs may be added, each independently of the others, means that a large number of chemically distinct forms of the FOXO molecule must be taken into account, and the program is used to generate them. Although the model does not yet include detailed representations of events upstream and downstream of FOXO, we show how it can qualitatively, and in some cases quantitatively, reproduce the known effects of certain treatments that induce various single and multiple PTMs, and allows for a complex spatiotemporal interplay of effects due to the activation of multiple PTM-inducing treatments. Thus, it provides an important framework to integrate current knowledge about the behaviour of FOXO. The approach should be generally applicable to other proteins experiencing multiple regulations. link: http://identifiers.org/pubmed/20567500

Smith2011 - Three Stage Innate Immune Response to a Pneumococcal Lung Infection: BIOMD0000000924v0.0.1

Pneumococcal pneumonia is a leading cause of death and a major source of human morbidity. The initial immune response pl…

Details

Pneumococcal pneumonia is a leading cause of death and a major source of human morbidity. The initial immune response plays a central role in determining the course and outcome of pneumococcal disease. We combine bacterial titer measurements from mice infected with Streptococcus pneumoniae with mathematical modeling to investigate the coordination of immune responses and the effects of initial inoculum on outcome. To evaluate the contributions of individual components, we systematically build a mathematical model from three subsystems that describe the succession of defensive cells in the lung: resident alveolar macrophages, neutrophils and monocyte-derived macrophages. The alveolar macrophage response, which can be modeled by a single differential equation, can by itself rapidly clear small initial numbers of pneumococci. Extending the model to include the neutrophil response required additional equations for recruitment cytokines and host cell status and damage. With these dynamics, two outcomes can be predicted: bacterial clearance or sustained bacterial growth. Finally, a model including monocyte-derived macrophage recruitment by neutrophils suggests that sustained bacterial growth is possible even in their presence. Our model quantifies the contributions of cytotoxicity and immune-mediated damage in pneumococcal pathogenesis. link: http://identifiers.org/pubmed/21300073

Parameters:

NameDescription
eta = 1.33; N_max = 180000.0Reaction: => Neutrophils__N; proinflammatory_cytokine__C, Rate Law: compartment*eta*proinflammatory_cytokine__C*(1-Neutrophils__N/N_max)
theta_M = 4.2E-8; d = 0.001; k_n = 1.4E-5; M_Astar = 1000000.0; alpha = 0.021; v = 0.029; kappa = 0.042Reaction: => proinflammatory_cytokine__C; Epithelial_cells_with_bacteria_attached__Ea, Neutrophils__N, Pneumococci___P, Rate Law: compartment*(alpha*Epithelial_cells_with_bacteria_attached__Ea/(1+k_n*Neutrophils__N)+v*theta_M*Pneumococci___P*M_Astar/(d+kappa+theta_M*Pneumococci___P*(1+k_n*Neutrophils__N)))
d_E = 0.167Reaction: Epithelial_cells_with_bacteria_attached__Ea =>, Rate Law: compartment*d_E*Epithelial_cells_with_bacteria_attached__Ea
d_C = 0.83Reaction: proinflammatory_cytokine__C =>, Rate Law: compartment*d_C*proinflammatory_cytokine__C
d_NP = 1.76E-7; d_E = 0.167; rho1 = 0.15; d_N = 0.063; rho2 = 0.001; rho3 = 1.0E-5Reaction: => Debris__D; Neutrophils__N, Pneumococci___P, Epithelial_cells_with_bacteria_attached__Ea, Rate Law: compartment*(rho1*d_NP*Neutrophils__N*Pneumococci___P+rho2*d_N*Neutrophils__N+rho3*d_E*Epithelial_cells_with_bacteria_attached__Ea)
d_D = 1.4E-7; M_Astar = 1000000.0Reaction: Debris__D =>, Rate Law: compartment*d_D*Debris__D*M_Astar
f_P_M_A = 0.00249376558603491; gamma_N = 1.0E-5; k_d = 5.0E-9; M_Astar = 1000000.0; gamma_M_A = 5.6E-6Reaction: Pneumococci___P => ; Debris__D, Neutrophils__N, Rate Law: compartment*(gamma_M_A*f_P_M_A/(1+k_d*Debris__D*M_Astar)*M_Astar*Pneumococci___P+gamma_N*Neutrophils__N*Pneumococci___P)
K_P = 3.41765197726012E8; r = 1.13Reaction: => Pneumococci___P, Rate Law: compartment*r*Pneumococci___P*(1-Pneumococci___P/K_P)
omega = 2.1E-8Reaction: Susceptible_epithelial_cells__EU => ; Pneumococci___P, Rate Law: compartment*omega*Pneumococci___P*Susceptible_epithelial_cells__EU
d_NP = 1.76E-7; d_N = 0.063Reaction: Neutrophils__N => ; Pneumococci___P, Rate Law: compartment*(d_NP*Neutrophils__N*Pneumococci___P+d_N*Neutrophils__N)

States:

NameDescription
Pneumococci P[C76384]
Epithelial cells with bacteria attached Ea[infected cell]
Susceptible epithelial cells EU[0006083]
Neutrophils N[0010527]
proinflammatory cytokine C[Cytokine]
Debris D[C120869]

Smith2011_HumanHeartMitochondrian_MetabolicModel: MODEL1106160000v0.0.1

This model is from the article: A metabolic model of the mitochondrion and its use in modelling diseases of the tricar…

Details

Mitochondria are a vital component of eukaryotic cells and their dysfunction is implicated in a large number of metabolic, degenerative and age-related human diseases. The mechanism or these disorders can be difficult to elucidate due to the inherent complexity of mitochondrial metabolism. To understand how mitochondrial metabolic dysfunction contributes to these diseases, a metabolic model of a human heart mitochondrion was created.A new model of mitochondrial metabolism was built on the principle of metabolite availability using MitoMiner, a mitochondrial proteomics database, to evaluate the subcellular localisation of reactions that have evidence for mitochondrial localisation. Extensive curation and manual refinement was used to create a model called iAS253, containing 253 reactions, 245 metabolites and 89 transport steps across the inner mitochondrial membrane. To demonstrate the predictive abilities of the model, flux balance analysis was used to calculate metabolite fluxes under normal conditions and to simulate three metabolic disorders that affect the TCA cycle: fumarase deficiency, succinate dehydrogenase deficiency and α-ketoglutarate dehydrogenase deficiency.The results of simulations using the new model corresponded closely with phenotypic data under normal conditions and provided insight into the complicated and unintuitive phenotypes of the three disorders, including the effect of interventions that may be of therapeutic benefit, such as low glucose diets or amino acid supplements. The model offers the ability to investigate other mitochondrial disorders and can provide the framework for the integration of experimental data in future studies. link: http://identifiers.org/pubmed/21714867

Smith2013 - Regulation of Insulin Signalling by Oxidative Stress: BIOMD0000000474v0.0.1

Smith2013 - Regulation of Insulin Signalling by Oxidative StressThe model describes insulin signalling (in rodent adipoc…

Details

Existing models of insulin signalling focus on short term dynamics, rather than the longer term dynamics necessary to understand many physiologically relevant behaviours. We have developed a model of insulin signalling in rodent adipocytes that includes both transcriptional feedback through the Forkhead box type O (FOXO) transcription factor, and interaction with oxidative stress, in addition to the core pathway. In the model Reactive Oxygen Species are both generated endogenously and can be applied externally. They regulate signalling though inhibition of phosphatases and induction of the activity of Stress Activated Protein Kinases, which themselves modulate feedbacks to insulin signalling and FOXO.Insulin and oxidative stress combined produce a lower degree of activation of insulin signalling than insulin alone. Fasting (nutrient withdrawal) and weak oxidative stress upregulate antioxidant defences while stronger oxidative stress leads to a short term activation of insulin signalling but if prolonged can have other effects including degradation of the insulin receptor substrate (IRS1) and FOXO. At high insulin the protective effect of moderate oxidative stress may disappear.Our model is consistent with a wide range of experimental data, some of which is difficult to explain. Oxidative stress can have effects that are both up- and down-regulatory on insulin signalling. Our model therefore shows the complexity of the interaction between the two pathways and highlights the need for such integrated computational models to give insight into the dysregulation of insulin signalling along with more data at the individual level.A complete SBML model file can be downloaded from BIOMODELS (https://www.ebi.ac.uk/biomodels-main) with unique identifier MODEL1212210000.Other files and scripts are available as additional files with this journal article and can be downloaded from https://github.com/graham1034/Smith2012insulinsignalling. link: http://identifiers.org/pubmed/23705851

Parameters:

NameDescription
k1 = 2.0E-5Reaction: Ins + InR => Ins_InR; InR, Ins, Rate Law: k1*Ins*extracellular*InR*cellsurface
kminusr16a = 1.0E-6Reaction: AS160_P => AS160; PP2A, AS160_P, PP2A, Rate Law: cytoplasm*kminusr16a*PP2A*cytoplasm*AS160_P*cytoplasm/cytoplasm
kminus9 = 0.0014; kminus9_basal = 2.7Reaction: PI345P3 => PIP2; PTEN, PI345P3, PTEN, Rate Law: cytoplasm*(kminus9_basal+kminus9*PTEN*cytoplasm)*PI345P3*cytoplasm/cytoplasm
ktr=0.125Reaction: nucleus_Foxo1_Pa1_Pd0_Pe1_pUb0 => dnabound_Foxo1_Pa1_Pd0_Pe1_pUb0; nucleus_Foxo1_Pa1_Pd0_Pe1_pUb0, Rate Law: nucleus_Foxo1_Pa1_Pd0_Pe1_pUb0*nucleus*ktr
kdeg=1.0E-4Reaction: cytoplasm_Foxo1_Pa1_Pd1_Pe1_pUb1 => degr_Foxo1; Proteasome, Proteasome, cytoplasm_Foxo1_Pa1_Pd1_Pe1_pUb1, Rate Law: cytoplasm*cytoplasm_Foxo1_Pa1_Pd1_Pe1_pUb1*cytoplasm*Proteasome*cytoplasm*kdeg/cytoplasm
kkin=3.0E-4Reaction: cytoplasm_Foxo1_Pa0_Pd1_Pe1_pUb1 => cytoplasm_Foxo1_Pa1_Pd1_Pe1_pUb1; SGK, SGK, cytoplasm_Foxo1_Pa0_Pd1_Pe1_pUb1, Rate Law: cytoplasm*cytoplasm_Foxo1_Pa0_Pd1_Pe1_pUb1*cytoplasm*SGK*cytoplasm*kkin/cytoplasm
k36f = 180.0Reaction: Mt => Mt + ROS; Mt, Rate Law: cytoplasm*k36f*Mt*cytoplasm/cytoplasm
kub=6.6E-5Reaction: dnabound_Foxo1_Pa1_Pd1_Pe0_pUb0 => dnabound_Foxo1_Pa1_Pd1_Pe0_pUb1; SCF, SCF, dnabound_Foxo1_Pa1_Pd1_Pe0_pUb0, Rate Law: dnabound*dnabound_Foxo1_Pa1_Pd1_Pe0_pUb0*dnabound*SCF*cytoplasm*kub/dnabound
ktr=0.055Reaction: nucleus_Foxo1_Pa0_Pd1_Pe1_pUb1 => cytoplasm_Foxo1_Pa0_Pd1_Pe1_pUb1; nucleus_Foxo1_Pa0_Pd1_Pe1_pUb1, Rate Law: nucleus_Foxo1_Pa0_Pd1_Pe1_pUb1*nucleus*ktr
kub=1.0E-6Reaction: dnabound_Foxo1_Pa0_Pd0_Pe1_pUb0 => dnabound_Foxo1_Pa0_Pd0_Pe1_pUb1; SCF, SCF, dnabound_Foxo1_Pa0_Pd0_Pe1_pUb0, Rate Law: dnabound*dnabound_Foxo1_Pa0_Pd0_Pe1_pUb0*dnabound*SCF*cytoplasm*kub/dnabound
k35f = 450.0Reaction: NOX => ROS + NOX; NOX, Rate Law: cytoplasm*k35f*NOX*cytoplasm/cytoplasm
kub=3.0E-6Reaction: nucleus_Foxo1_Pa1_Pd0_Pe0_pUb0 => nucleus_Foxo1_Pa1_Pd0_Pe0_pUb1; SCF, SCF, nucleus_Foxo1_Pa1_Pd0_Pe0_pUb0, Rate Law: nucleus*nucleus_Foxo1_Pa1_Pd0_Pe0_pUb0*nucleus*SCF*cytoplasm*kub/nucleus
kpdeg=0.0044Reaction: cytoplasm_InR => null; cytoplasm_InR, Rate Law: cytoplasm*cytoplasm_InR*cytoplasm*kpdeg/cytoplasm
kminus12 = 1.25E-6Reaction: PKC_P => PKC; PP2A, PKC_P, PP2A, Rate Law: cytoplasm*kminus12*PP2A*cytoplasm*PKC_P*cytoplasm/cytoplasm
kminus13 = 0.167Reaction: cellsurface_GLUT4 => cytoplasm_GLUT4; cellsurface_GLUT4, Rate Law: kminus13*cellsurface_GLUT4*cellsurface
k_ros_perm = 4.81Reaction: ROS => extracellular_ROS; ROS, Rate Law: k_ros_perm*extracellular/cytoplasm*ROS*cytoplasm
ktr=0.55Reaction: nucleus_Foxo1_Pa0_Pd1_Pe0_pUb0 => cytoplasm_Foxo1_Pa0_Pd1_Pe0_pUb0; nucleus_Foxo1_Pa0_Pd1_Pe0_pUb0, Rate Law: nucleus_Foxo1_Pa0_Pd1_Pe0_pUb0*nucleus*ktr
ktr=0.0909090909091Reaction: cytoplasm_Foxo1_Pa0_Pd1_Pe0_pUb0 => nucleus_Foxo1_Pa0_Pd1_Pe0_pUb0; cytoplasm_Foxo1_Pa0_Pd1_Pe0_pUb0, Rate Law: cytoplasm_Foxo1_Pa0_Pd1_Pe0_pUb0*cytoplasm*ktr
k30r = 0.005Reaction: PTP1B_ox + GSH => PTP1B + GSH; GSH, PTP1B_ox, Rate Law: cytoplasm*k30r*PTP1B_ox*cytoplasm*GSH*cytoplasm/cytoplasm
k13 = 7.5E-6; k13_basal = 0.015Reaction: cytoplasm_GLUT4 => cellsurface_GLUT4; AS160_P, AS160_P, cytoplasm_GLUT4, Rate Law: (k13_basal+k13*AS160_P*cytoplasm)*cytoplasm_GLUT4*cytoplasm
k32f = 6.0E-4Reaction: DUSP + ROS => DUSP_ox + ROS; DUSP, ROS, Rate Law: cytoplasm*k32f*DUSP*cytoplasm*ROS*cytoplasm/cytoplasm
k31r = 0.002Reaction: PTEN_ox + GSH => PTEN + GSH; GSH, PTEN_ox, Rate Law: cytoplasm*k31r*PTEN_ox*cytoplasm*GSH*cytoplasm/cytoplasm
by_jnk_phos_factor = 2.0; kkin=5.0E-5Reaction: dnabound_Foxo1_Pa0_Pd0_Pe0_pUb1 => dnabound_Foxo1_Pa0_Pd0_Pe1_pUb1; JNK_P, JNK_P, dnabound_Foxo1_Pa0_Pd0_Pe0_pUb1, Rate Law: dnabound*dnabound_Foxo1_Pa0_Pd0_Pe0_pUb1*dnabound*JNK_P*cytoplasm*by_jnk_phos_factor*kkin/dnabound
kminus11 = 1.1878E-6Reaction: Akt_P2 => Akt; PP2A, Akt_P2, PP2A, Rate Law: cytoplasm*kminus11*PP2A*cytoplasm*Akt_P2*cytoplasm/cytoplasm
k9 = 0.0055; k9_basal = 0.13145Reaction: PIP2 => PI345P3; IRS1_TyrP_PI3K, IRS1_TyrP_PI3K, PIP2, Rate Law: cytoplasm*(k9_basal+k9*IRS1_TyrP_PI3K*cytoplasm)*PIP2*cytoplasm/cytoplasm
by_ikk_phos_factor = 3.0; kkin=5.0E-5Reaction: dnabound_Foxo1_Pa0_Pd0_Pe1_pUb1 => dnabound_Foxo1_Pa0_Pd1_Pe1_pUb1; IKK_P, IKK_P, dnabound_Foxo1_Pa0_Pd0_Pe1_pUb1, Rate Law: dnabound*dnabound_Foxo1_Pa0_Pd0_Pe1_pUb1*dnabound*IKK_P*cytoplasm*by_ikk_phos_factor*kkin/dnabound
ktr=0.181818181818Reaction: cytoplasm_Foxo1_Pa0_Pd0_Pe0_pUb1 => nucleus_Foxo1_Pa0_Pd0_Pe0_pUb1; cytoplasm_Foxo1_Pa0_Pd0_Pe0_pUb1, Rate Law: cytoplasm_Foxo1_Pa0_Pd0_Pe0_pUb1*cytoplasm*ktr
pip3_basal = 200.0; k12 = 3.5E-5Reaction: PKC => PKC_P; PI345P3, PI345P3, PKC, Rate Law: cytoplasm*k12*PKC*cytoplasm*piecewise(PI345P3*cytoplasm-pip3_basal, (PI345P3*cytoplasm) > pip3_basal, 0)/cytoplasm
k31f = 2.7E-4Reaction: PTEN + ROS => PTEN_ox + ROS; PTEN, ROS, Rate Law: cytoplasm*k31f*PTEN*cytoplasm*ROS*cytoplasm/cytoplasm
pip3_basal = 200.0; k11 = 2.5E-5Reaction: Akt => Akt_P2; PI345P3, Akt, PI345P3, Rate Law: cytoplasm*k11*Akt*cytoplasm*piecewise(PI345P3*cytoplasm-pip3_basal, (PI345P3*cytoplasm) > pip3_basal, 0)/cytoplasm
ktranscr=0.24Reaction: null => nucleus_RNA_InR; dnabound_Foxo1_Pa1_Pd0_Pe0_pUb0, dnabound_Foxo1_Pa1_Pd0_Pe0_pUb0, Rate Law: dnabound_Foxo1_Pa1_Pd0_Pe0_pUb0*dnabound*ktranscr
k_irs1_basal_degr = 0.001Reaction: IRS1 => NULL; IRS1, Rate Law: cytoplasm*IRS1*cytoplasm*k_irs1_basal_degr/cytoplasm
kdephos=1.0E-6Reaction: dnabound_Foxo1_Pa1_Pd0_Pe1_pUb1 => dnabound_Foxo1_Pa0_Pd0_Pe1_pUb1; PP2A, PP2A, dnabound_Foxo1_Pa1_Pd0_Pe1_pUb1, Rate Law: dnabound*dnabound_Foxo1_Pa1_Pd0_Pe1_pUb1*dnabound*PP2A*cytoplasm*kdephos/dnabound

States:

NameDescription
dnabound Foxo1 Pa1 Pd1 Pe1 pUb1[double-stranded DNA; Forkhead box protein O1]
dnabound Foxo1 Pa1 Pd1 Pe1 pUb0[double-stranded DNA; Forkhead box protein O1]
PIP2[phosphatidylinositol bisphosphate]
PKC P[phosphorylated; Atypical protein kinase C]
PTEN[Phosphatase and tensin homologPhosphatase and tensin homolog, isoform CRA_aProtein tyrosine phosphatase and tensin homolog/mutated in multiple advanced cancers proteinProtein tyrosine phosphatase and tensin-like protein]
cytoplasm Foxo1 Pa0 Pd1 Pe0 pUb0[Forkhead box protein O1]
Akt[RAC-alpha serine/threonine-protein kinase]
cytoplasm Foxo1 Pa1 Pd0 Pe1 pUb1[Forkhead box protein O1]
cytoplasm Foxo1 Pa0 Pd0 Pe0 pUb1[Forkhead box protein O1]
nucleus Foxo1 Pa1 Pd1 Pe1 pUb0[Forkhead box protein O1]
nucleus Foxo1 Pa1 Pd0 Pe0 pUb0[Forkhead box protein O1]
dnabound Foxo1 Pa1 Pd0 Pe1 pUb0[double-stranded DNA; Forkhead box protein O1]
PTP1B oxPTP1B_ox
nucleus Foxo1 Pa1 Pd1 Pe1 pUb1[Forkhead box protein O1]
cytoplasm InR[Insulin receptor]
cellsurface GLUT4[Solute carrier family 2, facilitated glucose transporter member 4]
dnabound Foxo1 Pa0 Pd0 Pe1 pUb1[double-stranded DNA; Forkhead box protein O1]
ROS[reactive oxygen species]
cytoplasm Foxo1 Pa0 Pd1 Pe1 pUb1[Forkhead box protein O1]
NULLNULL
dnabound Foxo1 Pa0 Pd0 Pe0 pUb0[double-stranded DNA; Forkhead box protein O1]
nucleus Foxo1 Pa1 Pd0 Pe1 pUb0[Forkhead box protein O1]
PTEN ox[oxidized; Phosphatase and tensin homologPhosphatase and tensin homolog, isoform CRA_aProtein tyrosine phosphatase and tensin homolog/mutated in multiple advanced cancers proteinProtein tyrosine phosphatase and tensin-like protein]
nucleus RNA InR[ribonucleic acid; Insulin receptor]
dnabound Foxo1 Pa1 Pd1 Pe0 pUb0[double-stranded DNA; Forkhead box protein O1]
Akt P2[phosphorylated; RAC-alpha serine/threonine-protein kinase]
AS160[TBC1 domain family member 4]
cytoplasm GLUT4[Solute carrier family 2, facilitated glucose transporter member 4]
Ins[Insulin-1]
dnabound Foxo1 Pa1 Pd0 Pe0 pUb0[double-stranded DNA; Forkhead box protein O1]
cytoplasm Foxo1 Pa1 Pd1 Pe0 pUb1[Forkhead box protein O1]
cytoplasm Foxo1 Pa1 Pd1 Pe1 pUb1[Forkhead box protein O1]
PKC[Atypical protein kinase C]
extracellular ROS[extracellular region; reactive oxygen species]

Smith2016-Combination therapy to prevent bacterial coinfection during influenza.: MODEL1812040005v0.0.1

Secondary bacterial infections (SBIs) exacerbate influenza-associated disease and mortality. Antimicrobial agents can re…

Details

Secondary bacterial infections (SBIs) exacerbate influenza-associated disease and mortality. Antimicrobial agents can reduce the severity of SBIs, but many have limited efficacy or cause adverse effects. Thus, new treatment strategies are needed. Kinetic models describing the infection process can help determine optimal therapeutic targets, the time scale on which a drug will be most effective, and how infection dynamics will change under therapy. To understand how different therapies perturb the dynamics of influenza infection and bacterial coinfection and to quantify the benefit of increasing a drug's efficacy or targeting a different infection process, I analyzed data from mice treated with an antiviral, an antibiotic, or an immune modulatory agent with kinetic models. The results suggest that antivirals targeting the viral life cycle are most efficacious in the first 2 days of infection, potentially because of an improved immune response, and that increasing the clearance of infected cells is important for treatment later in the infection. For a coinfection, immunotherapy could control low bacterial loads with as little as 20 % efficacy, but more effective drugs would be necessary for high bacterial loads. Antibiotics targeting bacterial replication and administered 10 h after infection would require 100 % efficacy, which could be reduced to 40 % with prophylaxis. Combining immunotherapy with antibiotics could substantially increase treatment success. Taken together, the results suggest when and why some therapies fail, determine the efficacy needed for successful treatment, identify potential immune effects, and show how the regulation of underlying mechanisms can be used to design new therapeutic strategies. link: http://identifiers.org/pubmed/27679506

SmithAE2002_RanTransport: BIOMD0000000164v0.0.1

The model reproduces the compartmental model for Ran transport as depicted in Fig 3 of the paper. Model reproduced using…

Details

The separate components of nucleocytoplasmic transport have been well characterized, including the key regulatory role of Ran, a guanine nucleotide triphosphatase. However, the overall system behavior in intact cells is difficult to analyze because the dynamics of these components are interdependent. We used a combined experimental and computational approach to study Ran transport in vivo. The resulting model provides the first quantitative picture of Ran flux between the nuclear and cytoplasmic compartments in eukaryotic cells. The model predicts that the Ran exchange factor RCC1, and not the flux capacity of the nuclear pore complex (NPC), is the crucial regulator of steady-state flux across the NPC. Moreover, it provides the first estimate of the total in vivo flux (520 molecules per NPC per second and predicts that the transport system is robust. link: http://identifiers.org/pubmed/11799242

Parameters:

NameDescription
RCC1Kcat=8.5 s^(-1); RCC1Km=1.1 0.001*dimensionless*m^(-3)*molReaction: RanGDP_Nucleus => RanGTP_Nucleus; RCC1_Nucleus, NTF2_RanGDP_Nucleus, Rate Law: 0.75*RCC1Kcat*RCC1_Nucleus*RanGDP_Nucleus*1/(RCC1Km+RanGDP_Nucleus+NTF2_RanGDP_Nucleus)*Nucleus
I=0.0 dimensionless*A*m^(-2); NTF2_RanGDP_Kperm=3.73333 1E-6*dimensionless*m*s^(-1)Reaction: FNTF2_RanGDP_Cytosol => FNTF2_RanGDP_Nucleus, Rate Law: NTF2_RanGDP_Kperm*(FNTF2_RanGDP_Cytosol+(-FNTF2_RanGDP_Nucleus))*Nuc_membrane
Koff_RanBP1_binding=0.5 s^(-1); Kon_RanBP1_binding=100.0 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: FCarrier_RanGTP_Cytosol + RanBP1_Cytosol => FRanBP1_Carrier_RanGTP_Cytosol, Rate Law: (Kon_RanBP1_binding*FCarrier_RanGTP_Cytosol*RanBP1_Cytosol+(-Koff_RanBP1_binding*FRanBP1_Carrier_RanGTP_Cytosol))*Cytosol
I=0.0 dimensionless*A*m^(-2); Carrier_RanGTP_Kperm=0.173333 1E-6*dimensionless*m*s^(-1)Reaction: Carrier_RanGTP_Cytosol => Carrier_RanGTP_Nucleus, Rate Law: Carrier_RanGTP_Kperm*(Carrier_RanGTP_Cytosol+(-Carrier_RanGTP_Nucleus))*Nuc_membrane
Kon_RanGTP_Carrier_binding=100.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); Koff_RanGTP_Carrier_binding=1.0 s^(-1)Reaction: Carrier_Nucleus + FRanGTP_Nucleus => FCarrier_RanGTP_Nucleus, Rate Law: (Kon_RanGTP_Carrier_binding*Carrier_Nucleus*FRanGTP_Nucleus+(-Koff_RanGTP_Carrier_binding*FCarrier_RanGTP_Nucleus))*Nucleus
Vmax_RanGTP_dephosphorylation_RanGTP_dephosphorylation = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1); Km_RanGTP_dephosphorylation=0.43 0.001*dimensionless*m^(-3)*molReaction: RanGTP_Cytosol => RanGDP_Cytosol; RanGAP_Cytosol, Rate Law: Vmax_RanGTP_dephosphorylation_RanGTP_dephosphorylation*RanGTP_Cytosol*1/(Km_RanGTP_dephosphorylation+RanGTP_Cytosol)*Cytosol
Km_dephosphorylation=0.43 0.001*dimensionless*m^(-3)*mol; Vmax_dephosphorylation_dephosphorylationF = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: FRanBP1_Carrier_RanGTP_Cytosol => FRanGDP_Cytosol + RanBP1_Cytosol + Carrier_Cytosol; RanGAP_Cytosol, Rate Law: Vmax_dephosphorylation_dephosphorylationF*FRanBP1_Carrier_RanGTP_Cytosol*1/(Km_dephosphorylation+FRanBP1_Carrier_RanGTP_Cytosol)*Cytosol
Koff_NTF2_RanGDP_unbinding=2.5 s^(-1); Kon_NTF2_RanGDP_unbinding=100.0 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: NTF2_RanGDP_Nucleus => RanGDP_Nucleus + NTF2_Nucleus, Rate Law: (Koff_NTF2_RanGDP_unbinding*NTF2_RanGDP_Nucleus+(-Kon_NTF2_RanGDP_unbinding*RanGDP_Nucleus*NTF2_Nucleus))*Nucleus
I=0.0 dimensionless*A*m^(-2); Carrier_Kperm=1.86667 1E-6*dimensionless*m*s^(-1)Reaction: Carrier_Cytosol => Carrier_Nucleus, Rate Law: Carrier_Kperm*(Carrier_Cytosol+(-Carrier_Nucleus))*Nuc_membrane
RanGDP_Kperm=0.0 1E-6*dimensionless*m*s^(-1); I=0.0 dimensionless*A*m^(-2)Reaction: FRanGDP_Cytosol => FRanGDP_Nucleus, Rate Law: RanGDP_Kperm*(FRanGDP_Cytosol+(-FRanGDP_Nucleus))*Nuc_membrane
I=0.0 dimensionless*A*m^(-2); NTF2_Kperm=3.73333 1E-6*dimensionless*m*s^(-1)Reaction: NTF2_Cytosol => NTF2_Nucleus, Rate Law: NTF2_Kperm*(NTF2_Cytosol+(-NTF2_Nucleus))*Nuc_membrane
Koff_NTF2_RanGDP_binding=2.5 s^(-1); Kon_NTF2_RanGDP_binding=100.0 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: NTF2_Cytosol + FRanGDP_Cytosol => FNTF2_RanGDP_Cytosol, Rate Law: (Kon_NTF2_RanGDP_binding*NTF2_Cytosol*FRanGDP_Cytosol+(-Koff_NTF2_RanGDP_binding*FNTF2_RanGDP_Cytosol))*Cytosol
RanGTP_Kperm=0.0 1E-6*dimensionless*m*s^(-1); I=0.0 dimensionless*A*m^(-2)Reaction: RanGTP_Cytosol => RanGTP_Nucleus, Rate Law: RanGTP_Kperm*(RanGTP_Cytosol+(-RanGTP_Nucleus))*Nuc_membrane
Vmax_RanGTP_dephosphorylation_FRanGTP_dephosphorylation = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1); Km_RanGTP_dephosphorylation=0.43 0.001*dimensionless*m^(-3)*molReaction: FRanGTP_Cytosol => FRanGDP_Cytosol; RanGAP_Cytosol, Rate Law: Vmax_RanGTP_dephosphorylation_FRanGTP_dephosphorylation*FRanGTP_Cytosol*1/(Km_RanGTP_dephosphorylation+FRanGTP_Cytosol)*Cytosol
ar_for_Microinj = 0.0Reaction: => FRanGDP_Cytosol; Pipet_Cytosol, Rate Law: ar_for_Microinj*Cytosol*1
Koff_Carrier_RanGTP_binding=0.0 s^(-1); Kon_Carrier_RanGTP_binding=0.0 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: Carrier_Cytosol + FRanGTP_Cytosol => FCarrier_RanGTP_Cytosol, Rate Law: (Kon_Carrier_RanGTP_binding*Carrier_Cytosol*FRanGTP_Cytosol+(-Koff_Carrier_RanGTP_binding*FCarrier_RanGTP_Cytosol))*Cytosol
Km_dephosphorylation=0.43 0.001*dimensionless*m^(-3)*mol; Vmax_dephosphorylation_dephosphorylation = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: RanBP1_Carrier_RanGTP_Cytosol => RanGDP_Cytosol + Carrier_Cytosol + RanBP1_Cytosol; RanGAP_Cytosol, Rate Law: Vmax_dephosphorylation_dephosphorylation*RanBP1_Carrier_RanGTP_Cytosol*1/(Km_dephosphorylation+RanBP1_Carrier_RanGTP_Cytosol)*Cytosol

States:

NameDescription
Carrier CytosolCarrier_Cytosol
FNTF2 RanGDP Cytosol[Nuclear transport factor 2; GTP-binding nuclear protein Ran; GDP; GDP]
RanGTP Nucleus[GTP; GTP-binding nuclear protein Ran; GTP-binding nuclear protein Ran; GTP; GTP]
FRanGDP Cytosol[GTP-binding nuclear protein Ran; GDP; GDP]
Carrier RanGTP Nucleus[GTP; GTP-binding nuclear protein Ran; GTP-binding nuclear protein Ran; GTP; GTP]
RanGDP Nucleus[GTP-binding nuclear protein Ran; GDP; GDP]
FRanGDP Nucleus[GTP-binding nuclear protein Ran; GDP; GDP]
NTF2 Nucleus[Nuclear transport factor 2]
NTF2 Cytosol[Nuclear transport factor 2]
Carrier RanGTP Cytosol[GTP; GTP-binding nuclear protein Ran; GTP-binding nuclear protein Ran; GTP; GTP]
RanGTP Cytosol[GTP; GTP-binding nuclear protein Ran; GTP-binding nuclear protein Ran; GTP; GTP]
NTF2 RanGDP Nucleus[GTP-binding nuclear protein Ran; Nuclear transport factor 2; GDP; GDP]
RanBP1 Carrier RanGTP Cytosol[GTP; GTP-binding nuclear protein Ran; GTP]
FNTF2 RanGDP Nucleus[Nuclear transport factor 2; GTP-binding nuclear protein Ran; GDP; GDP]
RanGDP Cytosol[GTP-binding nuclear protein Ran; GDP; GDP]
NTF2 RanGDP Cytosol[Nuclear transport factor 2; GTP-binding nuclear protein Ran; GDP; GDP]
FCarrier RanGTP Cytosol[GTP; GTP-binding nuclear protein Ran; GTP-binding nuclear protein Ran; GTP; GTP]
FRanGTP Cytosol[GTP; GTP-binding nuclear protein Ran; GTP-binding nuclear protein Ran; GTP; GTP]
FCarrier RanGTP Nucleus[GTP; GTP-binding nuclear protein Ran; GTP-binding nuclear protein Ran; GTP; GTP]
FRanBP1 Carrier RanGTP Cytosol[GTP; GTP-binding nuclear protein Ran; GTP-binding nuclear protein Ran; GTP; GTP]
FRanGTP Nucleus[GTP; GTP-binding nuclear protein Ran; GTP-binding nuclear protein Ran; GTP; GTP]
RanBP1 CytosolRanBP1_Cytosol
Carrier NucleusCarrier_Nucleus

Smolen2002_CircClock: BIOMD0000000025v0.0.1

This model originates from BioModels Database: A Database of Annotated Published Models. It is copyright (c) 2005-2010 T…

Details

Although several detailed models of molecular processes essential for circadian oscillations have been developed, their complexity makes intuitive understanding of the oscillation mechanism difficult. The goal of the present study was to reduce a previously developed, detailed model to a minimal representation of the transcriptional regulation essential for circadian rhythmicity in Drosophila. The reduced model contains only two differential equations, each with time delays. A negative feedback loop is included, in which PER protein represses per transcription by binding the dCLOCK transcription factor. A positive feedback loop is also included, in which dCLOCK indirectly enhances its own formation. The model simulated circadian oscillations, light entrainment, and a phase-response curve with qualitative similarities to experiment. Time delays were found to be essential for simulation of circadian oscillations with this model. To examine the robustness of the simplified model to fluctuations in molecule numbers, a stochastic variant was constructed. Robust circadian oscillations and entrainment to light pulses were simulated with fewer than 80 molecules of each gene product present on average. Circadian oscillations persisted when the positive feedback loop was removed. Moreover, elimination of positive feedback did not decrease the robustness of oscillations to stochastic fluctuations or to variations in parameter values. Such reduced models can aid understanding of the oscillation mechanisms in Drosophila and in other organisms in which feedback regulation of transcription may play an important role. link: http://identifiers.org/pubmed/12414672

Parameters:

NameDescription
kdp = 0.5 per_hrReaction: dClk => EmptySet, Rate Law: kdp*dClk*CELL
K1 = 0.3 nM; dClkF_tau1 = NaN nM; Vsp = 0.5 nM_per_hrReaction: EmptySet => Per; dClkF, Rate Law: Vsp*dClkF_tau1/(K1+dClkF_tau1)*CELL
kdc = 0.5 per_hrReaction: Per => EmptySet, Rate Law: kdc*Per*CELL
K2 = 0.1 nM; Vsc = 0.25 nM_per_hr; dClkF_tau2 = NaN nMReaction: EmptySet => dClk; dClkF, Rate Law: CELL*Vsc*K2/(K2+dClkF_tau2)

States:

NameDescription
dClkF[Circadian locomoter output cycles protein kaput]
dClk[Period circadian protein; Circadian locomoter output cycles protein kaput]
Per[Period circadian protein]

Smolen2004_CircClock: BIOMD0000000034v0.0.1

No inititial conditions are specified in the paper. Because there is a basal rate of transcription for each gene, it doe…

Details

A model of Drosophila circadian rhythm generation was developed to represent feedback loops based on transcriptional regulation of per, Clk (dclock), Pdp-1, and vri (vrille). The model postulates that histone acetylation kinetics make transcriptional activation a nonlinear function of [CLK]. Such a nonlinearity is essential to simulate robust circadian oscillations of transcription in our model and in previous models. Simulations suggest that two positive feedback loops involving Clk are not essential for oscillations, because oscillations of [PER] were preserved when Clk, vri, or Pdp-1 expression was fixed. However, eliminating positive feedback by fixing vri expression altered the oscillation period. Eliminating the negative feedback loop in which PER represses per expression abolished oscillations. Simulations of per or Clk null mutations, of per overexpression, and of vri, Clk, or Pdp-1 heterozygous null mutations altered model behavior in ways similar to experimental data. The model simulated a photic phase-response curve resembling experimental curves, and oscillations entrained to simulated light-dark cycles. Temperature compensation of oscillation period could be simulated if temperature elevation slowed PER nuclear entry or PER phosphorylation. The model makes experimental predictions, some of which could be tested in transgenic Drosophila. link: http://identifiers.org/pubmed/15111397

Parameters:

NameDescription
parameter_0000048 = 0.00531Reaction: species_0000001 =>, Rate Law: compartment_0000001*parameter_0000048*species_0000001
parameter_0000043 = 0.001; parameter_0000042 = 0.3186Reaction: species_0000001 => species_0000002, Rate Law: compartment_0000001*parameter_0000042*species_0000001/(parameter_0000043+species_0000001)
parameter_0000010 = 0.54; parameter_0000008 = 0.54; parameter_0000030 = 1.062; parameter_0000033 = 0.001062Reaction: => species_0000008; species_0000009, species_0000007, Rate Law: compartment_0000001*(parameter_0000030*species_0000009^2/(species_0000009^2+parameter_0000010^2)*parameter_0000008^2/(species_0000007^2+parameter_0000008^2)+parameter_0000033)
parameter_0000040 = 1.6992; parameter_0000041 = 0.25Reaction: species_0000004 => species_0000005, Rate Law: compartment_0000002*parameter_0000040*species_0000004/(parameter_0000041+species_0000004)
parameter_0000027 = 10.62; parameter_0000031 = 0.02124; parameter_0000021 = NaNReaction: => species_0000004, Rate Law: compartment_0000002*(parameter_0000027*parameter_0000021+parameter_0000031)
parameter_0000037 = 0.7434Reaction: species_0000007 =>, Rate Law: compartment_0000001*parameter_0000037*species_0000007
parameter_0000038 = 0.6903Reaction: species_0000009 =>, Rate Law: compartment_0000001*parameter_0000038*species_0000009
parameter_0000046 = 5.31; parameter_0000047 = 0.01Reaction: species_0000003 =>, Rate Law: compartment_0000001*parameter_0000046*species_0000003/(parameter_0000047+species_0000003)
parameter_0000044 = 1.6992; parameter_0000045 = 0.25Reaction: species_0000006 => species_0000001, Rate Law: compartment_0000002*parameter_0000044*species_0000006/(parameter_0000045+species_0000006)
parameter_0000020 = NaN; parameter_0000028 = 76.464; parameter_0000032 = 0.19116Reaction: => species_0000007, Rate Law: compartment_0000001*(parameter_0000028*parameter_0000020+parameter_0000032)
parameter_0000036 = 0.2124Reaction: species_0000008 =>, Rate Law: compartment_0000001*parameter_0000036*species_0000008
parameter_0000029 = 344.09; parameter_0000022 = NaN; parameter_0000034 = 0.38232; parameter_0000039 = 2.8249Reaction: => species_0000009, Rate Law: compartment_0000001*delay(parameter_0000029*parameter_0000022+parameter_0000034, parameter_0000039)

States:

NameDescription
species 0000008[Circadian locomoter output cycles protein kaput; Period circadian protein]
species 0000005[Period circadian protein]
species 0000002[Period circadian protein]
species 0000003[Period circadian protein]
species 0000001[Period circadian protein]
species 0000007[BZIP transcription factor]
species 0000004[Period circadian protein]
species 0000009[PAR domain protein 1-epislonPAR domain protein 1-epsilon]
species 0000006[Period circadian protein]

Smolen2018 - Paradoxical LTP maintenance with inhibition of protein synthesis and the proteasome: BIOMD0000000853v0.0.1

This is a mathematical model describing the formation of long-term potentiation (LTP) at the Schaffer collateral of CA1…

Details

The transition from early long-term potentiation (E-LTP) to late long-term potentiation (L-LTP) is a multistep process that involves both protein synthesis and degradation. The ways in which these two opposing processes interact to establish L-LTP are not well understood, however. For example, L-LTP is attenuated by inhibiting either protein synthesis or proteasome-dependent degradation prior to and during a tetanic stimulus (e.g., Huang et al., 1996; Karpova et al., 2006), but paradoxically, L-LTP is not attenuated when synthesis and degradation are inhibited simultaneously (Fonseca et al., 2006). These paradoxical results suggest that counter-acting 'positive' and 'negative' proteins regulate L-LTP. To investigate the basis of this paradox, we developed a model of LTP at the Schaffer collateral to CA1 pyramidal cell synapse. The model consists of nine ordinary differential equations that describe the levels of both positive- and negative-regulator proteins (PP and NP, respectively) and the transitions among five discrete synaptic states, including a basal state (BAS), three states corresponding to E-LTP (EP1, EP2, and ED), and a L-LTP state (LP). An LTP-inducing stimulus: 1) initiates the transition from BAS to EP1 and from EP1 to EP2; 2) initiates the synthesis of PP and NP; and finally; 3) activates the ubiquitin-proteasome system (UPS), which in turn, mediates transitions of EP1 and EP2 to ED and the degradation of NP. The conversion of E-LTP to L-LTP is mediated by the PP-dependent transition from ED to LP, whereas NP mediates reversal of EP2 to BAS. We found that the inclusion of the five discrete synaptic states was necessary to simulate key empirical observations: 1) normal L-LTP, 2) block of L-LTP by either proteasome inhibitor or protein synthesis inhibitor alone, and 3) preservation of L-LTP when both inhibitors are applied together. Although our model is abstract, elements of the model can be correlated with specific molecular processes. Moreover, the model correctly captures the dynamics of protein synthesis- and degradation-dependent phases of LTP, and it makes testable predictions, such as a unique synaptic state (ED) that precedes the transition from E-LTP to L-LTP, and a well-defined time window for the action of the UPS (i.e., during the transitions from EP1 and EP2 to ED). Tests of these predictions will provide new insights into the processes and dynamics of long-term synaptic plasticity. link: http://identifiers.org/pubmed/30138630

Parameters:

NameDescription
kdeg2 = 0.01; LAC = 0.0Reaction: NP => ; UPS, Rate Law: compartment*kdeg2*UPS*NP*(1-LAC)
PSI = 0.0; STIM = 1.0; ksyn2 = 2.0; ksyn2bas = 0.0014Reaction: => NP, Rate Law: compartment*(1-PSI)*(ksyn2*STIM+ksyn2bas)
kdeg3 = 0.02Reaction: STAB =>, Rate Law: compartment*kdeg3*STAB
kb3 = 0.02Reaction: ED => BAS, Rate Law: compartment*kb3*ED
kf4 = 0.02; LAC = 0.0Reaction: EP2 => ED; UPS, Rate Law: compartment*kf4*UPS*(1-LAC)*EP2
ksyn1 = 1.0; PSI = 0.0; ksyn1bas = 0.0035; STIM = 1.0Reaction: => PP, Rate Law: compartment*(1-PSI)*(ksyn1*STIM+ksyn1bas)
ksyn3 = 1.0; STIM = 1.0Reaction: => STAB, Rate Law: compartment*ksyn3*STIM
kf1bas = 0.0; STIM = 1.0Reaction: BAS => EP1, Rate Law: compartment*kf1bas*(1-STIM)*BAS
kb1 = 0.005Reaction: EP1 => BAS, Rate Law: compartment*kb1*EP1
kdeact = 0.0143Reaction: UPS =>, Rate Law: compartment*kdeact*UPS
kactbas = 0.00214Reaction: => UPS, Rate Law: compartment*kactbas
kf3 = 0.01Reaction: EP1 => EP2; STAB, Rate Law: compartment*kf3*STAB*EP1
kact = 0.2; STIM = 1.0Reaction: => UPS, Rate Law: compartment*kact*STIM
kf2 = 0.02; LAC = 0.0Reaction: EP1 => ED; UPS, Rate Law: compartment*kf2*UPS*(1-LAC)*EP1
STIM = 1.0; kf1 = 0.1Reaction: BAS => EP1, Rate Law: compartment*kf1*STIM*BAS
ksyn3bas = 0.008Reaction: => STAB, Rate Law: compartment*ksyn3bas
kb4 = 0.001Reaction: LP => BAS, Rate Law: compartment*kb4*LP
kf5 = 5.0E-4Reaction: ED => LP; PP, Rate Law: compartment*kf5*PP^2*ED
kb2 = 7.0E-4Reaction: EP2 => BAS; NP, Rate Law: compartment*kb2*EP2*NP
kdeg1 = 0.005Reaction: PP =>, Rate Law: compartment*kdeg1*PP
kdeg2bas = 0.002Reaction: NP =>, Rate Law: compartment*kdeg2bas*NP

States:

NameDescription
ED[C13281; C61589]
NP[Protein; Inhibitor]
EP1[C13281; C61589]
PP[Protein; SBO:0000459]
UPS[PW:0000144]
LP[C13281; C25322]
BAS[C13281; C90067]
EP2[C13281; C61589]
STAB[PR:000009238]

Sneppen2009 - Modeling proteasome dynamics in Parkinson's disease: BIOMD0000000548v0.0.1

Sneppen2009 - Modeling proteasome dynamics in Parkinson's diseaseThis model is described in the article: [Modeling prot…

Details

In Parkinson's disease (PD), there is evidence that alpha-synuclein (alphaSN) aggregation is coupled to dysfunctional or overburdened protein quality control systems, in particular the ubiquitin-proteasome system. Here, we develop a simple dynamical model for the on-going conflict between alphaSN aggregation and the maintenance of a functional proteasome in the healthy cell, based on the premise that proteasomal activity can be titrated out by mature alphaSN fibrils and their protofilament precursors. In the presence of excess proteasomes the cell easily maintains homeostasis. However, when the ratio between the available proteasome and the alphaSN protofilaments is reduced below a threshold level, we predict a collapse of homeostasis and onset of oscillations in the proteasome concentration. Depleted proteasome opens for accumulation of oligomers. Our analysis suggests that the onset of PD is associated with a proteasome population that becomes occupied in periodic degradation of aggregates. This behavior is found to be the general state of a proteasome/chaperone system under pressure, and suggests new interpretations of other diseases where protein aggregation could stress elements of the protein quality control system. link: http://identifiers.org/pubmed/19411740

Parameters:

NameDescription
m = 25.0; gamma = 1.0Reaction: F = m/(1+P)-gamma*F*P, Rate Law: m/(1+P)-gamma*F*P
sigma = 1.0; nu = 1.0; gamma = 1.0Reaction: P = ((sigma-P)-gamma*F*P)+nu*C, Rate Law: ((sigma-P)-gamma*F*P)+nu*C
nu = 1.0; gamma = 1.0Reaction: C = gamma*F*P-nu*C, Rate Law: gamma*F*P-nu*C

States:

NameDescription
P[proteasome complex]
C[Alpha-synuclein; proteasome complex; supramolecular fiber]
F[supramolecular fiber; Alpha-synuclein]

Sneyd1995_CalciumWave_IP3diffusion: MODEL1006230107v0.0.1

This a model from the article: Intercellular calcium waves mediated by diffusion of inositol trisphosphate: a two-dime…

Details

In response to mechanical stimulation of a single cell, airway epithelial cells in culture exhibit a wave of increased intracellular free Ca2+ concentration that spreads from cell to cell over a limited distance through the culture. We present a detailed analysis of the intercellular wave in a two-dimensional sheet of cells. The model is based on the hypothesis that the wave is the result of diffusion of inositol trisphosphate (IP3) from the stimulated cell. The two-dimensional model agrees well with experimental data and makes the following quantitative predictions: as the distance from the stimulated cells increases, 1) the intercellular delay increases exponentially, 2) the intracellular wave speed decreases exponentially, and 3) the arrival time increases exponentially. Furthermore, 4) a proportion of the cells at the periphery of the response will exhibit waves of decreased amplitude, 5) the intercellular membrane permeability to IP3 must be approximately 2 microns/s or greater, and 6) the ratio of the maximum concentration of IP3 in the stimulated cell to the Km of the IP3 receptor (with respect to IP3) must be approximately 300 or greater. These predictions constitute a rigorous test of the hypothesis that the intercellular Ca2+ waves are mediated by IP3 diffusion. link: http://identifiers.org/pubmed/7611375

Sneyd2002_IP3_Receptor: BIOMD0000000057v0.0.1

This model was successfully tested on Jarnac and MathSBML. The model reproduces the time profile of "Open Probability" o…

Details

The dynamic properties of the inositol (1,4,5)-trisphosphate (IP(3)) receptor are crucial for the control of intracellular Ca(2+), including the generation of Ca(2+) oscillations and waves. However, many models of this receptor do not agree with recent experimental data on the dynamic responses of the receptor. We construct a model of the IP(3) receptor and fit the model to dynamic and steady-state experimental data from type-2 IP(3) receptors. Our results indicate that, (i) Ca(2+) binds to the receptor using saturating, not mass-action, kinetics; (ii) Ca(2+) decreases the rate of IP(3) binding while simultaneously increasing the steady-state sensitivity of the receptor to IP(3); (iii) the rate of Ca(2+)-induced receptor activation increases with Ca(2+) and is faster than Ca(2+)-induced receptor inactivation; and (iv) IP(3) receptors are sequentially activated and inactivated by Ca(2+) even when IP(3) is bound. Our results emphasize that measurement of steady-state properties alone is insufficient to characterize the functional properties of the receptor. link: http://identifiers.org/pubmed/11842185

Parameters:

NameDescription
lminus2 = 0.8; kminus1 = 0.04; lminus2=0.8; kminus1=0.04; Phi5 = 0.0Reaction: A => I2, Rate Law: compartment*(Phi5*A-(kminus1+lminus2)*I2)
kminus3 = 29.8; kminus3=29.8; Phi3 = 0.0Reaction: O => S, Rate Law: compartment*(Phi3*O-kminus3*S)
Phi4 = 0.0; Phi_minus4 = 0.0Reaction: O => A, Rate Law: compartment*(Phi4*O-Phi_minus4*A)
lminus2 = 0.8; kminus1 = 0.04; Phi1 = 0.0; lminus2=0.8; kminus1=0.04Reaction: R => I1, Rate Law: compartment*(Phi1*R-(kminus1+lminus2)*I1)
IP3=10.0; IP3 = 10.0; Phi_minus2 = 0.0; Phi2 = 0.0Reaction: R => O, Rate Law: compartment*(Phi2*IP3*R-Phi_minus2*O)

States:

NameDescription
I1[IPR000493]
I2[IPR000493]
S[IPR000493]
A[IPR000493]
R[IPR000493]
O[IPR000493]

Sobaleva2005_ProlactinRegulation: MODEL7896869925v0.0.1

This a model from the article: Mathematical modelling of prolactin-receptor interaction and the corollary for prolacti…

Details

A mathematical model of prolactin regulating its own receptors was developed, and compared with experimental data on a qualitative level. The model incorporates the kinetics of prolactin-receptor interactions and subsequent signalling by prolactin-receptor dimers to regulate the production of receptor mRNA and hence the receptor population. The model relates changes in plasma prolactin concentration to prolactin receptor (PRLR) gene expression, and can be used for predictive purposes. The cell signalling that leads to the activation of target genes, and the mechanisms for regulation of transcription, were treated empirically in the model. The model's parameters were adjusted so that model simulations agreed with experimentally observed responses to administration of prolactin in sheep. In particular, the model correctly predicts insensitivity of receptor mRNA regulation to a series of subcutaneous injections of prolactin, versus sensitivity to prolonged infusion of prolactin. In the latter case, response was an acute down-regulation followed by a prolonged up-regulation of mRNA, with the magnitude of the up-regulation increasing with the duration of infusion period. The model demonstrates the feasibility of predicting the in vivo response of prolactin target genes to external manipulation of plasma prolactin, and could provide a useful tool for identifying optimal prolactin treatments for desirable outcomes. link: http://identifiers.org/pubmed/15757685

Sohn2010 - Genome-scale metabolic network of Pichia pastoris (PpaMBEL1254): MODEL1507180050v0.0.1

Sohn2010 - Genome-scale metabolic network of Pichia pastoris (PpaMBEL1254)This model is described in the article: [Geno…

Details

The methylotrophic yeast Pichia pastoris has gained much attention during the last decade as a platform for producing heterologous recombinant proteins of pharmaceutical importance, due to its ability to reproduce post-translational modification similar to higher eukaryotes. With the recent release of the full genome sequence for P. pastoris, in-depth study of its functions has become feasible. Here we present the first reconstruction of the genome-scale metabolic model of the eukaryote P. pastoris type strain DSMZ 70382, PpaMBEL1254, consisting of 1254 metabolic reactions and 1147 metabolites compartmentalized into eight different regions to represent organelles. Additionally, equations describing the production of two heterologous proteins, human serum albumin and human superoxide dismutase, were incorporated. The protein-producing model versions of PpaMBEL1254 were then analyzed to examine the impact on oxygen limitation on protein production. link: http://identifiers.org/pubmed/20503221

Sohn2010 - Genome-scale metabolic network of Pseudomonas putida (PpuMBEL1071): MODEL1507180043v0.0.1

Sohn2010 - Genome-scale metabolic network of Pseudomonas putida (PpuMBEL1071)This model is described in the article: [I…

Details

Genome-scale metabolic models have been appearing with increasing frequency and have been employed in a wide range of biotechnological applications as well as in biological studies. With the metabolic model as a platform, engineering strategies have become more systematic and focused, unlike the random shotgun approach used in the past. Here we present the genome-scale metabolic model of the versatile Gram-negative bacterium Pseudomonas putida, which has gained widespread interest for various biotechnological applications. With the construction of the genome-scale metabolic model of P. putida KT2440, PpuMBEL1071, we investigated various characteristics of P. putida, such as its capacity for synthesizing polyhydroxyalkanoates (PHA) and degrading aromatics. Although P. putida has been characterized as a strict aerobic bacterium, the physiological characteristics required to achieve anaerobic survival were investigated. Through analysis of PpuMBEL1071, extended survival of P. putida under anaerobic stress was achieved by introducing the ackA gene from Pseudomonas aeruginosa and Escherichia coli. link: http://identifiers.org/pubmed/20540110

Sohn2012 - Genome-scale metabolic network of Schizosaccharomyces pombe (SpoMBEL1693): MODEL1507180061v0.0.1

Sohn2012 - Genome-scale metabolic network of Schizosaccharomyces pombe (SpoMBEL1693)This model is described in the artic…

Details

BACKGROUND: Over the last decade, the genome-scale metabolic models have been playing increasingly important roles in elucidating metabolic characteristics of biological systems for a wide range of applications including, but not limited to, system-wide identification of drug targets and production of high value biochemical compounds. However, these genome-scale metabolic models must be able to first predict known in vivo phenotypes before it is applied towards these applications with high confidence. One benchmark for measuring the in silico capability in predicting in vivo phenotypes is the use of single-gene mutant libraries to measure the accuracy of knockout simulations in predicting mutant growth phenotypes. RESULTS: Here we employed a systematic and iterative process, designated as Reconciling In silico/in vivo mutaNt Growth (RING), to settle discrepancies between in silico prediction and in vivo observations to a newly reconstructed genome-scale metabolic model of the fission yeast, Schizosaccharomyces pombe, SpoMBEL1693. The predictive capabilities of the genome-scale metabolic model in predicting single-gene mutant growth phenotypes were measured against the single-gene mutant library of S. pombe. The use of RING resulted in improving the overall predictive capability of SpoMBEL1693 by 21.5%, from 61.2% to 82.7% (92.5% of the negative predictions matched the observed growth phenotype and 79.7% the positive predictions matched the observed growth phenotype). CONCLUSION: This study presents validation and refinement of a newly reconstructed metabolic model of the yeast S. pombe, through improving the metabolic model's predictive capabilities by reconciling the in silico predicted growth phenotypes of single-gene knockout mutants, with experimental in vivo growth data. link: http://identifiers.org/pubmed/22631437

Solis-perez2019 - A fractional mathematical model of breast cancer competition model: BIOMD0000000903v0.0.1

A fractional mathematical model of breast cancer competition model Author links open overlay panelJ.E.Solís-PérezaJ.F.Gó…

Details

In this paper, a mathematical model which considers population dynamics among cancer stem cells, tumor cells, healthy cells, the effects of excess estrogen and the body’s natural immune response on the cell populations was considered. Fractional derivatives with power law and exponential decay law in Liouville–Caputo sense were considered. Special solutions using an iterative scheme via Laplace transform were obtained. Furthermore, numerical simulations of the model considering both derivatives were obtained using the Atangana–Toufik numerical method. Also, random model described by a system of random differential equations was presented. The use of fractional derivatives provides more useful information about the complexity of the dynamics of the breast cancer competition model.

Volume 127, October 2019, Pages 38-54 link: http://identifiers.org/doi/10.1016/j.chaos.2019.06.027

Parameters:

NameDescription
a3 = 1250000.0; p3 = 100.0; delta = 6.0E-8Reaction: H => ; T, E, Rate Law: compartment*(delta*H*T+p3*H*E/(a3+H))
gamma1 = 3.0E-7Reaction: C => ; I, Rate Law: compartment*gamma1*I*C
tau = 2700.0Reaction: => E, Rate Law: compartment*tau
q = 0.7; M3 = 2.5E7Reaction: => H, Rate Law: compartment*q*H*(1-H/M3)
a2 = 1.135E7; mu = 0.97; a3 = 1250000.0; a1 = 1135000.0; d1 = 0.01; d2 = 0.01; d3 = 0.01Reaction: E => ; C, T, H, Rate Law: compartment*(mu+d1*C/(a1+C)+d2*T/(a2+T)+d3*H/(a3+H))*E
gamma2 = 3.0E-6; n1 = 0.01Reaction: T => ; I, Rate Law: compartment*(n1*T+gamma2*I*T)
s = 13000.0; w = 300000.0; p = 0.2Reaction: => I; T, Rate Law: compartment*(s+p*I*T/(w+T))
p1 = 600.0; M1 = 2270000.0; k1 = 0.75; a1 = 1135000.0Reaction: => C; E, Rate Law: compartment*(k1*C*(1-C/M1)+p1*C*E/(a1+C))
p2 = 0.0; M2 = 2.27E7; a2 = 1.135E7; M1 = 2270000.0; k2 = 0.514Reaction: => T; C, E, Rate Law: compartment*(k2*C*C/M1*(1-T/M2)+p2*T*E/(a2+T))
gamma3 = 1.0E-7; v = 400.0; u = 0.2; n2 = 0.29Reaction: I => ; T, E, Rate Law: compartment*(gamma3*I*T+n2*I+u*I*E/(v+E))

States:

NameDescription
I[Immune Cell]
T[Neoplastic Cell]
C[BTO:0006293]
EE
H[Healthy]

Somogyi1990_CaOscillations: BIOMD0000000114v0.0.1

This model encoded according to the paper *Hormone induced Calcium Oscillations in Liver Cells Can Be Explained by a Sim…

Details

Hormone-induced oscillations of the free intracellular calcium concentration are thought to be relevant for frequency encoding of hormone signals. In liver cells, such Ca2+ oscillations occur in response to stimulation by hormones acting via phosphoinositide breakdown. This observation may be explained by cooperative, positive feedback of Ca2+ on its own release from one inositol 1,4,5-trisphosphate-sensitive pool, obviating oscillations of inositol 1,4,5-trisphosphate. The kinetic rate laws of the associated model have a mathematical structure reminiscent of the Brusselator, a hypothetical chemical model involving a rather improbable trimolecular reaction step, thus giving a realistic biological interpretation to this hallmark of dissipative structures. We propose that calmodulin is involved in mediating this cooperativity and positive feedback, as suggested by the presented experiments. For one, hormone-induced calcium oscillations can be inhibited by the (nonphenothiazine) calmodulin antagonists calmidazolium or CGS 9343 B. Alternatively, in cells overstimulated by hormone, as characterized by a non-oscillatory elevated Ca2+ concentration, these antagonists could again restore sustained calcium oscillations. The experimental observations, including modulation of the oscillations by extracellular calcium, were in qualitative agreement with the predictions of our mathematical model. link: http://identifiers.org/pubmed/1904060

Parameters:

NameDescription
alpha = 5.0; fy = NaNReaction: x => y, Rate Law: alpha*fy*x*cytoplasm
k = 0.01; k1 = 2.0Reaction: x => y, Rate Law: k*x*cytoplasm-k1*y*ER
beta = 1.0Reaction: y =>, Rate Law: beta*y*extracellular
gamma = 1.0Reaction: => y, Rate Law: gamma*cytoplasm

States:

NameDescription
x[calcium(2+); Calcium cation]
y[calcium(2+); Calcium cation]

Somogyi1990_CaOscillations_SingleCaSpike: BIOMD0000000115v0.0.1

Another model from *Hormone induced Calcium Oscillations in Liver Cells Can Be Explained by a Simply One Pool Model.* A…

Details

Hormone-induced oscillations of the free intracellular calcium concentration are thought to be relevant for frequency encoding of hormone signals. In liver cells, such Ca2+ oscillations occur in response to stimulation by hormones acting via phosphoinositide breakdown. This observation may be explained by cooperative, positive feedback of Ca2+ on its own release from one inositol 1,4,5-trisphosphate-sensitive pool, obviating oscillations of inositol 1,4,5-trisphosphate. The kinetic rate laws of the associated model have a mathematical structure reminiscent of the Brusselator, a hypothetical chemical model involving a rather improbable trimolecular reaction step, thus giving a realistic biological interpretation to this hallmark of dissipative structures. We propose that calmodulin is involved in mediating this cooperativity and positive feedback, as suggested by the presented experiments. For one, hormone-induced calcium oscillations can be inhibited by the (nonphenothiazine) calmodulin antagonists calmidazolium or CGS 9343 B. Alternatively, in cells overstimulated by hormone, as characterized by a non-oscillatory elevated Ca2+ concentration, these antagonists could again restore sustained calcium oscillations. The experimental observations, including modulation of the oscillations by extracellular calcium, were in qualitative agreement with the predictions of our mathematical model. link: http://identifiers.org/pubmed/1904060

Parameters:

NameDescription
alpha = 10.0; fy = NaNReaction: x => y, Rate Law: alpha*fy*(x-y)*Cytosol
k = 0.01Reaction: x => y, Rate Law: Cytosol*k*(x-y)
k1 = 2.0Reaction: y => x, Rate Law: k1*y*ER
beta = 1.0Reaction: y =>, Rate Law: beta*y*Extracellular
gamma = 1.0Reaction: => y, Rate Law: gamma*Cytosol

States:

NameDescription
x[calcium(2+); Calcium cation]
y[calcium(2+); Calcium cation]

Soni2018 - IL6 induced M2 Phenotype in Leishmania major infected macrophage: BIOMD0000000873v0.0.1

IL-6 has been proposed to favor the development of Th2 responses and play an important role in the communication between…

Details

IL-6 has been proposed to favor the development of Th2 responses and play an important role in the communication between cells of multicellular organisms. They are involved in the regulation of complex cellular processes such as proliferation, differentiation and act as key player during inflammation and immune response. Th2 cytokines play an immunoregulatory role in early infection. Literature says in mice infected with L. major, IL-6 may promote the development of both Th1 and Th2 responses. IL-4 is also considered to be the signature cytokine of Th-2 response. IL-10 was initially characterized as a Th2 cytokine but later on it was proved to be a pleiotropic cytokine, secreted from different cell types including the macrophages. A major challenge is to understand how these complex non-linear processes are connected and regulated. Systems biology approaches may be used to tackle this challenge in an iterative process of quantitative mathematical analysis. In this study, we created an in silico model of IL6 mediated macrophage activation which suffers from an excessive impact of the negative feedback loop involving SOCS3. The strategy adopted in this framework may help to reduce the complexity of the leishmanial IL6 model analysis and also laydown various physiological or pathological conditions of IL6 signaling in future. link: http://identifiers.org/pubmed/29128405

Parameters:

NameDescription
mwce7a28b5_2f60_4228_a88f_ccd3a6213c3e=0.23Reaction: mw6339814d_af4c_4eee_9455_7e20795f6aeb => mwc42127ea_2b78_4381_b230_30e95cd5a9d6, Rate Law: mwce7a28b5_2f60_4228_a88f_ccd3a6213c3e*mw6339814d_af4c_4eee_9455_7e20795f6aeb*mw664a2e7f_0c35_423c_ac5d_34090e629a69
mw8d03d46b_3832_4fe6_8a6b_467ef38af206=10.0; mw658feef9_67a1_431f_95fa_7358c6b294d2=1.0E-15; mwf50b6519_aaf9_4cb0_a651_d5a3159d7390=10.0Reaction: mwacd53f34_4935_4b6a_8267_024f0d966c8c + mw57b4236e_c789_4799_a4f9_a03437a5593a => mw57b4236e_c789_4799_a4f9_a03437a5593a + mwacd53f34_4935_4b6a_8267_024f0d966c8c, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw658feef9_67a1_431f_95fa_7358c6b294d2*mw57b4236e_c789_4799_a4f9_a03437a5593a*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/(mw8d03d46b_3832_4fe6_8a6b_467ef38af206*(1+mwacd53f34_4935_4b6a_8267_024f0d966c8c*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwf50b6519_aaf9_4cb0_a651_d5a3159d7390)+mw57b4236e_c789_4799_a4f9_a03437a5593a*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22)/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mwe479dc91_2774_4239_8212_60a330a84a76=0.86Reaction: mw869055b5_5d27_4f4a_a390_b3fa48d6780e => mwc84af692_e3fc_4ede_99b6_b0cce3729bf7, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mwe479dc91_2774_4239_8212_60a330a84a76*mw869055b5_5d27_4f4a_a390_b3fa48d6780e*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw1aca690a_9ee1_460f_bc28_780fb04c1a32=1.0; mwf604d18f_ae32_4d08_bfef_f778aae8f24b=1.0Reaction: mw6f8ce639_1c28_444f_b6e6_30ff06ab0d6e + mwb9a40fab_a7c9_4984_805f_045fefc4ff32 => mwd5f166e6_df0a_45bf_b662_e87b91b79a27, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mwf604d18f_ae32_4d08_bfef_f778aae8f24b*mwb9a40fab_a7c9_4984_805f_045fefc4ff32*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/(mw1aca690a_9ee1_460f_bc28_780fb04c1a32+mwb9a40fab_a7c9_4984_805f_045fefc4ff32*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22)/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw0930c7cf_99a6_4a20_a33d_02fd90e424bb=0.24Reaction: mwc42127ea_2b78_4381_b230_30e95cd5a9d6 => mw4fd8b902_e2a2_4910_899c_2a7d3425e0e0, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw0930c7cf_99a6_4a20_a33d_02fd90e424bb*mwc42127ea_2b78_4381_b230_30e95cd5a9d6*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mwe250484c_acc9_4453_9eb6_49cda54b2f1e=0.3Reaction: mw10b15557_55f5_4525_b19d_161b056f5791 => mw6f8ce639_1c28_444f_b6e6_30ff06ab0d6e, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mwe250484c_acc9_4453_9eb6_49cda54b2f1e*mw10b15557_55f5_4525_b19d_161b056f5791*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mwd84cf612_edaf_4dd9_9abd_003cc0569864=0.0Reaction: mw9275f30d_c42b_459c_91c5_67b7e08b6486 => mwbe46ba92_97de_4cc4_970d_2dec54671573, Rate Law: mwd84cf612_edaf_4dd9_9abd_003cc0569864*mw9275f30d_c42b_459c_91c5_67b7e08b6486*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw3c07a768_8e7a_49bb_8a1d_fdc92192f92b=1.0E-15; mw2a60d98a_bc06_4e05_b965_c920da8987dc=10.0; mw9704b67a_2c16_4ac7_a311_2096ab426758=10.0Reaction: mwacd53f34_4935_4b6a_8267_024f0d966c8c + mw7f261959_39d2_4e8b_92b6_4466c2504544 => mw7f261959_39d2_4e8b_92b6_4466c2504544 + mwacd53f34_4935_4b6a_8267_024f0d966c8c, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw3c07a768_8e7a_49bb_8a1d_fdc92192f92b*mw7f261959_39d2_4e8b_92b6_4466c2504544*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/(mw9704b67a_2c16_4ac7_a311_2096ab426758*(1+mwacd53f34_4935_4b6a_8267_024f0d966c8c*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mw2a60d98a_bc06_4e05_b965_c920da8987dc)+mw7f261959_39d2_4e8b_92b6_4466c2504544*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22)/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw4594ab28_d538_4a72_875a_d878e907020d=1.0E-15Reaction: mwb167e768_b778_4072_8798_3cf19e96d1d7 + mw5b252d78_9ab9_438c_8b81_2189b1f76357 => mwf219928e_abba_4c09_9597_5d6910f7e4d9, Rate Law: mw664a2e7f_0c35_423c_ac5d_34090e629a69*mw4594ab28_d538_4a72_875a_d878e907020d*mwb167e768_b778_4072_8798_3cf19e96d1d7*mw664a2e7f_0c35_423c_ac5d_34090e629a69*mw5b252d78_9ab9_438c_8b81_2189b1f76357*mw664a2e7f_0c35_423c_ac5d_34090e629a69/mw664a2e7f_0c35_423c_ac5d_34090e629a69
mw9c4a9937_faff_4853_8a76_d17a084948d9=0.16Reaction: mwbba20281_3d8b_48c3_8e13_dee78e87dfb8 => mw78198c86_4b16_4117_8592_6a95c3953126, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw9c4a9937_faff_4853_8a76_d17a084948d9*mwbba20281_3d8b_48c3_8e13_dee78e87dfb8*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mwd01d5494_fdcd_494e_8652_87201fa5b291=0.002Reaction: mwf219928e_abba_4c09_9597_5d6910f7e4d9 => mw57b4236e_c789_4799_a4f9_a03437a5593a, Rate Law: mwd01d5494_fdcd_494e_8652_87201fa5b291*mwf219928e_abba_4c09_9597_5d6910f7e4d9*mw664a2e7f_0c35_423c_ac5d_34090e629a69
mw7b7a1d20_f258_4e3d_9234_62db60126326=6.0E-5Reaction: mw9b410665_6c5e_4f37_a7f2_0cb0963b98b1 + mw56c26af9_9f4a_4f13_936a_94ae6364342b => mwb167e768_b778_4072_8798_3cf19e96d1d7, Rate Law: mw664a2e7f_0c35_423c_ac5d_34090e629a69*mw7b7a1d20_f258_4e3d_9234_62db60126326*mw9b410665_6c5e_4f37_a7f2_0cb0963b98b1*mw664a2e7f_0c35_423c_ac5d_34090e629a69*mw56c26af9_9f4a_4f13_936a_94ae6364342b*mw664a2e7f_0c35_423c_ac5d_34090e629a69/mw664a2e7f_0c35_423c_ac5d_34090e629a69
mw87e9394c_7eef_4271_85bc_d1b0a96db1f2=0.0Reaction: mwd5f166e6_df0a_45bf_b662_e87b91b79a27 => mw9275f30d_c42b_459c_91c5_67b7e08b6486, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw87e9394c_7eef_4271_85bc_d1b0a96db1f2*mwd5f166e6_df0a_45bf_b662_e87b91b79a27*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw8f586bdf_8d42_49c1_807a_670a1c49cbd9=1.0; mw2ba0a425_a224_4fa1_affc_bcc206027a3e=1.0Reaction: mw6f8ce639_1c28_444f_b6e6_30ff06ab0d6e + mw58370246_a992_4253_8029_12fbb07a417d => mw869055b5_5d27_4f4a_a390_b3fa48d6780e, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw8f586bdf_8d42_49c1_807a_670a1c49cbd9*mw58370246_a992_4253_8029_12fbb07a417d*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/(mw2ba0a425_a224_4fa1_affc_bcc206027a3e+mw58370246_a992_4253_8029_12fbb07a417d*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22)/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw58dfe861_b815_48b4_89e9_6f71cccb561e=0.095Reaction: mw5607cee0_ee75_4065_9368_d07c3abbe18b => mw8675a533_92fb_4fbf_b747_7bca05c5841c, Rate Law: mw58dfe861_b815_48b4_89e9_6f71cccb561e*mw5607cee0_ee75_4065_9368_d07c3abbe18b*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9
mw106608bc_52fb_40e5_babb_cbb58aaaeed4=0.24Reaction: mwf219928e_abba_4c09_9597_5d6910f7e4d9 => mwce6e4efd_3187_4379_ad47_104c95e0eb3b, Rate Law: mw106608bc_52fb_40e5_babb_cbb58aaaeed4*mwf219928e_abba_4c09_9597_5d6910f7e4d9*mw664a2e7f_0c35_423c_ac5d_34090e629a69
mwd7b0f9fb_1180_47bb_b8c0_be2f89aa1c59=1.0; mw91a51f27_e4be_41a6_a967_b6a63c909652=1.0Reaction: mw7f261959_39d2_4e8b_92b6_4466c2504544 => mwd5f166e6_df0a_45bf_b662_e87b91b79a27, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw91a51f27_e4be_41a6_a967_b6a63c909652*mw7f261959_39d2_4e8b_92b6_4466c2504544*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/(mwd7b0f9fb_1180_47bb_b8c0_be2f89aa1c59+mw7f261959_39d2_4e8b_92b6_4466c2504544*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22)/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mwf8062aeb_69d6_416b_b877_9687ef6fbc80=1.0; mw7ff2e94a_ac19_47ed_9127_14d385a1e544=1.0Reaction: mw57b4236e_c789_4799_a4f9_a03437a5593a + mwb9a40fab_a7c9_4984_805f_045fefc4ff32 => mwd5f166e6_df0a_45bf_b662_e87b91b79a27, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw7ff2e94a_ac19_47ed_9127_14d385a1e544*mwb9a40fab_a7c9_4984_805f_045fefc4ff32*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/(mwf8062aeb_69d6_416b_b877_9687ef6fbc80+mwb9a40fab_a7c9_4984_805f_045fefc4ff32*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22)/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mwdf0efe39_4f88_4fc1_930b_55865b8d52f0=0.08Reaction: mw8675a533_92fb_4fbf_b747_7bca05c5841c => mw56c26af9_9f4a_4f13_936a_94ae6364342b, Rate Law: mwdf0efe39_4f88_4fc1_930b_55865b8d52f0*mw8675a533_92fb_4fbf_b747_7bca05c5841c*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw5ea6c681_bf85_4ac5_9aac_536087a97950=1.0; mw65a608c7_1026_4108_bb3d_f3358430aaf2=1.0Reaction: mwce6e4efd_3187_4379_ad47_104c95e0eb3b + mw58370246_a992_4253_8029_12fbb07a417d => mw869055b5_5d27_4f4a_a390_b3fa48d6780e, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw65a608c7_1026_4108_bb3d_f3358430aaf2*mw58370246_a992_4253_8029_12fbb07a417d*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/(mw5ea6c681_bf85_4ac5_9aac_536087a97950+mw58370246_a992_4253_8029_12fbb07a417d*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22)/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw1981fa4d_c122_41a6_8c85_55a49d408c00=400.0; mw340bc412_34ab_498a_bd42_47dd6cf025bd=1.0; mw6cf7ac60_5bab_4b73_bb47_dc064486f000=2000.0Reaction: mwba110304_bd9a_4fd0_9b4c_b8bfc975e30b => mw5607cee0_ee75_4065_9368_d07c3abbe18b + mw364ca1a4_fc56_4138_8031_16341ac865de + mw4fc13b75_10cc_41fb_b9f8_1ce95fccae73, Rate Law: mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9*mw1981fa4d_c122_41a6_8c85_55a49d408c00*(mwba110304_bd9a_4fd0_9b4c_b8bfc975e30b*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mw340bc412_34ab_498a_bd42_47dd6cf025bd/(mw6cf7ac60_5bab_4b73_bb47_dc064486f000+(mwba110304_bd9a_4fd0_9b4c_b8bfc975e30b*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mw340bc412_34ab_498a_bd42_47dd6cf025bd)/mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9
mw2081c907_9d37_48a0_8581_8b8f3d5f7148=0.1Reaction: mwd7270399_0429_4c85_920a_de2e0ae74440 => mw7f261959_39d2_4e8b_92b6_4466c2504544, Rate Law: mw2081c907_9d37_48a0_8581_8b8f3d5f7148*mwd7270399_0429_4c85_920a_de2e0ae74440*mw664a2e7f_0c35_423c_ac5d_34090e629a69
mw3bf6abba_fc8d_4254_bc9c_037ec64d3e12=1000.0; mw4c1174b0_1cba_4d7c_bedb_c29d9f65c10c=1.0; mw85474b5d_1b19_4388_bcee_098fbb23f2e0=700.0Reaction: mw55b6f083_2b28_4a1a_ab90_82a751525d72 => mwcbadb505_1cfb_4903_9975_0e53de2ba877, Rate Law: mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9*mw3bf6abba_fc8d_4254_bc9c_037ec64d3e12*(mw55b6f083_2b28_4a1a_ab90_82a751525d72*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mw4c1174b0_1cba_4d7c_bedb_c29d9f65c10c/(mw85474b5d_1b19_4388_bcee_098fbb23f2e0+(mw55b6f083_2b28_4a1a_ab90_82a751525d72*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mw4c1174b0_1cba_4d7c_bedb_c29d9f65c10c)/mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9
mw202bb744_d70a_4cf2_b2db_0d9d360bdfb5=0.1Reaction: mw78198c86_4b16_4117_8592_6a95c3953126 => mwf26f605f_29e9_4454_834c_7b3edab4bbc2, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw202bb744_d70a_4cf2_b2db_0d9d360bdfb5*mw78198c86_4b16_4117_8592_6a95c3953126*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw3288f227_0208_435a_a41c_b1778f50008c=0.09Reaction: mwf26f605f_29e9_4454_834c_7b3edab4bbc2 => mw10b15557_55f5_4525_b19d_161b056f5791, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw3288f227_0208_435a_a41c_b1778f50008c*mwf26f605f_29e9_4454_834c_7b3edab4bbc2*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mwf5ba0e23_9a0b_4bf6_b0ea_9fbc91843b5c=0.1Reaction: mwc84af692_e3fc_4ede_99b6_b0cce3729bf7 => mw55b6f083_2b28_4a1a_ab90_82a751525d72, Rate Law: mwf5ba0e23_9a0b_4bf6_b0ea_9fbc91843b5c*mwc84af692_e3fc_4ede_99b6_b0cce3729bf7*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw5b67154e_1851_4ba0_9491_7a14b2064c42=0.06Reaction: mw364ca1a4_fc56_4138_8031_16341ac865de => mwb9a40fab_a7c9_4984_805f_045fefc4ff32, Rate Law: mw5b67154e_1851_4ba0_9491_7a14b2064c42*mw364ca1a4_fc56_4138_8031_16341ac865de*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9
mwb8d480f9_5783_4feb_aaf1_2f861b09e009=0.14Reaction: mw78198c86_4b16_4117_8592_6a95c3953126 => mw7938fab7_d0c6_497b_8fb6_75922fcc19d5, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mwb8d480f9_5783_4feb_aaf1_2f861b09e009*mw78198c86_4b16_4117_8592_6a95c3953126*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw7246cef3_7a78_43b0_acb7_21533c394daa=0.001Reaction: mw85ae78ed_34f7_460c_b906_1f512a83810c + mw49322d55_ad63_4e7c_b1eb_42835c9b577a => mwd7270399_0429_4c85_920a_de2e0ae74440, Rate Law: mw664a2e7f_0c35_423c_ac5d_34090e629a69*mw7246cef3_7a78_43b0_acb7_21533c394daa*mw85ae78ed_34f7_460c_b906_1f512a83810c*mw664a2e7f_0c35_423c_ac5d_34090e629a69*mw49322d55_ad63_4e7c_b1eb_42835c9b577a*mw664a2e7f_0c35_423c_ac5d_34090e629a69/mw664a2e7f_0c35_423c_ac5d_34090e629a69
mwe808b2da_9ed2_421e_a8d9_3c3534d42ee0=0.2Reaction: mw4fc13b75_10cc_41fb_b9f8_1ce95fccae73 => mw58370246_a992_4253_8029_12fbb07a417d, Rate Law: mwe808b2da_9ed2_421e_a8d9_3c3534d42ee0*mw4fc13b75_10cc_41fb_b9f8_1ce95fccae73*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9
mw322ca6ab_81cf_4f6c_835e_48bb5f8b11de=0.0; mw051e0a03_1d45_448b_be4a_9d489e1b3ff9=0.0; mwf9b630f8_c8a5_4fd0_bef1_92819b1257d2=0.0Reaction: mwbe46ba92_97de_4cc4_970d_2dec54671573 => mw3d58864c_79c7_4ff2_98fe_1a85b2ccc43d, Rate Law: mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9*mw051e0a03_1d45_448b_be4a_9d489e1b3ff9*(mwbe46ba92_97de_4cc4_970d_2dec54671573*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mwf9b630f8_c8a5_4fd0_bef1_92819b1257d2/(mw322ca6ab_81cf_4f6c_835e_48bb5f8b11de+(mwbe46ba92_97de_4cc4_970d_2dec54671573*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mwf9b630f8_c8a5_4fd0_bef1_92819b1257d2)/mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9
mw5805aaa6_5a0a_495b_a21b_16e6ba6f39bc=0.3Reaction: mw3d58864c_79c7_4ff2_98fe_1a85b2ccc43d => mwacd53f34_4935_4b6a_8267_024f0d966c8c, Rate Law: mw5805aaa6_5a0a_495b_a21b_16e6ba6f39bc*mw3d58864c_79c7_4ff2_98fe_1a85b2ccc43d*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9
mw5b78650d_d92f_4602_bbe9_66d900ff312e=0.22Reaction: mw4fd8b902_e2a2_4910_899c_2a7d3425e0e0 => mwf06c0537_577b_4f13_a9a3_521f9d2217fb, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mw5b78650d_d92f_4602_bbe9_66d900ff312e*mw4fd8b902_e2a2_4910_899c_2a7d3425e0e0*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mw9ca6d980_6fbd_4207_9f2c_c05e2b5b0502=0.0; mwf32cef57_32ba_4c0f_bbaf_bbc90af4a8aa=0.0; mwcffa9e49_67c6_4908_9314_6c16030c8989=0.0Reaction: mwbe46ba92_97de_4cc4_970d_2dec54671573 => mw6949b379_107d_4822_b13e_680a491d2425, Rate Law: mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9*mwf32cef57_32ba_4c0f_bbaf_bbc90af4a8aa*(mwbe46ba92_97de_4cc4_970d_2dec54671573*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mwcffa9e49_67c6_4908_9314_6c16030c8989/(mw9ca6d980_6fbd_4207_9f2c_c05e2b5b0502+(mwbe46ba92_97de_4cc4_970d_2dec54671573*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mwcffa9e49_67c6_4908_9314_6c16030c8989)/mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9
mw2db26dbd_dda3_4770_b923_d2725a80ccba=1.0; mwb86b112e_674a_4e98_8ca4_3d0273a134d4=10.0; mw287bd1ba_fbc1_43a0_94ab_38a6336bbebb=10.0Reaction: mw55b6f083_2b28_4a1a_ab90_82a751525d72 => mwe3af2471_5087_4796_982a_56fad9a9a972, Rate Law: mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9*mwb86b112e_674a_4e98_8ca4_3d0273a134d4*(mw55b6f083_2b28_4a1a_ab90_82a751525d72*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mw2db26dbd_dda3_4770_b923_d2725a80ccba/(mw287bd1ba_fbc1_43a0_94ab_38a6336bbebb+(mw55b6f083_2b28_4a1a_ab90_82a751525d72*mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9)^mw2db26dbd_dda3_4770_b923_d2725a80ccba)/mwdfcbcdb1_3058_4a8b_9166_5b5e144c52c9
mwf80a94ba_03ba_44f0_b793_4f60c20fd074=8.0E-9Reaction: mwfaacd7f7_6aa9_4e6c_a7d8_281f2022ba2f + mwa244ea2a_3e41_473b_9ae7_d0db512fc366 + mw763cffc8_c121_4004_afdb_97e9de9f0081 => mw6339814d_af4c_4eee_9455_7e20795f6aeb, Rate Law: mw664a2e7f_0c35_423c_ac5d_34090e629a69*mwf80a94ba_03ba_44f0_b793_4f60c20fd074*mwfaacd7f7_6aa9_4e6c_a7d8_281f2022ba2f*mw664a2e7f_0c35_423c_ac5d_34090e629a69*mwa244ea2a_3e41_473b_9ae7_d0db512fc366*mw664a2e7f_0c35_423c_ac5d_34090e629a69*mw763cffc8_c121_4004_afdb_97e9de9f0081*mw664a2e7f_0c35_423c_ac5d_34090e629a69/mw664a2e7f_0c35_423c_ac5d_34090e629a69
mwec4950e3_ecf8_4077_9cd1_b70818e911b9=0.19Reaction: mwf06c0537_577b_4f13_a9a3_521f9d2217fb => mwbba20281_3d8b_48c3_8e13_dee78e87dfb8, Rate Law: mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22*mwec4950e3_ecf8_4077_9cd1_b70818e911b9*mwf06c0537_577b_4f13_a9a3_521f9d2217fb*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22/mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22
mwe0f7b31e_2efc_481e_90d0_e86c3b8d0be4=0.125Reaction: mw7938fab7_d0c6_497b_8fb6_75922fcc19d5 => mwba110304_bd9a_4fd0_9b4c_b8bfc975e30b, Rate Law: mwe0f7b31e_2efc_481e_90d0_e86c3b8d0be4*mw7938fab7_d0c6_497b_8fb6_75922fcc19d5*mwfef402b9_4b7e_4fbd_bba2_ff8998ab0b22

States:

NameDescription
mw763cffc8 c121 4004 afdb 97e9de9f0081LPG
mw56c26af9 9f4a 4f13 936a 94ae6364342b[Interleukin-6 receptor subunit alpha]
mw6f8ce639 1c28 444f b6e6 30ff06ab0d6e[Q4QAU2]
mw9b410665 6c5e 4f37 a7f2 0cb0963b98b1[Interleukin-6 receptor subunit alpha]
mwacd53f34 4935 4b6a 8267 024f0d966c8c[O64645]
mwa244ea2a 3e41 473b 9ae7 d0db512fc366[Q9Y2C9]
mw9275f30d c42b 459c 91c5 67b7e08b6486[Signal transducer and activator of transcription 1-alpha/beta]
mw7938fab7 d0c6 497b 8fb6 75922fcc19d5[O64645]
mw6949b379 107d 4822 b13e 680a491d2425[Nitric oxide synthase, inducible]
mw5b252d78 9ab9 438c 8b81 2189b1f76357[Q87041]
mwc42127ea 2b78 4381 b230 30e95cd5a9d6[Myeloid differentiation primary response protein MyD88]
mwf219928e abba 4c09 9597 5d6910f7e4d9[Interleukin-6 receptor subunit alpha]
mw8675a533 92fb 4fbf b747 7bca05c5841c[Interleukin-6 receptor subunit alpha]
mw7f261959 39d2 4e8b 92b6 4466c2504544[Tyrosine-protein kinase JAK1]
mw5607cee0 ee75 4065 9368 d07c3abbe18b[Interleukin-6 receptor subunit alpha]
mw3d58864c 79c7 4ff2 98fe 1a85b2ccc43d[O64645]
mwbe46ba92 97de 4cc4 970d 2dec54671573[Signal transducer and activator of transcription 1-alpha/beta]
mw4fd8b902 e2a2 4910 899c 2a7d3425e0e0[Interleukin-1 receptor-associated kinase 1]
mwce6e4efd 3187 4379 ad47 104c95e0eb3b[Tyrosine-protein kinase JAK1]
mw55b6f083 2b28 4a1a ab90 82a751525d72[Signal transducer and activator of transcription 1-alpha/beta]
mwd5f166e6 df0a 45bf b662 e87b91b79a27[5416611]
mwba110304 bd9a 4fd0 9b4c b8bfc975e30b[O64645]
mw78198c86 4b16 4117 8592 6a95c3953126[Q9VEZ5]
mw10b15557 55f5 4525 b19d 161b056f5791[A0A061RQ89]
mw6339814d af4c 4eee 9455 7e20795f6aeb[Q9Y2C9]
mwb9a40fab a7c9 4984 805f 045fefc4ff32[5416611]
mwf26f605f 29e9 4454 834c 7b3edab4bbc2[B3EWE5]
mwb167e768 b778 4072 8798 3cf19e96d1d7[Interleukin-6 receptor subunit alpha]
mwc84af692 e3fc 4ede 99b6 b0cce3729bf7[Signal transducer and activator of transcription 1-alpha/beta]
mwfaacd7f7 6aa9 4e6c a7d8 281f2022ba2f[Toll-like receptor 2]
mw58370246 a992 4253 8029 12fbb07a417d[5416611]
mw57b4236e c789 4799 a4f9 a03437a5593a[Tyrosine-protein kinase JAK1]
mw869055b5 5d27 4f4a a390 b3fa48d6780e[5416611]
mwe3af2471 5087 4796 982a 56fad9a9a972[O64645]
mw364ca1a4 fc56 4138 8031 16341ac865de[5416611]
mw4fc13b75 10cc 41fb b9f8 1ce95fccae73[5416611]
mw49322d55 ad63 4e7c b1eb 42835c9b577a[Interferon gamma]
mwd7270399 0429 4c85 920a de2e0ae74440[Interferon gamma]
mwf06c0537 577b 4f13 a9a3 521f9d2217fb[TNF receptor-associated factor 2]
mwcbadb505 1cfb 4903 9975 0e53de2ba877[Q9BRQ8]
mw85ae78ed 34f7 460c b906 1f512a83810c[Interferon gamma]
mwbba20281 3d8b 48c3 8e13 dee78e87dfb8[Q9V3Q6]

Sonntag2012 - mTOR model - IRS dependent regulation of AMPK by insulin: BIOMD0000000580v0.0.1

Sonntag2012 - mTOR model - IRS dependent regulation of AMPK by insulinTSC1-TSC2 complex has two states: 1) active (TSC1_…

Details

Mammalian target of rapamycin (mTOR) kinase responds to growth factors, nutrients and cellular energy status and is a central controller of cellular growth. mTOR exists in two multiprotein complexes that are embedded into a complex signalling network. Adenosine monophosphate-dependent kinase (AMPK) is activated by energy deprivation and shuts off adenosine 5'-triphosphate (ATP)-consuming anabolic processes, in part via the inactivation of mTORC1. Surprisingly, we observed that AMPK not only responds to energy deprivation but can also be activated by insulin, and is further induced in mTORC1-deficient cells. We have recently modelled the mTOR network, covering both mTOR complexes and their insulin and nutrient inputs. In the present study we extended the network by an AMPK module to generate the to date most comprehensive data-driven dynamic AMPK-mTOR network model. In order to define the intersection via which AMPK is activated by the insulin network, we compared simulations for six different hypothetical model structures to our observed AMPK dynamics. Hypotheses ranking suggested that the most probable intersection between insulin and AMPK was the insulin receptor substrate (IRS) and that the effects of canonical IRS downstream cues on AMPK would be mediated via an mTORC1-driven negative-feedback loop. We tested these predictions experimentally in multiple set-ups, where we inhibited or induced players along the insulin-mTORC1 signalling axis and observed AMPK induction or inhibition. We confirmed the identified model and therefore report a novel connection within the insulin-mTOR-AMPK network: we conclude that AMPK is positively regulated by IRS and can be inhibited via the negative-feedback loop. link: http://identifiers.org/pubmed/22452783

Parameters:

NameDescription
scale_PRAS40_pS183_obs = 1.0Reaction: PRAS40_pS183_obs = scale_PRAS40_pS183_obs*PRAS40_pS183, Rate Law: missing
AMPK_pT172_dephosphorylation = 0.0107214736590526Reaction: AMPK_pT172 => AMPK; AMPK_pT172, Rate Law: Cell*AMPK_pT172_dephosphorylation*AMPK_pT172
Akt_T308_phosphorylation_by_IRS1_p = 6.91810637938108Reaction: Akt_T308 => Akt_pT308; IRS1_p, Akt_T308, IRS1_p, Rate Law: Cell*Akt_T308_phosphorylation_by_IRS1_p*Akt_T308*IRS1_p
IRS1_pS636_dephosphorylation = 0.0130499987407289Reaction: IRS1_pS636 => IRS1; IRS1_pS636, Rate Law: Cell*IRS1_pS636_dephosphorylation*IRS1_pS636
PRAS40_pS183_dephosphorylation = 2.33014390064544Reaction: PRAS40_pS183 => PRAS40_S183; PRAS40_pS183, Rate Law: Cell*PRAS40_pS183_dephosphorylation*PRAS40_pS183
scale_Akt_pS473_obs = 1.0Reaction: Akt_pS473_obs = scale_Akt_pS473_obs*Akt_pS473, Rate Law: missing
scale_TSC1_TSC2_pS1387_obs = 1.0Reaction: TSC1_TSC2_pS1387_obs = scale_TSC1_TSC2_pS1387_obs*TSC1_TSC2_pS1387, Rate Law: missing
scale_AMPK_pT172_obs = 1.0Reaction: AMPK_pT172_obs = scale_AMPK_pT172_obs*AMPK_pT172, Rate Law: missing
mTORC1_S2448_activation_by_Amino_Acids = 0.00438915524637669Reaction: mTORC1 => mTORC1_pS2448; Amino_Acids, Amino_Acids, mTORC1, Rate Law: Cell*mTORC1_S2448_activation_by_Amino_Acids*mTORC1*Amino_Acids
scale_mTOR_pS2448_obs = 1.0Reaction: mTOR_pS2448_obs = scale_mTOR_pS2448_obs*mTORC1_pS2448, Rate Law: missing
IR_beta_ready = 0.0532769862975496Reaction: IR_beta_refractory => IR_beta; IR_beta_refractory, Rate Law: Cell*IR_beta_ready*IR_beta_refractory
PI3K_variant_phosphorylation_by_IR_beta_pY1146 = 0.01Reaction: PI3K_variant => PI3K_variant_p; IR_beta_pY1146, IR_beta_pY1146, PI3K_variant, Rate Law: Cell*PI3K_variant_phosphorylation_by_IR_beta_pY1146*PI3K_variant*IR_beta_pY1146
PI3K_variant_p_dephosphorylation = 10.0Reaction: PI3K_variant_p => PI3K_variant; PI3K_variant_p, Rate Law: Cell*PI3K_variant_p_dephosphorylation*PI3K_variant_p
TSC1_TSC2_T1462_phosphorylation_by_Akt_pT308 = 0.0177561800881718Reaction: TSC1_TSC2_pS1387 => TSC1_TSC2_pT1462; Akt_pT308, Akt_pT308, TSC1_TSC2_pS1387, Rate Law: Cell*TSC1_TSC2_T1462_phosphorylation_by_Akt_pT308*TSC1_TSC2_pS1387*Akt_pT308
PRAS40_S183_phosphorylation_by_mTORC1_pS2448 = 0.187621138099883Reaction: PRAS40_S183 => PRAS40_pS183; mTORC1_pS2448, PRAS40_S183, mTORC1_pS2448, Rate Law: Cell*PRAS40_S183_phosphorylation_by_mTORC1_pS2448*PRAS40_S183*mTORC1_pS2448
TSC1_TSC2_S1387_phosphorylation_by_AMPK_pT172 = 0.036558856656738Reaction: TSC1_TSC2_pT1462 => TSC1_TSC2_pS1387; AMPK_pT172, AMPK_pT172, TSC1_TSC2_pT1462, Rate Law: Cell*TSC1_TSC2_S1387_phosphorylation_by_AMPK_pT172*TSC1_TSC2_pT1462*AMPK_pT172
mTORC2_pS2481_dephosphorylation = 0.0183734532316308Reaction: mTORC2_pS2481 => mTORC2; mTORC2_pS2481, Rate Law: Cell*mTORC2_pS2481_dephosphorylation*mTORC2_pS2481
IR_beta_phosphorylation_by_Insulin = 0.124273166818913Reaction: IR_beta => IR_beta_pY1146; Insulin, IR_beta, Insulin, Rate Law: Cell*IR_beta_phosphorylation_by_Insulin*IR_beta*Insulin
IRS1_phosphorylation_by_IR_beta_pY1146 = 0.00491598674814158Reaction: IRS1 => IRS1_p; IR_beta_pY1146, IRS1, IR_beta_pY1146, Rate Law: Cell*IRS1_phosphorylation_by_IR_beta_pY1146*IRS1*IR_beta_pY1146
AMPK_T172_phosphorylation = 9.79765849087796Reaction: AMPK => AMPK_pT172; IRS1_p, AMPK, IRS1_p, Rate Law: Cell*AMPK_T172_phosphorylation*AMPK*IRS1_p
Akt_pS473_dephosphorylation = 0.00640215551178824Reaction: Akt_pS473 => Akt_S473; Akt_pS473, Rate Law: Cell*Akt_pS473_dephosphorylation*Akt_pS473
mTORC1_pS2448_dephosphorylation_by_TSC1_TSC2_pS1387 = 0.0106651971237991Reaction: mTORC1_pS2448 => mTORC1; TSC1_TSC2_pS1387, TSC1_TSC2_pS1387, mTORC1_pS2448, Rate Law: Cell*mTORC1_pS2448_dephosphorylation_by_TSC1_TSC2_pS1387*mTORC1_pS2448*TSC1_TSC2_pS1387
scale_mTOR_pS2481_obs = 1.0Reaction: mTOR_pS2481_obs = scale_mTOR_pS2481_obs*mTORC2_pS2481, Rate Law: missing
scale_p70S6K_pT389_obs = 1.0Reaction: p70S6K_pT389_obs = scale_p70S6K_pT389_obs*p70S6K_pT389, Rate Law: missing
PRAS40_T246_phosphorylation_by_Akt_pT308 = 0.137729484208433Reaction: PRAS40_T246 => PRAS40_pT246; Akt_pT308, Akt_pT308, PRAS40_T246, Rate Law: Cell*PRAS40_T246_phosphorylation_by_Akt_pT308*PRAS40_T246*Akt_pT308
mTORC2_S2481_phosphorylation_by_PI3K_variant_p = 0.37535264623552Reaction: mTORC2 => mTORC2_pS2481; PI3K_variant_p, PI3K_variant_p, mTORC2, Rate Law: Cell*mTORC2_S2481_phosphorylation_by_PI3K_variant_p*mTORC2*PI3K_variant_p
PRAS40_pT246_dephosphorylation = 1.60512543108081Reaction: PRAS40_pT246 => PRAS40_T246; PRAS40_pT246, Rate Law: Cell*PRAS40_pT246_dephosphorylation*PRAS40_pT246
Akt_S473_phosphorylation_by_mTORC2_pS2481_n_IRS1_p = 13.1441708920036Reaction: Akt_S473 => Akt_pS473; mTORC2_pS2481, IRS1_p, Akt_S473, IRS1_p, mTORC2_pS2481, Rate Law: Cell*Akt_S473_phosphorylation_by_mTORC2_pS2481_n_IRS1_p*Akt_S473*mTORC2_pS2481*IRS1_p
p70S6K_pT389_dephosphorylation = 0.0113511588360422Reaction: p70S6K_pT389 => p70S6K; p70S6K_pT389, Rate Law: Cell*p70S6K_pT389_dephosphorylation*p70S6K_pT389
p70S6K_T389_phosphorylation_by_mTORC1_pS2448 = 0.00184042775983938Reaction: p70S6K => p70S6K_pT389; mTORC1_pS2448, mTORC1_pS2448, p70S6K, Rate Law: Cell*p70S6K_T389_phosphorylation_by_mTORC1_pS2448*p70S6K*mTORC1_pS2448
scale_IR_beta_pY1146_obs = 1.0Reaction: IR_beta_pY1146_obs = scale_IR_beta_pY1146_obs*IR_beta_pY1146, Rate Law: missing
scale_Akt_pT308_obs = 1.0Reaction: Akt_pT308_obs = scale_Akt_pT308_obs*Akt_pT308, Rate Law: missing
IRS1_p_phosphorylation_by_p70S6K_pT389 = 1682.74838380846Reaction: IRS1_p => IRS1_pS636; p70S6K_pT389, IRS1_p, p70S6K_pT389, Rate Law: Cell*IRS1_p_phosphorylation_by_p70S6K_pT389*IRS1_p*p70S6K_pT389
Akt_pT308_dephosphorylation = 0.00335544587646129Reaction: Akt_pT308 => Akt_T308; Akt_pT308, Rate Law: Cell*Akt_pT308_dephosphorylation*Akt_pT308
IR_beta_pY1146_dephosphorylation = 0.396235078561935Reaction: IR_beta_pY1146 => IR_beta_refractory; IR_beta_pY1146, Rate Law: Cell*IR_beta_pY1146_dephosphorylation*IR_beta_pY1146
scale_PRAS40_pT246_obs = 1.0Reaction: PRAS40_pT246_obs = scale_PRAS40_pT246_obs*PRAS40_pT246, Rate Law: missing
scale_IRS1_pS636_obs = 1.0Reaction: IRS1_pS636_obs = scale_IRS1_pS636_obs*IRS1_pS636, Rate Law: missing

States:

NameDescription
mTORC1 pS2448[Serine/threonine-protein kinase mTOR]
IRS1 pS636[Insulin receptor substrate 1; Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform]
mTOR pS2448 obs[Serine/threonine-protein kinase mTOR]
AMPK pT172 obs[5'-AMP-activated protein kinase subunit gamma-1; 5'-AMP-activated protein kinase catalytic subunit alpha-1; 5'-AMP-activated protein kinase subunit beta-1]
Akt S473[RAC-alpha serine/threonine-protein kinase]
IRS1 pS636 obs[Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform; Insulin receptor substrate 1]
IRS1 p[Insulin receptor substrate 1; Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform]
IR beta[Insulin receptor]
AMPK pT172[5'-AMP-activated protein kinase subunit gamma-1; 5'-AMP-activated protein kinase subunit beta-1; 5'-AMP-activated protein kinase catalytic subunit alpha-1]
PRAS40 S183[Proline-rich AKT1 substrate 1]
PRAS40 pT246[Proline-rich AKT1 substrate 1]
PI3K variantPI3K_variant
PI3K variant pPI3K_variant_p
PRAS40 pT246 obs[Proline-rich AKT1 substrate 1]
PRAS40 pS183[Proline-rich AKT1 substrate 1]
mTOR pS2481 obs[Serine/threonine-protein kinase mTOR]
Akt pS473[RAC-alpha serine/threonine-protein kinase]
p70S6K pT389 obs[Ribosomal protein S6 kinase beta-1]
PRAS40 T246[Proline-rich AKT1 substrate 1]
mTORC2[Serine/threonine-protein kinase mTOR]
Akt pS473 obs[RAC-alpha serine/threonine-protein kinase]
TSC1 TSC2 pT1462[Hamartin; Tuberin]
IR beta refractory[Insulin receptor]
Akt pT308[RAC-alpha serine/threonine-protein kinase]
PRAS40 pS183 obs[Proline-rich AKT1 substrate 1]
Akt T308[RAC-alpha serine/threonine-protein kinase]
Insulin[Insulin]
mTORC1[Serine/threonine-protein kinase mTOR]
AMPK[5'-AMP-activated protein kinase subunit beta-1; 5'-AMP-activated protein kinase subunit gamma-1; 5'-AMP-activated protein kinase catalytic subunit alpha-1]
Amino AcidsAmino_Acids
IRS1[Insulin receptor substrate 1; Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform]
IR beta pY1146[Insulin receptor]
TSC1 TSC2 pS1387[Tuberin; Hamartin]
p70S6K pT389[Ribosomal protein S6 kinase beta-1]
IR beta pY1146 obs[Insulin receptor]
mTORC2 pS2481[Serine/threonine-protein kinase mTOR]
TSC1 TSC2 pS1387 obs[Tuberin; Hamartin]
p70S6K[Ribosomal protein S6 kinase beta-1]
Akt pT308 obs[RAC-alpha serine/threonine-protein kinase]

Sorokina2009_PhyA_FHL_ON_OFF_9h: MODEL0911120000v0.0.1

Final version obtained by merging of the PhyA_FHL model with On_OFF 9h long experiment This model originates from BioMod…

Details

Advances in synthetic biology will require spatio-temporal regulation of biological processes in heterologous host cells. We develop a light-switchable, two-hybrid interaction in yeast, based upon the Arabidopsis proteins PHYTOCHROME A and FAR-RED ELONGATED HYPOCOTYL 1-LIKE. Light input to this regulatory module allows dynamic control of a light-emitting LUCIFERASE reporter gene, which we detect by real-time imaging of yeast colonies on solid media.The reversible activation of the phytochrome by red light, and its inactivation by far-red light, is retained. We use this quantitative readout to construct a mathematical model that matches the system's behaviour and predicts the molecular targets for future manipulation.Our model, methods and materials together constitute a novel system for a eukaryotic host with the potential to convert a dynamic pattern of light input into a predictable gene expression response. This system could be applied for the regulation of genetic networks - both known and synthetic. link: http://identifiers.org/pubmed/19761615

Sorokina2011_StarchMetabolism_Ostreococcus: MODEL1204240000v0.0.1

This model is from the article: Microarray data can predict diurnal changes of starch content in the picoalga Ostreoco…

Details

The storage of photosynthetic carbohydrate products such as starch is subject to complex regulation, effected at both transcriptional and post-translational levels. The relevant genes in plants show pronounced daily regulation. Their temporal RNA expression profiles, however, do not predict the dynamics of metabolite levels, due to the divergence of enzyme activity from the RNA profiles.Unicellular phytoplankton retains the complexity of plant carbohydrate metabolism, and recent transcriptomic profiling suggests a major input of transcriptional regulation.We used a quasi-steady-state, constraint-based modelling approach to infer the dynamics of starch content during the 12 h light/12 h dark cycle in the model alga Ostreococcus tauri. Measured RNA expression datasets from microarray analysis were integrated with a detailed stoichiometric reconstruction of starch metabolism in O. tauri in order to predict the optimal flux distribution and the dynamics of the starch content in the light/dark cycle. The predicted starch profile was validated by experimental data over the 24 h cycle. The main genetic regulatory targets within the pathway were predicted by in silico analysis.A single-reaction description of starch production is not able to account for the observed variability of diurnal activity profiles of starch-related enzymes. We developed a detailed reaction model of starch metabolism, which, to our knowledge, is the first attempt to describe this polysaccharide polymerization while preserving the mass balance relationships. Our model and method demonstrate the utility of a quasi-steady-state approach for inferring dynamic metabolic information in O. tauri directly from time-series gene expression data. link: http://identifiers.org/pubmed/21352558

Sotolongo-Costa2003 - Behavior of tumors under nonstationary therapy: BIOMD0000000785v0.0.1

This model describes the interaction dynamics of a lymphocyte-tumor cell population.

Details

We present a model for the interaction dynamics of lymphocytes-tumor cells population. This model reproduces all known states for the tumor. Further, we develop it taking into account periodical immunotherapy treatment with cytokines alone. A detailed analysis for the evolution of tumor cells as a function of frequency and therapy burden applied for the periodical treatment is carried out. Certain threshold values for the frequency and applied doses are derived from this analysis. So it seems possible to control and reduce the growth of the tumor. Also, constant values for cytokines doses seems to be a successful treatment. link: http://identifiers.org/doi/10.1016/S0167-2789(03)00005-8

Parameters:

NameDescription
alpha = 2.0Reaction: y_Lymphocytes =>, Rate Law: compartment*y_Lymphocytes/alpha
k = 0.2Reaction: y_Lymphocytes => ; x_Malignant_Cells, Rate Law: compartment*k*x_Malignant_Cells
sigma = 0.25Reaction: => y_Lymphocytes, Rate Law: compartment*sigma

States:

NameDescription
x Malignant Cells[neoplastic cell]
y Lymphocytes[lymphocyte]

Sriram2007_CellCycle: BIOMD0000000181v0.0.1

The model reproduces the time profile of species depicted in Figure 12a and 12 b. The authors communicated to the curato…

Details

A novel topology of regulatory networks abstracted from the budding yeast cell cycle is studied by constructing a simple nonlinear model. A ternary positive feedback loop with only positive regulations is constructed with elements that activates the subsequent element in a clockwise fashion. A ternary negative feedback loop with only negative regulations is constructed with the elements that inhibit the subsequent element in an anticlockwise fashion. Positive feedback loop exhibits bistability, whereas the negative feedback loop exhibits limit cycle oscillations. The novelty of the topology is that the corresponding elements in these two homogeneous feedback loops are linked by the binary positive feedback loops with only positive regulations. This results in the emergence of mixed feedback loops in the network that displays complex behaviour like the coexistence of multiple steady states, relaxation oscillations and chaos. Importantly, the arrangement of the feedback loops brings in the notion of checkpoint in the model. The model also exhibits domino-like behaviour, where the limit cycle oscillations take place in a stepwise fashion. As the aforementioned topology is abstracted from the budding yeast cell cycle, the events that govern the cell cycle are considered for the present study. In budding yeast, the sequential activation of the transcription factors, cyclins and their inhibitors form mixed feedback loops. The transcription factors that involve in the positive regulation in a clockwise orientation generates ternary positive feedback loop, while the cyclins and their inhibitors that involve in the negative regulation in an anticlockwise orientation generates ternary negative feedback loop. The mutual regulation between the corresponding elements in the transcription factors and the cyclins and their inhibitors generates binary positive feedback loops. The bifurcation diagram constructed for the whole system can be related to the different events of the cell cycle in terms of dynamical system theory. The checkpoint mechanism that plays an important role in different phases of the cell cycle are accounted for by silencing appropriate feedback loops in the model. link: http://identifiers.org/pubmed/18203579

Parameters:

NameDescription
kc2 = 0.22 min_1Reaction: => T2; C2, Rate Law: compartment*kc2*C2
j1 = 0.9 nM_min_1Reaction: => T1, Rate Law: compartment*j1
vd1 = 6.0 nM_min_1; km1 = 5.0 nM; n = 2.0 dimensionlessReaction: => T1; T3, Rate Law: compartment*vd1*T3^n/(km1^n+T3^n)
k110 = 10.0 nM; n = 2.0 dimensionless; v11 = 15.0 nM_min_1Reaction: => C2; T2, C3, Rate Law: compartment*v11*T2^n/(k110^n+T2^n+C3^n)
kd4 = 0.16 min_1Reaction: C1 =>, Rate Law: compartment*kd4*C1
kc1 = 0.2 min_1Reaction: => T1; C1, Rate Law: compartment*kc1*C1
j2 = 0.5 nM_min_1Reaction: => T2, Rate Law: compartment*j2
kc3 = 0.6 min_1Reaction: => T3; C3, Rate Law: compartment*kc3*C3
v12 = 15.0 nM_min_1; n = 2.0 dimensionless; k120 = 10.0 nMReaction: => C1; T1, C2, Rate Law: compartment*v12*T1^n/(k120^n+T1^n+C2^n)
vd3 = 3.0 nM_min_1; km3 = 5.0 nM; n = 2.0 dimensionlessReaction: => T3; T2, Rate Law: compartment*vd3*T2^n/(km3^n+T2^n)
kd2 = 0.9 min_1Reaction: T2 =>, Rate Law: compartment*kd2*T2
v10 = 15.0 nM_min_1; k100 = 10.0 nM; n = 2.0 dimensionlessReaction: => C3; T3, C1, Rate Law: compartment*v10*T3^n/(k100^n+T3^n+C1^n)
vd2 = 1.052 nM_min_1; n = 2.0 dimensionless; km2 = 5.0 nMReaction: => T2; T1, Rate Law: compartment*vd2*T1^n/(km2^n+T1^n)
kd6 = 0.16 min_1Reaction: C3 =>, Rate Law: compartment*kd6*C3
kd5 = 0.16 min_1Reaction: C2 =>, Rate Law: compartment*kd5*C2
kd3 = 0.8 min_1Reaction: T3 =>, Rate Law: compartment*kd3*T3
j3 = 0.2 nM_min_1Reaction: => T3, Rate Law: compartment*j3
kd1 = 0.8 min_1Reaction: T1 =>, Rate Law: compartment*kd1*T1

States:

NameDescription
T3[Transcriptional factor SWI5]
C1[G1/S-specific cyclin CLN1; Cyclin-dependent kinase 1]
C2[G2/mitotic-specific cyclin-1; Cyclin-dependent kinase 1]
C3[Protein SIC1]
T1[Regulatory protein SWI4]
T2[Pheromone receptor transcription factor]

Srividhya2006_CellCycle: BIOMD0000000196v0.0.1

In this model the values of "free CDK" (Id: x2), "cdc25_P" (x4) "Wee1_P" (Id: y5) and "APC" (Id: y6) are assigned using…

Details

We propose a seven variable model with time delay in one of the variables for the cell cycle in higher eukaryotes. The model consists of four important phosphorylation-dephosphorylation (P-D) cycles that govern the cell cycle, namely Pre-MPF-MPF, Cdc25P-Cdc25, Wee1P-Wee1 and APCP-APC. Other variables are cyclin, free cyclin dependent kinase (Cdk) and mass. The mass acts as a G2/M checkpoint and the checkpoint is represented by a saddle node loop bifurcation. The key feature of the model is that a time lag has been introduced in the activation of anaphase promoting complex (APC) by maturation promoting factor (MPF). This is effected by treating MPF as a time-delayed variable in the activation step of APC. The time lag acts as a spindle checkpoint. Absence of time delay induces a bistability in our model. Time delay also brings about variability in G1 phase timings. The model also reproduces the mutant phenotype experiments on wee1 cells. Stochasticity has been introduced in the model to simulate the dependence of the cycle time on cell birth length. Mutant phenotypes in the stochastic model reproduce the experimental observations better than the deterministic model. link: http://identifiers.org/pubmed/16473373

Parameters:

NameDescription
Ka = 0.5; j1_2 = 0.01; vM1_2 = 0.55; B1 = 5.0Reaction: x3 => Pre_MPF; x5, Rate Law: vM1_2*(1+B1*x5/(Ka+x5))*x3/(j1_2+x3)
Ka = 0.5; j1 = 0.01; a1 = 1.2; vM1 = 0.7Reaction: Pre_MPF => x3; x4, Rate Law: vM1*(1+a1*x4/(Ka+x4))*Pre_MPF/(j1+Pre_MPF)
totwee1 = 1.0Reaction: y5 = totwee1-x5, Rate Law: missing
kf = 1.0Reaction: x1 + x2 => Pre_MPF, Rate Law: kf*x1*x2
vM3 = 1.0; j3_2 = 0.01Reaction: y5 => x5, Rate Law: vM3*(1-x5)/((j3_2+1)-x5)
B3 = 1.0; Ka = 0.5; j3 = 0.01; vM3_2 = 1.0Reaction: x5 => y5; x3, m, Rate Law: vM3_2*(1+B3*m*x3/(Ka+m*x3))*x5/(j3+x5)
totcdc25 = 1.0Reaction: x4 = totcdc25-y4, Rate Law: missing
kd = 0.2; B2 = 3.3Reaction: x3 => x2; x6, Rate Law: kd*(1+B2*x6)*x3
c = 1.1Reaction: x2 = (c-Pre_MPF)-x3, Rate Law: missing
a = 10.0; mu = 0.01Reaction: => m, Rate Law: mu*m*(1-m/a)
vM2_2 = 1.0; j2_2 = 0.01Reaction: x4 => y4, Rate Law: vM2_2*x4/(j2_2+x4)
Ka = 0.5; vM2 = 0.41; j2 = 0.01; a2 = 1.0Reaction: y4 => x4; m, x3, Rate Law: vM2*(1+a2*m*x3/(Ka+m*x3))*(1-x4)/((j2+1)-x4)
vf = 0.215Reaction: => x1, Rate Law: vf
Bc = 3.5; kc = 0.05Reaction: x1 => ; x6, Rate Law: x1*(kc+Bc*x6)
j4_2 = 0.01; vM4_2 = 1.0Reaction: x6 => y6, Rate Law: vM4_2*x6/(j4_2+x6)
Ka = 0.5; j4 = 0.01; a4 = 2.0; vM4 = 0.7; tau = 5.0Reaction: y6 => x6; m, x3, Rate Law: vM4*(1+a4*m*delay(x3, tau)/(Ka+m*delay(x3, tau)))*(1-x6)/(j4+(1-x6))
totAPC = 1.0Reaction: y6 = totAPC-x6, Rate Law: missing

States:

NameDescription
x5[Wee1-like protein kinase]
Pre MPF[Cyclin-dependent kinase 1; IPR015454; MPF complex; cyclin-dependent protein kinase holoenzyme complex]
x4[Cell division control protein 25; M-phase inducer phosphatase 1; Phosphoprotein]
x6[Phosphoprotein; anaphase-promoting complex]
x2[Cyclin-dependent kinase 1; protein kinase activity]
x3[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin-B1; MPF complex]
x1[G2/mitotic-specific cyclin-B1; cyclin-dependent protein serine/threonine kinase regulator activity]
y4[Cell division control protein 25; M-phase inducer phosphatase 1]
m[cell]
y6[anaphase-promoting complex]
y5[Wee1-like protein kinase; Phosphoprotein]

Stanford2013 - Kinetic model of yeast metabolic network (regulation): BIOMD0000000497v0.0.1

Stanford2013 - Kinetic model of yeast metabolic network (standard)Large-scale model construction based on a logical laye…

Details

The quantitative effects of environmental and genetic perturbations on metabolism can be studied in silico using kinetic models. We present a strategy for large-scale model construction based on a logical layering of data such as reaction fluxes, metabolite concentrations, and kinetic constants. The resulting models contain realistic standard rate laws and plausible parameters, adhere to the laws of thermodynamics, and reproduce a predefined steady state. These features have not been simultaneously achieved by previous workflows. We demonstrate the advantages and limitations of the workflow by translating the yeast consensus metabolic network into a kinetic model. Despite crudely selected data, the model shows realistic control behaviour, a stable dynamic, and realistic response to perturbations in extracellular glucose concentrations. The paper concludes by outlining how new data can continuously be fed into the workflow and how iterative model building can assist in directing experiments. link: http://identifiers.org/pubmed/24324546

Parameters:

NameDescription
Vmax_r_0529=4.51989; kmp_s_0735r_0529=0.601873; kms_s_1315r_0529=12.8511; Keq_r_0529=0.0515178; kms_s_0657r_0529=0.549; kmp_s_0659r_0529=0.549Reaction: s_0657 + s_1315 => s_0659 + s_0735; s_0657, s_0659, s_0735, s_1315, Rate Law: intracellular*Vmax_r_0529*(1/kms_s_0657r_0529)^1*(1/kms_s_1315r_0529)^1*(s_0657^1*s_1315^1-s_0659^1*s_0735^1/Keq_r_0529)/(((1+s_0657/kms_s_0657r_0529)*(1+s_1315/kms_s_1315r_0529)+(1+s_0659/kmp_s_0659r_0529)*(1+s_0735/kmp_s_0735r_0529))-1)/intracellular
kmp_s_0692r_0171=0.549; Keq_r_0171=1.38552; kmp_s_0434r_0171=1.25956; Vmax_r_0171=0.395998; kms_s_1053r_0171=0.549Reaction: s_1053 => s_0434 + s_0692; s_0434, s_0692, s_1053, Rate Law: intracellular*Vmax_r_0171*(1/kms_s_1053r_0171)^1*(s_1053^1-s_0434^1*s_0692^1/Keq_r_0171)/((1+s_1053/kms_s_1053r_0171+(1+s_0434/kmp_s_0434r_0171)*(1+s_0692/kmp_s_0692r_0171))-1)/intracellular
Vmax_r_1036=0.14014; kms_s_0731r_1036=0.0436363; kmp_s_0561r_1036=0.549; Keq_r_1036=13.8394; kmp_s_0427r_1036=0.549; kms_s_1304r_1036=0.549Reaction: s_0731 + s_1304 => s_0427 + s_0561; s_0427, s_0561, s_0731, s_1304, Rate Law: intracellular*Vmax_r_1036*(1/kms_s_0731r_1036)^1*(1/kms_s_1304r_1036)^1*(s_0731^1*s_1304^1-s_0427^1*s_0561^1/Keq_r_1036)/(((1+s_0731/kms_s_0731r_1036)*(1+s_1304/kms_s_1304r_1036)+(1+s_0427/kmp_s_0427r_1036)*(1+s_0561/kmp_s_0561r_1036))-1)/intracellular
kms_s_1096r_0467=0.549; kmp_s_0514r_0467=0.549; kms_s_0763_br_0467=0.549; kms_s_1187r_0467=0.549; Vmax_r_0467=0.00599719; Keq_r_0467=3.64962; kmp_s_1091r_0467=0.549; kms_s_1005r_0467=0.549; kmp_s_1334r_0467=0.549; kmp_s_0470r_0467=1.0; kmp_s_1434_br_0467=0.549Reaction: s_0763_b + s_1005 + s_1096 + s_1187 => s_0470 + s_0514 + s_1091 + s_1334 + s_1434_b; s_0470, s_0514, s_0763_b, s_1005, s_1091, s_1096, s_1187, s_1334, s_1434_b, Rate Law: intracellular*Vmax_r_0467*(1/kms_s_0763_br_0467)^3*(1/kms_s_1005r_0467)^1*(1/kms_s_1096r_0467)^2*(1/kms_s_1187r_0467)^1*(s_0763_b^3*s_1005^1*s_1096^2*s_1187^1-s_0470^1*s_0514^1*s_1091^2*s_1334^1*s_1434_b^1/Keq_r_0467)/(((1+s_0763_b/kms_s_0763_br_0467)*(1+s_1005/kms_s_1005r_0467)*(1+s_1096/kms_s_1096r_0467)*(1+s_1187/kms_s_1187r_0467)+(1+s_0470/kmp_s_0470r_0467)*(1+s_0514/kmp_s_0514r_0467)*(1+s_1091/kmp_s_1091r_0467)*(1+s_1334/kmp_s_1334r_0467)*(1+s_1434_b/kmp_s_1434_br_0467))-1)/intracellular
Vmax_r_1038=0.1001; kms_s_1434_br_1038=0.549; Keq_r_1038=1.1; kms_s_0419r_1038=0.549; kmp_s_0416r_1038=0.549; kmp_s_1207r_1038=0.549Reaction: s_0419 + s_1434_b => s_0416 + s_1207; s_0416, s_0419, s_1207, s_1434_b, Rate Law: intracellular*Vmax_r_1038*(1/kms_s_0419r_1038)^1*(1/kms_s_1434_br_1038)^1*(s_0419^1*s_1434_b^1-s_0416^1*s_1207^1/Keq_r_1038)/(((1+s_0419/kms_s_0419r_1038)*(1+s_1434_b/kms_s_1434_br_1038)+(1+s_0416/kmp_s_0416r_1038)*(1+s_1207/kmp_s_1207r_1038))-1)/intracellular
Keq_r_0886=0.950614; kmp_s_0009r_0886=0.549; kms_s_0446r_0886=1.09208; kmp_s_1207r_0886=0.549; kmp_s_0763_br_0886=0.549; kms_s_0318r_0886=0.549; Vmax_r_0886=1.53571; kms_s_0881r_0886=0.549; kmp_s_0400r_0886=1.71907Reaction: s_0318 + s_0446 + s_0881 => s_0009 + s_0400 + s_0763_b + s_1207; s_0009, s_0318, s_0400, s_0446, s_0763_b, s_0881, s_1207, Rate Law: intracellular*Vmax_r_0886*(1/kms_s_0318r_0886)^1*(1/kms_s_0446r_0886)^1*(1/kms_s_0881r_0886)^1*(s_0318^1*s_0446^1*s_0881^1-s_0009^1*s_0400^1*s_0763_b^1*s_1207^1/Keq_r_0886)/(((1+s_0318/kms_s_0318r_0886)*(1+s_0446/kms_s_0446r_0886)*(1+s_0881/kms_s_0881r_0886)+(1+s_0009/kmp_s_0009r_0886)*(1+s_0400/kmp_s_0400r_0886)*(1+s_0763_b/kmp_s_0763_br_0886)*(1+s_1207/kmp_s_1207r_0886))-1)/intracellular
kmp_s_0514r_0442=0.549; kmp_s_1132r_0442=0.549; Vmax_r_0442=0.001914; kms_s_0605r_0442=0.549; kmp_s_0446r_0442=1.09208; kms_s_0434r_0442=1.25956; kms_s_1140r_0442=0.549; Keq_r_0442=0.953736Reaction: s_0434 + s_0605 + s_1140 => s_0446 + s_0514 + s_1132; s_0434, s_0446, s_0514, s_0605, s_1132, s_1140, Rate Law: intracellular*Vmax_r_0442*(1/kms_s_0434r_0442)^1*(1/kms_s_0605r_0442)^1*(1/kms_s_1140r_0442)^1*(s_0434^1*s_0605^1*s_1140^1-s_0446^1*s_0514^1*s_1132^1/Keq_r_0442)/(((1+s_0434/kms_s_0434r_0442)*(1+s_0605/kms_s_0605r_0442)*(1+s_1140/kms_s_1140r_0442)+(1+s_0446/kmp_s_0446r_0442)*(1+s_0514/kmp_s_0514r_0442)*(1+s_1132/kmp_s_1132r_0442))-1)/intracellular
Keq_r_0528=0.0128394; Vmax_r_0528=3.48809; kmp_s_0732r_0528=0.15; kms_s_1315r_0528=12.8511; kms_s_1434_br_0528=0.549; kmp_s_1207r_0528=0.549Reaction: s_1315 + s_1434_b => s_0732 + s_1207; s_0732, s_1207, s_1315, s_1434_b, Rate Law: intracellular*Vmax_r_0528*(1/kms_s_1315r_0528)^1*(1/kms_s_1434_br_0528)^1*(s_1315^1*s_1434_b^1-s_0732^1*s_1207^1/Keq_r_0528)/(((1+s_1315/kms_s_1315r_0528)*(1+s_1434_b/kms_s_1434_br_0528)+(1+s_0732/kmp_s_0732r_0528)*(1+s_1207/kmp_s_1207r_0528))-1)/intracellular
Vmax_r_0021=1.60931; Keq_r_0021=40.5765; kms_s_1243r_0021=0.0271093; kms_s_1434_br_0021=0.549; kmp_s_0356r_0021=0.549; kmp_s_1207r_0021=0.549; kms_s_0533r_0021=0.549Reaction: s_0533 + s_1243 + s_1434_b => s_0356 + s_1207; s_0356, s_0533, s_1207, s_1243, s_1434_b, Rate Law: intracellular*Vmax_r_0021*(1/kms_s_0533r_0021)^1*(1/kms_s_1243r_0021)^1*(1/kms_s_1434_br_0021)^1*(s_0533^1*s_1243^1*s_1434_b^1-s_0356^1*s_1207^1/Keq_r_0021)/(((1+s_0533/kms_s_0533r_0021)*(1+s_1243/kms_s_1243r_0021)*(1+s_1434_b/kms_s_1434_br_0021)+(1+s_0356/kmp_s_0356r_0021)*(1+s_1207/kmp_s_1207r_0021))-1)/intracellular
kms_s_0366r_0183=0.120104; kmp_s_1082r_0183=1.50326; kms_s_0763_br_0183=0.549; kms_s_1087r_0183=0.0867353; Keq_r_0183=14456.7; Vmax_r_0183=99.1; kmp_s_0650r_0183=50.0Reaction: s_0366 + s_0763_b + s_1087 => s_0650 + s_1082; s_0366, s_0650, s_0763_b, s_1082, s_1087, Rate Law: intracellular*Vmax_r_0183*(1/kms_s_0366r_0183)^1*(1/kms_s_0763_br_0183)^1*(1/kms_s_1087r_0183)^1*(s_0366^1*s_0763_b^1*s_1087^1-s_0650^1*s_1082^1/Keq_r_0183)/(((1+s_0366/kms_s_0366r_0183)*(1+s_0763_b/kms_s_0763_br_0183)*(1+s_1087/kms_s_1087r_0183)+(1+s_0650/kmp_s_0650r_0183)*(1+s_1082/kmp_s_1082r_0183))-1)/intracellular
Keq_r_0465=3.64962; kmp_s_0470r_0465=1.0; kmp_s_1044r_0465=0.549; kms_s_0763_br_0465=0.549; kmp_s_1091r_0465=0.549; kms_s_0977r_0465=0.549; Vmax_r_0465=0.0179399; kms_s_1005r_0465=0.549; kmp_s_0514r_0465=0.549; kmp_s_1434_br_0465=0.549; kms_s_1096r_0465=0.549Reaction: s_0763_b + s_0977 + s_1005 + s_1096 => s_0470 + s_0514 + s_1044 + s_1091 + s_1434_b; s_0470, s_0514, s_0763_b, s_0977, s_1005, s_1044, s_1091, s_1096, s_1434_b, Rate Law: intracellular*Vmax_r_0465*(1/kms_s_0763_br_0465)^3*(1/kms_s_0977r_0465)^1*(1/kms_s_1005r_0465)^1*(1/kms_s_1096r_0465)^2*(s_0763_b^3*s_0977^1*s_1005^1*s_1096^2-s_0470^1*s_0514^1*s_1044^1*s_1091^2*s_1434_b^1/Keq_r_0465)/(((1+s_0763_b/kms_s_0763_br_0465)*(1+s_0977/kms_s_0977r_0465)*(1+s_1005/kms_s_1005r_0465)*(1+s_1096/kms_s_1096r_0465)+(1+s_0470/kmp_s_0470r_0465)*(1+s_0514/kmp_s_0514r_0465)*(1+s_1044/kmp_s_1044r_0465)*(1+s_1091/kmp_s_1091r_0465)*(1+s_1434_b/kmp_s_1434_br_0465))-1)/intracellular
kms_s_0069r_0485=0.549; Keq_r_0485=0.6039; Vmax_r_0485=2.08449; kmp_s_0692r_0485=0.549; kmp_s_1434_br_0485=0.549Reaction: s_0069 => s_0692 + s_1434_b; s_0069, s_0692, s_1434_b, Rate Law: intracellular*Vmax_r_0485*(1/kms_s_0069r_0485)^1*(s_0069^1-s_0692^1*s_1434_b^1/Keq_r_0485)/((1+s_0069/kms_s_0069r_0485+(1+s_0692/kmp_s_0692r_0485)*(1+s_1434_b/kmp_s_1434_br_0485))-1)/intracellular
Vmax_r_0509=38.2031; kmp_s_0899r_0509=0.549; kms_s_0763_br_0509=0.549; kms_s_1096r_0509=0.549; kmp_s_1091r_0509=0.549; kms_s_0185r_0509=0.549; Keq_r_0509=2.00364; kmp_s_1434_br_0509=0.549; kms_s_0430r_0509=0.549Reaction: s_0185 + s_0430 + s_0763_b + s_1096 => s_0899 + s_1091 + s_1434_b; s_0185, s_0430, s_0763_b, s_0899, s_1091, s_1096, s_1434_b, Rate Law: intracellular*Vmax_r_0509*(1/kms_s_0185r_0509)^1*(1/kms_s_0430r_0509)^1*(1/kms_s_0763_br_0509)^1*(1/kms_s_1096r_0509)^1*(s_0185^1*s_0430^1*s_0763_b^1*s_1096^1-s_0899^1*s_1091^1*s_1434_b^1/Keq_r_0509)/(((1+s_0185/kms_s_0185r_0509)*(1+s_0430/kms_s_0430r_0509)*(1+s_0763_b/kms_s_0763_br_0509)*(1+s_1096/kms_s_1096r_0509)+(1+s_0899/kmp_s_0899r_0509)*(1+s_1091/kmp_s_1091r_0509)*(1+s_1434_b/kmp_s_1434_br_0509))-1)/intracellular
kms_s_0635r_0995=0.549; kmp_s_1434_br_0995=0.549; Keq_r_0995=1.1; Vmax_r_0995=0.0034727; kms_s_0663r_0995=0.549; kmp_s_0641r_0995=0.549Reaction: s_0635 + s_0663 => s_0641 + s_1434_b; s_0635, s_0641, s_0663, s_1434_b, Rate Law: intracellular*Vmax_r_0995*(1/kms_s_0635r_0995)^1*(1/kms_s_0663r_0995)^1*(s_0635^1*s_0663^1-s_0641^1*s_1434_b^1/Keq_r_0995)/(((1+s_0635/kms_s_0635r_0995)*(1+s_0663/kms_s_0663r_0995)+(1+s_0641/kmp_s_0641r_0995)*(1+s_1434_b/kmp_s_1434_br_0995))-1)/intracellular
kmp_s_0530r_0351=0.549; kms_s_1087r_0351=0.0867353; Vmax_r_0351=3.30331; Keq_r_0351=34.7263; kms_s_0763_br_0351=0.549; kmp_s_1082r_0351=1.50326; kms_s_0529r_0351=0.549Reaction: s_0529 + s_0763_b + s_1087 => s_0530 + s_1082; s_0529, s_0530, s_0763_b, s_1082, s_1087, Rate Law: intracellular*Vmax_r_0351*(1/kms_s_0529r_0351)^1*(1/kms_s_0763_br_0351)^1*(1/kms_s_1087r_0351)^1*(s_0529^1*s_0763_b^1*s_1087^1-s_0530^1*s_1082^1/Keq_r_0351)/(((1+s_0529/kms_s_0529r_0351)*(1+s_0763_b/kms_s_0763_br_0351)*(1+s_1087/kms_s_1087r_0351)+(1+s_0530/kmp_s_0530r_0351)*(1+s_1082/kmp_s_1082r_0351))-1)/intracellular
kms_s_0446r_0891=1.09208; kmp_s_0331r_0891=0.549; kms_s_0427r_0891=0.549; Keq_r_0891=0.696514; kmp_s_0434r_0891=1.25956; Vmax_r_0891=2.25059; kmp_s_0763_br_0891=0.549Reaction: s_0427 + s_0446 => s_0331 + s_0434 + s_0763_b; s_0331, s_0427, s_0434, s_0446, s_0763_b, Rate Law: intracellular*Vmax_r_0891*(1/kms_s_0427r_0891)^1*(1/kms_s_0446r_0891)^1*(s_0427^1*s_0446^1-s_0331^1*s_0434^1*s_0763_b^1/Keq_r_0891)/(((1+s_0427/kms_s_0427r_0891)*(1+s_0446/kms_s_0446r_0891)+(1+s_0331/kmp_s_0331r_0891)*(1+s_0434/kmp_s_0434r_0891)*(1+s_0763_b/kmp_s_0763_br_0891))-1)/intracellular
Keq_r_0031=2.00364; kmp_s_0297r_0031=0.549; kmp_s_0470r_0031=1.0; kms_s_0010r_0031=0.549; Vmax_r_0031=1.0703; kms_s_0763_br_0031=0.549Reaction: s_0010 + s_0763_b => s_0297 + s_0470; s_0010, s_0297, s_0470, s_0763_b, Rate Law: intracellular*Vmax_r_0031*(1/kms_s_0010r_0031)^1*(1/kms_s_0763_br_0031)^1*(s_0010^1*s_0763_b^1-s_0297^1*s_0470^1/Keq_r_0031)/(((1+s_0010/kms_s_0010r_0031)*(1+s_0763_b/kms_s_0763_br_0031)+(1+s_0297/kmp_s_0297r_0031)*(1+s_0470/kmp_s_0470r_0031))-1)/intracellular
kmp_s_0079r_0881=0.549; Keq_r_0881=2.00364; kms_s_1434_br_0881=0.549; Vmax_r_0881=0.229351; kms_s_0080r_0881=0.549Reaction: s_0080 + s_1434_b => s_0079; s_0079, s_0080, s_1434_b, Rate Law: intracellular*Vmax_r_0881*(1/kms_s_0080r_0881)^1*(1/kms_s_1434_br_0881)^1*(s_0080^1*s_1434_b^1-s_0079^1/Keq_r_0881)/(((1+s_0080/kms_s_0080r_0881)*(1+s_1434_b/kms_s_1434_br_0881)+1+s_0079/kmp_s_0079r_0881)-1)/intracellular
kms_s_0315r_0604=0.549; kms_s_0907r_0604=0.549; kmp_s_0763_br_0604=0.549; Vmax_r_0604=0.871524; kmp_s_0899r_0604=0.549; kmp_s_0317r_0604=0.549; Keq_r_0604=0.331541; kmp_s_0532r_0604=0.549Reaction: s_0315 + s_0907 => s_0317 + s_0532 + s_0763_b + s_0899; s_0315, s_0317, s_0532, s_0763_b, s_0899, s_0907, Rate Law: intracellular*Vmax_r_0604*(1/kms_s_0315r_0604)^1*(1/kms_s_0907r_0604)^1*(s_0315^1*s_0907^1-s_0317^1*s_0532^1*s_0763_b^1*s_0899^1/Keq_r_0604)/(((1+s_0315/kms_s_0315r_0604)*(1+s_0907/kms_s_0907r_0604)+(1+s_0317/kmp_s_0317r_0604)*(1+s_0532/kmp_s_0532r_0604)*(1+s_0763_b/kmp_s_0763_br_0604)*(1+s_0899/kmp_s_0899r_0604))-1)/intracellular
kmp_s_0564r_0360=0.549; kmp_s_0446r_0360=1.09208; Keq_r_0360=0.698801; Vmax_r_0360=0.015323; kms_s_0400r_0360=1.71907; kms_s_0562r_0360=0.549Reaction: s_0400 + s_0562 => s_0446 + s_0564; s_0400, s_0446, s_0562, s_0564, Rate Law: intracellular*Vmax_r_0360*(1/kms_s_0400r_0360)^1*(1/kms_s_0562r_0360)^1*(s_0400^1*s_0562^1-s_0446^1*s_0564^1/Keq_r_0360)/(((1+s_0400/kms_s_0400r_0360)*(1+s_0562/kms_s_0562r_0360)+(1+s_0446/kmp_s_0446r_0360)*(1+s_0564/kmp_s_0564r_0360))-1)/intracellular
Keq_r_0064=0.0348439; kms_s_1082r_0064=1.50326; Vmax_r_0064=1.68189; kmp_s_0763_br_0064=0.549; kmp_s_1087r_0064=0.0867353; kms_s_0008r_0064=0.549; kmp_s_0010r_0064=0.549Reaction: s_0008 + s_1082 => s_0010 + s_0763_b + s_1087; s_0008, s_0010, s_0763_b, s_1082, s_1087, Rate Law: intracellular*Vmax_r_0064*(1/kms_s_0008r_0064)^1*(1/kms_s_1082r_0064)^1*(s_0008^1*s_1082^1-s_0010^1*s_0763_b^1*s_1087^1/Keq_r_0064)/(((1+s_0008/kms_s_0008r_0064)*(1+s_1082/kms_s_1082r_0064)+(1+s_0010/kmp_s_0010r_0064)*(1+s_0763_b/kmp_s_0763_br_0064)*(1+s_1087/kmp_s_1087r_0064))-1)/intracellular
kmp_s_0763_br_0856=0.549; kmp_s_1517r_0856=0.549; kms_s_1521r_0856=0.549; Vmax_r_0856=1.07843; Keq_r_0856=0.182016; kmp_s_1349r_0856=0.549; kmp_s_0397r_0856=0.549; kms_s_0206r_0856=0.549Reaction: s_0206 + s_1521 => s_0397 + s_0763_b + s_1349 + s_1517; s_0206, s_0397, s_0763_b, s_1349, s_1517, s_1521, Rate Law: intracellular*Vmax_r_0856*(1/kms_s_0206r_0856)^1*(1/kms_s_1521r_0856)^1*(s_0206^1*s_1521^1-s_0397^1*s_0763_b^2*s_1349^1*s_1517^1/Keq_r_0856)/(((1+s_0206/kms_s_0206r_0856)*(1+s_1521/kms_s_1521r_0856)+(1+s_0397/kmp_s_0397r_0856)*(1+s_0763_b/kmp_s_0763_br_0856)*(1+s_1349/kmp_s_1349r_0856)*(1+s_1517/kmp_s_1517r_0856))-1)/intracellular
kms_s_0539r_1035=0.104555; Vmax_r_1035=0.14014; kms_s_0533r_1035=0.549; kmp_s_0731r_1035=0.0436363; kmp_s_1304r_1035=0.549; Keq_r_1035=0.459088Reaction: s_0533 + s_0539 => s_0731 + s_1304; s_0533, s_0539, s_0731, s_1304, Rate Law: intracellular*Vmax_r_1035*(1/kms_s_0533r_1035)^1*(1/kms_s_0539r_1035)^1*(s_0533^1*s_0539^1-s_0731^1*s_1304^1/Keq_r_1035)/(((1+s_0533/kms_s_0533r_1035)*(1+s_0539/kms_s_0539r_1035)+(1+s_0731/kmp_s_0731r_1035)*(1+s_1304/kmp_s_1304r_1035))-1)/intracellular
kms_s_0561r_0965=0.549; Keq_r_0965=1.1; Vmax_r_0965=0.5577; kmp_s_0557r_0965=0.549Reaction: s_0561 => s_0557; s_0557, s_0561, Rate Law: intracellular*Vmax_r_0965*(1/kms_s_0561r_0965)^1*(s_0561^1-s_0557^1/Keq_r_0965)/((1+s_0561/kms_s_0561r_0965+1+s_0557/kmp_s_0557r_0965)-1)/intracellular
kmp_s_0386r_1672=0.549; Keq_r_1672=1.1; Vmax_r_1672=0.026268; kms_s_1342r_1672=0.549Reaction: s_1342 => s_0386; s_0386, s_1342, Rate Law: intracellular*Vmax_r_1672*(1/kms_s_1342r_1672)^1*(s_1342^1-s_0386^1/Keq_r_1672)/((1+s_1342/kms_s_1342r_1672+1+s_0386/kmp_s_0386r_1672)-1)/intracellular
kmp_s_0400r_0573=1.71907; Vmax_r_0573=1.99579; kms_s_0545r_0573=0.0987587; kmp_s_0410r_0573=0.549; Keq_r_0573=2000.0; kms_s_0446r_0573=1.09208; kmp_s_0763_br_0573=0.549Reaction: s_0446 + s_0545 => s_0400 + s_0410 + s_0763_b; s_0400, s_0410, s_0446, s_0545, s_0763_b, Rate Law: intracellular*Vmax_r_0573*(1/kms_s_0446r_0573)^1*(1/kms_s_0545r_0573)^1*(s_0446^1*s_0545^1-s_0400^1*s_0410^1*s_0763_b^1/Keq_r_0573)/(((1+s_0446/kms_s_0446r_0573)*(1+s_0545/kms_s_0545r_0573)+(1+s_0400/kmp_s_0400r_0573)*(1+s_0410/kmp_s_0410r_0573)*(1+s_0763_b/kmp_s_0763_br_0573))-1)/intracellular
kmp_s_0763_br_0526=0.549; Keq_r_0526=2.21027; kmp_s_1096r_0526=0.549; kms_s_1091r_0526=0.549; Vmax_r_0526=5.48128; kmp_s_0734r_0526=0.549; kms_s_0732r_0526=0.15Reaction: s_0732 + s_1091 => s_0734 + s_0763_b + s_1096; s_0732, s_0734, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0526*(1/kms_s_0732r_0526)^1*(1/kms_s_1091r_0526)^1*(s_0732^1*s_1091^1-s_0734^1*s_0763_b^1*s_1096^1/Keq_r_0526)/(((1+s_0732/kms_s_0732r_0526)*(1+s_1091/kms_s_1091r_0526)+(1+s_0734/kmp_s_0734r_0526)*(1+s_0763_b/kmp_s_0763_br_0526)*(1+s_1096/kmp_s_1096r_0526))-1)/intracellular
kmp_s_0533r_1037=0.549; Keq_r_1037=72.6682; kmp_s_0561r_1037=0.549; kms_s_0731r_1037=0.0436363; Vmax_r_1037=1.1627; kms_s_0539r_1037=0.104555Reaction: s_0539 + s_0731 => s_0533 + s_0561; s_0533, s_0539, s_0561, s_0731, Rate Law: intracellular*Vmax_r_1037*(1/kms_s_0539r_1037)^1*(1/kms_s_0731r_1037)^1*(s_0539^1*s_0731^1-s_0533^1*s_0561^1/Keq_r_1037)/(((1+s_0539/kms_s_0539r_1037)*(1+s_0731/kms_s_0731r_1037)+(1+s_0533/kmp_s_0533r_1037)*(1+s_0561/kmp_s_0561r_1037))-1)/intracellular
kms_s_0755r_0170=0.549; kms_s_0816r_0170=0.549; Keq_r_0170=0.331541; kmp_s_1053r_0170=0.549; kmp_s_0706r_0170=0.549; kmp_s_1207r_0170=0.549; kmp_s_0763_br_0170=0.549; kms_s_0881r_0170=0.549; Vmax_r_0170=1.8216Reaction: s_0755 + s_0816 + s_0881 => s_0706 + s_0763_b + s_1053 + s_1207; s_0706, s_0755, s_0763_b, s_0816, s_0881, s_1053, s_1207, Rate Law: intracellular*Vmax_r_0170*(1/kms_s_0755r_0170)^1*(1/kms_s_0816r_0170)^1*(1/kms_s_0881r_0170)^1*(s_0755^1*s_0816^1*s_0881^1-s_0706^1*s_0763_b^2*s_1053^1*s_1207^1/Keq_r_0170)/(((1+s_0755/kms_s_0755r_0170)*(1+s_0816/kms_s_0816r_0170)*(1+s_0881/kms_s_0881r_0170)+(1+s_0706/kmp_s_0706r_0170)*(1+s_0763_b/kmp_s_0763_br_0170)*(1+s_1053/kmp_s_1053r_0170)*(1+s_1207/kmp_s_1207r_0170))-1)/intracellular
kmp_s_0317r_0169=0.549; kms_s_0009r_0169=0.549; Vmax_r_0169=0.333848; Keq_r_0169=0.6039; kmp_s_0692r_0169=0.549Reaction: s_0009 => s_0317 + s_0692; s_0009, s_0317, s_0692, Rate Law: intracellular*Vmax_r_0169*(1/kms_s_0009r_0169)^1*(s_0009^1-s_0317^1*s_0692^1/Keq_r_0169)/((1+s_0009/kms_s_0009r_0169+(1+s_0317/kmp_s_0317r_0169)*(1+s_0692/kmp_s_0692r_0169))-1)/intracellular
Vmax_r_0936=0.863944; kmp_s_1091r_0936=0.549; kms_s_0763_br_0936=0.549; Keq_r_0936=3.64962; kms_s_0120r_0936=0.549; kmp_s_0939r_0936=0.549; kms_s_1096r_0936=0.549Reaction: s_0120 + s_0763_b + s_1096 => s_0939 + s_1091; s_0120, s_0763_b, s_0939, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0936*(1/kms_s_0120r_0936)^1*(1/kms_s_0763_br_0936)^2*(1/kms_s_1096r_0936)^1*(s_0120^1*s_0763_b^2*s_1096^1-s_0939^1*s_1091^1/Keq_r_0936)/(((1+s_0120/kms_s_0120r_0936)*(1+s_0763_b/kms_s_0763_br_0936)*(1+s_1096/kms_s_1096r_0936)+(1+s_0939/kmp_s_0939r_0936)*(1+s_1091/kmp_s_1091r_0936))-1)/intracellular
kms_s_0514r_0437=0.549; kmp_s_0434r_0437=1.25956; Keq_r_0437=1.26869; kmp_s_1355r_0437=0.549; kms_s_0987r_0437=0.549; Vmax_r_0437=0.0038115; kms_s_0446r_0437=1.09208; kmp_s_0605r_0437=0.549Reaction: s_0446 + s_0514 + s_0987 => s_0434 + s_0605 + s_1355; s_0434, s_0446, s_0514, s_0605, s_0987, s_1355, Rate Law: intracellular*Vmax_r_0437*(1/kms_s_0446r_0437)^1*(1/kms_s_0514r_0437)^1*(1/kms_s_0987r_0437)^1*(s_0446^1*s_0514^1*s_0987^1-s_0434^1*s_0605^1*s_1355^1/Keq_r_0437)/(((1+s_0446/kms_s_0446r_0437)*(1+s_0514/kms_s_0514r_0437)*(1+s_0987/kms_s_0987r_0437)+(1+s_0434/kmp_s_0434r_0437)*(1+s_0605/kmp_s_0605r_0437)*(1+s_1355/kmp_s_1355r_0437))-1)/intracellular
Vmax_r_0875=1.5048; Keq_r_0875=1.1; kms_s_0554r_0875=0.549; kmp_s_0553r_0875=0.549Reaction: s_0554 => s_0553; s_0553, s_0554, Rate Law: intracellular*Vmax_r_0875*(1/kms_s_0554r_0875)^1*(s_0554^1-s_0553^1/Keq_r_0875)/((1+s_0554/kms_s_0554r_0875+1+s_0553/kmp_s_0553r_0875)-1)/intracellular
Keq_r_0725=1.1; kmp_s_1207r_0725=0.549; kms_s_1434_br_0725=0.549; kms_s_0128r_0725=0.549; Vmax_r_0725=0.006545; kmp_s_1020r_0725=0.549Reaction: s_0128 + s_1434_b => s_1020 + s_1207; s_0128, s_1020, s_1207, s_1434_b, Rate Law: intracellular*Vmax_r_0725*(1/kms_s_0128r_0725)^1*(1/kms_s_1434_br_0725)^1*(s_0128^1*s_1434_b^1-s_1020^1*s_1207^1/Keq_r_0725)/(((1+s_0128/kms_s_0128r_0725)*(1+s_1434_b/kms_s_1434_br_0725)+(1+s_1020/kmp_s_1020r_0725)*(1+s_1207/kmp_s_1207r_0725))-1)/intracellular
kms_s_0446r_0130=1.09208; Vmax_r_0130=0.58058; kmp_s_1070r_0130=0.549; Keq_r_0130=1.73154; kmp_s_0400r_0130=1.71907; kms_s_1071r_0130=0.549Reaction: s_0446 + s_1071 => s_0400 + s_1070; s_0400, s_0446, s_1070, s_1071, Rate Law: intracellular*Vmax_r_0130*(1/kms_s_0446r_0130)^1*(1/kms_s_1071r_0130)^1*(s_0446^1*s_1071^1-s_0400^1*s_1070^1/Keq_r_0130)/(((1+s_0446/kms_s_0446r_0130)*(1+s_1071/kms_s_1071r_0130)+(1+s_0400/kmp_s_0400r_0130)*(1+s_1070/kmp_s_1070r_0130))-1)/intracellular
Vmax_r_0225=0.414697; kms_s_0017r_0225=0.549; kmp_s_0692r_0225=0.549; Keq_r_0225=0.6039; kmp_s_0873r_0225=0.549Reaction: s_0017 => s_0692 + s_0873; s_0017, s_0692, s_0873, Rate Law: intracellular*Vmax_r_0225*(1/kms_s_0017r_0225)^1*(s_0017^1-s_0692^1*s_0873^1/Keq_r_0225)/((1+s_0017/kms_s_0017r_0225+(1+s_0692/kmp_s_0692r_0225)*(1+s_0873/kmp_s_0873r_0225))-1)/intracellular
kmp_s_1207r_0728=0.549; kmp_s_0149r_0728=0.549; kms_s_1070r_0728=0.549; kms_s_0763_br_0728=0.549; Vmax_r_0728=1.2441; Keq_r_0728=1.1; kms_s_1096r_0728=0.549; kmp_s_1091r_0728=0.549Reaction: s_0763_b + s_1070 + s_1096 => s_0149 + s_1091 + s_1207; s_0149, s_0763_b, s_1070, s_1091, s_1096, s_1207, Rate Law: intracellular*Vmax_r_0728*(1/kms_s_0763_br_0728)^1*(1/kms_s_1070r_0728)^1*(1/kms_s_1096r_0728)^1*(s_0763_b^1*s_1070^1*s_1096^1-s_0149^1*s_1091^1*s_1207^1/Keq_r_0728)/(((1+s_0763_b/kms_s_0763_br_0728)*(1+s_1070/kms_s_1070r_0728)*(1+s_1096/kms_s_1096r_0728)+(1+s_0149/kmp_s_0149r_0728)*(1+s_1091/kmp_s_1091r_0728)*(1+s_1207/kmp_s_1207r_0728))-1)/intracellular
kmp_s_0514r_0534=0.549; kms_s_0386r_0534=0.549; kms_s_1315r_0534=12.8511; Vmax_r_0534=0.0421077; kmp_s_0763_br_0534=0.549; Keq_r_0534=0.0141635; kmp_s_0083r_0534=0.549Reaction: s_0386 + s_1315 => s_0083 + s_0514 + s_0763_b; s_0083, s_0386, s_0514, s_0763_b, s_1315, Rate Law: intracellular*Vmax_r_0534*(1/kms_s_0386r_0534)^1*(1/kms_s_1315r_0534)^1*(s_0386^1*s_1315^1-s_0083^1*s_0514^1*s_0763_b^2/Keq_r_0534)/(((1+s_0386/kms_s_0386r_0534)*(1+s_1315/kms_s_1315r_0534)+(1+s_0083/kmp_s_0083r_0534)*(1+s_0514/kmp_s_0514r_0534)*(1+s_0763_b/kmp_s_0763_br_0534))-1)/intracellular
kms_s_0816r_0607=0.549; kms_s_1434_br_0607=0.549; Vmax_r_0607=0.501598; Keq_r_0607=0.063468; kmp_s_0306r_0607=0.549; kmp_s_1087r_0607=0.0867353; kms_s_1082r_0607=1.50326; kmp_s_0763_br_0607=0.549Reaction: s_0816 + s_1082 + s_1434_b => s_0306 + s_0763_b + s_1087; s_0306, s_0763_b, s_0816, s_1082, s_1087, s_1434_b, Rate Law: intracellular*Vmax_r_0607*(1/kms_s_0816r_0607)^1*(1/kms_s_1082r_0607)^1*(1/kms_s_1434_br_0607)^1*(s_0816^1*s_1082^1*s_1434_b^1-s_0306^1*s_0763_b^1*s_1087^1/Keq_r_0607)/(((1+s_0816/kms_s_0816r_0607)*(1+s_1082/kms_s_1082r_0607)*(1+s_1434_b/kms_s_1434_br_0607)+(1+s_0306/kmp_s_0306r_0607)*(1+s_0763_b/kmp_s_0763_br_0607)*(1+s_1087/kmp_s_1087r_0607))-1)/intracellular
kms_s_0079r_0008=0.549; kmp_s_0315r_0008=0.549; Vmax_r_0008=0.13761; Keq_r_0008=1.1Reaction: s_0079 => s_0315; s_0079, s_0315, Rate Law: intracellular*Vmax_r_0008*(1/kms_s_0079r_0008)^1*(s_0079^1-s_0315^1/Keq_r_0008)/((1+s_0079/kms_s_0079r_0008+1+s_0315/kmp_s_0315r_0008)-1)/intracellular
kmp_s_1347r_1507=0.549; Vmax_r_1507=0.0190579; kms_s_1348_br_1507=42.2; Keq_r_1507=1.0Reaction: s_1348_b => s_1347; s_1347, s_1348_b, Rate Law: Vmax_r_1507*(1/kms_s_1348_br_1507)^1*(s_1348_b^1-s_1347^1/Keq_r_1507)/((1+s_1348_b/kms_s_1348_br_1507+1+s_1347/kmp_s_1347r_1507)-1)
Keq_r_0165=0.805968; kmp_s_0434r_0165=1.25956; kmp_s_0755r_0165=0.549; Vmax_r_0165=4.0656; kms_s_0706r_0165=0.549; kms_s_0400r_0165=1.71907Reaction: s_0400 + s_0706 => s_0434 + s_0755; s_0400, s_0434, s_0706, s_0755, Rate Law: intracellular*Vmax_r_0165*(1/kms_s_0400r_0165)^1*(1/kms_s_0706r_0165)^1*(s_0400^1*s_0706^1-s_0434^1*s_0755^1/Keq_r_0165)/(((1+s_0400/kms_s_0400r_0165)*(1+s_0706/kms_s_0706r_0165)+(1+s_0434/kmp_s_0434r_0165)*(1+s_0755/kmp_s_0755r_0165))-1)/intracellular
kmp_s_0309r_0889=0.549; kmp_s_1052r_0889=0.549; kms_s_0122r_0889=0.549; kmp_s_0763_br_0889=0.549; Vmax_r_0889=0.734467; kms_s_1048r_0889=0.549; Keq_r_0889=0.6039Reaction: s_0122 + s_1048 => s_0309 + s_0763_b + s_1052; s_0122, s_0309, s_0763_b, s_1048, s_1052, Rate Law: intracellular*Vmax_r_0889*(1/kms_s_0122r_0889)^1*(1/kms_s_1048r_0889)^1*(s_0122^1*s_1048^1-s_0309^1*s_0763_b^1*s_1052^1/Keq_r_0889)/(((1+s_0122/kms_s_0122r_0889)*(1+s_1048/kms_s_1048r_0889)+(1+s_0309/kmp_s_0309r_0889)*(1+s_0763_b/kmp_s_0763_br_0889)*(1+s_1052/kmp_s_1052r_0889))-1)/intracellular
kms_s_0446r_0123=1.09208; kmp_s_0400r_0123=1.71907; kmp_s_0763_br_0123=0.549; kmp_s_1207r_0123=0.549; Keq_r_0123=0.950614; Vmax_r_0123=0.105501; kmp_s_1005r_0123=0.549; kms_s_0458r_0123=0.549; kms_s_0380r_0123=0.549Reaction: s_0380 + s_0446 + s_0458 => s_0400 + s_0763_b + s_1005 + s_1207; s_0380, s_0400, s_0446, s_0458, s_0763_b, s_1005, s_1207, Rate Law: intracellular*Vmax_r_0123*(1/kms_s_0380r_0123)^1*(1/kms_s_0446r_0123)^1*(1/kms_s_0458r_0123)^1*(s_0380^1*s_0446^1*s_0458^1-s_0400^1*s_0763_b^1*s_1005^1*s_1207^1/Keq_r_0123)/(((1+s_0380/kms_s_0380r_0123)*(1+s_0446/kms_s_0446r_0123)*(1+s_0458/kms_s_0458r_0123)+(1+s_0400/kmp_s_0400r_0123)*(1+s_0763_b/kmp_s_0763_br_0123)*(1+s_1005/kmp_s_1005r_0123)*(1+s_1207/kmp_s_1207r_0123))-1)/intracellular
Keq_r_0213=0.6039; kmp_s_0763_br_0213=0.549; kms_s_0410r_0213=0.549; Vmax_r_0213=0.174824; kmp_s_0419r_0213=0.549; kmp_s_1411r_0213=0.549; kmI_s_1415rm_0213=6.0; kms_s_1415r_0213=0.549Reaction: s_0410 + s_1415 => s_0419 + s_0763_b + s_1411; s_1415, s_0410, s_1415, s_0419, s_0763_b, s_1411, Rate Law: intracellular*Vmax_r_0213*(1/kms_s_0410r_0213)^1*(1/kms_s_1415r_0213)^1*(s_0410^1*s_1415^1-s_0419^1*s_0763_b^1*s_1411^1/Keq_r_0213)/(((1+s_0410/kms_s_0410r_0213)*(1+s_1415/kms_s_1415r_0213)+(1+s_0419/kmp_s_0419r_0213)*(1+s_0763_b/kmp_s_0763_br_0213)*(1+s_1411/kmp_s_1411r_0213)+1+s_1415/kmI_s_1415rm_0213)-1)/intracellular
kms_s_1349r_1008=0.549; Keq_r_1008=3.64962; kms_s_0763_br_1008=0.549; kmp_s_1434_br_1008=0.549; kms_s_1096r_1008=0.549; Vmax_r_1008=0.851402; kmp_s_0805r_1008=0.549; kmp_s_1091r_1008=0.549Reaction: s_0763_b + s_1096 + s_1349 => s_0805 + s_1091 + s_1434_b; s_0763_b, s_0805, s_1091, s_1096, s_1349, s_1434_b, Rate Law: intracellular*Vmax_r_1008*(1/kms_s_0763_br_1008)^5*(1/kms_s_1096r_1008)^3*(1/kms_s_1349r_1008)^1*(s_0763_b^5*s_1096^3*s_1349^1-s_0805^1*s_1091^3*s_1434_b^3/Keq_r_1008)/(((1+s_0763_b/kms_s_0763_br_1008)*(1+s_1096/kms_s_1096r_1008)*(1+s_1349/kms_s_1349r_1008)+(1+s_0805/kmp_s_0805r_1008)*(1+s_1091/kmp_s_1091r_1008)*(1+s_1434_b/kmp_s_1434_br_1008))-1)/intracellular
kmp_s_0316r_0884=0.549; Keq_r_0884=0.286516; kmp_s_0763_br_0884=0.549; kmp_s_1207r_0884=0.549; kmp_s_0400r_0884=1.71907; kms_s_0446r_0884=1.09208; Vmax_r_0884=1.26862; kms_s_0158r_0884=0.549Reaction: s_0158 + s_0446 => s_0316 + s_0400 + s_0763_b + s_1207; s_0158, s_0316, s_0400, s_0446, s_0763_b, s_1207, Rate Law: intracellular*Vmax_r_0884*(1/kms_s_0158r_0884)^1*(1/kms_s_0446r_0884)^1*(s_0158^1*s_0446^1-s_0316^1*s_0400^1*s_0763_b^2*s_1207^1/Keq_r_0884)/(((1+s_0158/kms_s_0158r_0884)*(1+s_0446/kms_s_0446r_0884)+(1+s_0316/kmp_s_0316r_0884)*(1+s_0400/kmp_s_0400r_0884)*(1+s_0763_b/kmp_s_0763_br_0884)*(1+s_1207/kmp_s_1207r_0884))-1)/intracellular
kmp_s_1342r_1003=0.549; kms_s_0514r_1003=0.549; kmp_s_1207r_1003=0.549; Keq_r_1003=1.73154; kms_s_0446r_1003=1.09208; Vmax_r_1003=0.13134; kms_s_1338r_1003=0.549; kmp_s_0400r_1003=1.71907Reaction: s_0446 + s_0514 + s_1338 => s_0400 + s_1207 + s_1342; s_0400, s_0446, s_0514, s_1207, s_1338, s_1342, Rate Law: intracellular*Vmax_r_1003*(1/kms_s_0446r_1003)^1*(1/kms_s_0514r_1003)^1*(1/kms_s_1338r_1003)^1*(s_0446^1*s_0514^1*s_1338^1-s_0400^1*s_1207^1*s_1342^1/Keq_r_1003)/(((1+s_0446/kms_s_0446r_1003)*(1+s_0514/kms_s_0514r_1003)*(1+s_1338/kms_s_1338r_1003)+(1+s_0400/kmp_s_0400r_1003)*(1+s_1207/kmp_s_1207r_1003)*(1+s_1342/kmp_s_1342r_1003))-1)/intracellular
kms_s_1156r_0688=0.549; kms_s_0763_br_0688=0.549; kmp_s_0069r_0688=0.549; Vmax_r_0688=4.58593; kmp_s_1082r_0688=1.50326; kms_s_1087r_0688=0.0867353; Keq_r_0688=34.7263Reaction: s_0763_b + s_1087 + s_1156 => s_0069 + s_1082; s_0069, s_0763_b, s_1082, s_1087, s_1156, Rate Law: intracellular*Vmax_r_0688*(1/kms_s_0763_br_0688)^1*(1/kms_s_1087r_0688)^1*(1/kms_s_1156r_0688)^1*(s_0763_b^1*s_1087^1*s_1156^1-s_0069^1*s_1082^1/Keq_r_0688)/(((1+s_0763_b/kms_s_0763_br_0688)*(1+s_1087/kms_s_1087r_0688)*(1+s_1156/kms_s_1156r_0688)+(1+s_0069/kmp_s_0069r_0688)*(1+s_1082/kmp_s_1082r_0688))-1)/intracellular
kmp_s_0798r_0582=0.549; kms_s_0185r_0582=0.549; Keq_r_0582=1.1; kmp_s_0763_br_0582=0.549; Vmax_r_0582=2.1945; kmp_s_0514r_0582=0.549; kms_s_1434_br_0582=0.549; kms_s_0380r_0582=0.549Reaction: s_0185 + s_0380 + s_1434_b => s_0514 + s_0763_b + s_0798; s_0185, s_0380, s_0514, s_0763_b, s_0798, s_1434_b, Rate Law: intracellular*Vmax_r_0582*(1/kms_s_0185r_0582)^1*(1/kms_s_0380r_0582)^1*(1/kms_s_1434_br_0582)^1*(s_0185^1*s_0380^1*s_1434_b^1-s_0514^1*s_0763_b^1*s_0798^1/Keq_r_0582)/(((1+s_0185/kms_s_0185r_0582)*(1+s_0380/kms_s_0380r_0582)*(1+s_1434_b/kms_s_1434_br_0582)+(1+s_0514/kmp_s_0514r_0582)*(1+s_0763_b/kmp_s_0763_br_0582)*(1+s_0798/kmp_s_0798r_0582))-1)/intracellular
kmp_s_0434r_0551=1.25956; kmp_s_0899r_0551=0.549; kmp_s_0752r_0551=0.549; kmp_s_0605r_0551=0.549; kms_s_0907r_0551=0.549; kms_s_1434_br_0551=0.549; Keq_r_0551=0.382386; kms_s_0446r_0551=1.09208; kms_s_0306r_0551=0.549; kmp_s_0763_br_0551=0.549; Vmax_r_0551=1.57168Reaction: s_0306 + s_0446 + s_0907 + s_1434_b => s_0434 + s_0605 + s_0752 + s_0763_b + s_0899; s_0306, s_0434, s_0446, s_0605, s_0752, s_0763_b, s_0899, s_0907, s_1434_b, Rate Law: intracellular*Vmax_r_0551*(1/kms_s_0306r_0551)^1*(1/kms_s_0446r_0551)^1*(1/kms_s_0907r_0551)^1*(1/kms_s_1434_br_0551)^1*(s_0306^1*s_0446^1*s_0907^1*s_1434_b^1-s_0434^1*s_0605^1*s_0752^1*s_0763_b^2*s_0899^1/Keq_r_0551)/(((1+s_0306/kms_s_0306r_0551)*(1+s_0446/kms_s_0446r_0551)*(1+s_0907/kms_s_0907r_0551)*(1+s_1434_b/kms_s_1434_br_0551)+(1+s_0434/kmp_s_0434r_0551)*(1+s_0605/kmp_s_0605r_0551)*(1+s_0752/kmp_s_0752r_0551)*(1+s_0763_b/kmp_s_0763_br_0551)*(1+s_0899/kmp_s_0899r_0551))-1)/intracellular
kms_s_1434_br_0699=0.549; kmp_s_0122r_0699=0.549; Keq_r_0699=1.1; Vmax_r_0699=1.2166; kms_s_0015r_0699=0.549; kmp_s_0763_br_0699=0.549Reaction: s_0015 + s_1434_b => s_0122 + s_0763_b; s_0015, s_0122, s_0763_b, s_1434_b, Rate Law: intracellular*Vmax_r_0699*(1/kms_s_0015r_0699)^1*(1/kms_s_1434_br_0699)^1*(s_0015^1*s_1434_b^1-s_0122^1*s_0763_b^1/Keq_r_0699)/(((1+s_0015/kms_s_0015r_0699)*(1+s_1434_b/kms_s_1434_br_0699)+(1+s_0122/kmp_s_0122r_0699)*(1+s_0763_b/kmp_s_0763_br_0699))-1)/intracellular
kms_s_0317r_0885=0.549; kmp_s_0325r_0885=0.549; kms_s_0122r_0885=0.549; Keq_r_0885=1.1; kmp_s_0309r_0885=0.549; Vmax_r_0885=0.7854Reaction: s_0122 + s_0317 => s_0309 + s_0325; s_0122, s_0309, s_0317, s_0325, Rate Law: intracellular*Vmax_r_0885*(1/kms_s_0122r_0885)^1*(1/kms_s_0317r_0885)^1*(s_0122^1*s_0317^1-s_0309^1*s_0325^1/Keq_r_0885)/(((1+s_0122/kms_s_0122r_0885)*(1+s_0317/kms_s_0317r_0885)+(1+s_0309/kmp_s_0309r_0885)*(1+s_0325/kmp_s_0325r_0885))-1)/intracellular
kmp_s_0867r_0650=0.549; kms_s_0763_br_0650=0.549; kmp_s_0434r_0650=1.25956; Vmax_r_0650=4.53532; kmp_s_0605r_0650=0.549; kms_s_1087r_0650=0.0867353; kmp_s_1082r_0650=1.50326; kms_s_0446r_0650=1.09208; Keq_r_0650=21.9885; kms_s_0861r_0650=0.549Reaction: s_0446 + s_0763_b + s_0861 + s_1087 => s_0434 + s_0605 + s_0867 + s_1082; s_0434, s_0446, s_0605, s_0763_b, s_0861, s_0867, s_1082, s_1087, Rate Law: intracellular*Vmax_r_0650*(1/kms_s_0446r_0650)^1*(1/kms_s_0763_br_0650)^1*(1/kms_s_0861r_0650)^1*(1/kms_s_1087r_0650)^1*(s_0446^1*s_0763_b^1*s_0861^1*s_1087^1-s_0434^1*s_0605^1*s_0867^1*s_1082^1/Keq_r_0650)/(((1+s_0446/kms_s_0446r_0650)*(1+s_0763_b/kms_s_0763_br_0650)*(1+s_0861/kms_s_0861r_0650)*(1+s_1087/kms_s_1087r_0650)+(1+s_0434/kmp_s_0434r_0650)*(1+s_0605/kmp_s_0605r_0650)*(1+s_0867/kmp_s_0867r_0650)*(1+s_1082/kmp_s_1082r_0650))-1)/intracellular
Keq_r_0425=40.2; Vmax_r_0425=0.0118696; kmp_s_0987r_0425=0.549; kmp_s_1091r_0425=0.549; kms_s_1005r_0425=0.549; kms_s_1329r_0425=0.549; kmp_s_0470r_0425=1.0; kmp_s_0514r_0425=0.549; kms_s_0763_br_0425=0.549; kmp_s_1434_br_0425=0.549; kms_s_1096r_0425=0.549Reaction: s_0763_b + s_1005 + s_1096 + s_1329 => s_0470 + s_0514 + s_0987 + s_1091 + s_1434_b; s_0470, s_0514, s_0763_b, s_0987, s_1005, s_1091, s_1096, s_1329, s_1434_b, Rate Law: intracellular*Vmax_r_0425*(1/kms_s_0763_br_0425)^9*(1/kms_s_1005r_0425)^3*(1/kms_s_1096r_0425)^6*(1/kms_s_1329r_0425)^1*(s_0763_b^9*s_1005^3*s_1096^6*s_1329^1-s_0470^3*s_0514^3*s_0987^1*s_1091^6*s_1434_b^3/Keq_r_0425)/(((1+s_0763_b/kms_s_0763_br_0425)*(1+s_1005/kms_s_1005r_0425)*(1+s_1096/kms_s_1096r_0425)*(1+s_1329/kms_s_1329r_0425)+(1+s_0470/kmp_s_0470r_0425)*(1+s_0514/kmp_s_0514r_0425)*(1+s_0987/kmp_s_0987r_0425)*(1+s_1091/kmp_s_1091r_0425)*(1+s_1434_b/kmp_s_1434_br_0425))-1)/intracellular
Vmax_r_0963=0.5544; kms_s_0557r_0963=0.549; kmp_s_0427r_0963=0.549; Keq_r_0963=1.1Reaction: s_0557 => s_0427; s_0427, s_0557, Rate Law: intracellular*Vmax_r_0963*(1/kms_s_0557r_0963)^1*(s_0557^1-s_0427^1/Keq_r_0963)/((1+s_0557/kms_s_0557r_0963+1+s_0427/kmp_s_0427r_0963)-1)/intracellular
Keq_r_0357=0.6039; kms_s_0430r_0357=0.549; kmp_s_0569r_0357=0.549; Vmax_r_0357=0.0163349; kmp_s_1434_br_0357=0.549; kmp_s_0763_br_0357=0.549; kms_s_0624r_0357=0.549Reaction: s_0430 + s_0624 => s_0569 + s_0763_b + s_1434_b; s_0430, s_0569, s_0624, s_0763_b, s_1434_b, Rate Law: intracellular*Vmax_r_0357*(1/kms_s_0430r_0357)^1*(1/kms_s_0624r_0357)^1*(s_0430^1*s_0624^1-s_0569^1*s_0763_b^1*s_1434_b^1/Keq_r_0357)/(((1+s_0430/kms_s_0430r_0357)*(1+s_0624/kms_s_0624r_0357)+(1+s_0569/kmp_s_0569r_0357)*(1+s_0763_b/kmp_s_0763_br_0357)*(1+s_1434_b/kmp_s_1434_br_0357))-1)/intracellular
kmp_s_1140r_0430=0.549; Vmax_r_0430=0.0237906; kmp_s_0470r_0430=1.0; kmp_s_1091r_0430=0.549; kms_s_1005r_0430=0.549; kmp_s_0514r_0430=0.549; Keq_r_0430=40.2; kms_s_0380r_0430=0.549; kmp_s_1434_br_0430=0.549; kms_s_1096r_0430=0.549; kms_s_0763_br_0430=0.549Reaction: s_0380 + s_0763_b + s_1005 + s_1096 => s_0470 + s_0514 + s_1091 + s_1140 + s_1434_b; s_0380, s_0470, s_0514, s_0763_b, s_1005, s_1091, s_1096, s_1140, s_1434_b, Rate Law: intracellular*Vmax_r_0430*(1/kms_s_0380r_0430)^1*(1/kms_s_0763_br_0430)^9*(1/kms_s_1005r_0430)^3*(1/kms_s_1096r_0430)^6*(s_0380^1*s_0763_b^9*s_1005^3*s_1096^6-s_0470^3*s_0514^3*s_1091^6*s_1140^1*s_1434_b^3/Keq_r_0430)/(((1+s_0380/kms_s_0380r_0430)*(1+s_0763_b/kms_s_0763_br_0430)*(1+s_1005/kms_s_1005r_0430)*(1+s_1096/kms_s_1096r_0430)+(1+s_0470/kmp_s_0470r_0430)*(1+s_0514/kmp_s_0514r_0430)*(1+s_1091/kmp_s_1091r_0430)*(1+s_1140/kmp_s_1140r_0430)*(1+s_1434_b/kmp_s_1434_br_0430))-1)/intracellular
Vmax_r_0290=0.00279509; kms_s_1325r_0290=0.549; kmp_s_0763_br_0290=0.549; kmp_s_0514r_0290=0.549; kmp_s_1080r_0290=0.549; Keq_r_0290=0.6039; kms_s_1355r_0290=0.549Reaction: s_1325 + s_1355 => s_0514 + s_0763_b + s_1080; s_0514, s_0763_b, s_1080, s_1325, s_1355, Rate Law: intracellular*Vmax_r_0290*(1/kms_s_1325r_0290)^1*(1/kms_s_1355r_0290)^1*(s_1325^1*s_1355^1-s_0514^1*s_0763_b^1*s_1080^1/Keq_r_0290)/(((1+s_1325/kms_s_1325r_0290)*(1+s_1355/kms_s_1355r_0290)+(1+s_0514/kmp_s_0514r_0290)*(1+s_0763_b/kmp_s_0763_br_0290)*(1+s_1080/kmp_s_1080r_0290))-1)/intracellular
kms_s_0659r_0488=0.549; kmp_s_1338r_0488=0.549; Vmax_r_0488=4.5199; Keq_r_0488=1.1; kmp_s_0657r_0488=0.549; kms_s_0692r_0488=0.549Reaction: s_0659 + s_0692 => s_0657 + s_1338; s_0657, s_0659, s_0692, s_1338, Rate Law: intracellular*Vmax_r_0488*(1/kms_s_0659r_0488)^1*(1/kms_s_0692r_0488)^1*(s_0659^1*s_0692^1-s_0657^1*s_1338^1/Keq_r_0488)/(((1+s_0659/kms_s_0659r_0488)*(1+s_0692/kms_s_0692r_0488)+(1+s_0657/kmp_s_0657r_0488)*(1+s_1338/kmp_s_1338r_0488))-1)/intracellular
Vmax_r_0063=0.764505; kmp_s_0008r_0063=0.549; Keq_r_0063=2.00364; kms_s_0170r_0063=0.549; kms_s_1434_br_0063=0.549Reaction: s_0170 + s_1434_b => s_0008; s_0008, s_0170, s_1434_b, Rate Law: intracellular*Vmax_r_0063*(1/kms_s_0170r_0063)^1*(1/kms_s_1434_br_0063)^1*(s_0170^1*s_1434_b^1-s_0008^1/Keq_r_0063)/(((1+s_0170/kms_s_0170r_0063)*(1+s_1434_b/kms_s_1434_br_0063)+1+s_0008/kmp_s_0008r_0063)-1)/intracellular
kmp_s_0763_br_0859=0.549; kmI_s_0446mr_0859=4.0; intracellular=1.0; kmp_s_0400r_0859=1.71907; Keq_r_0859=12.2086; Vmax_r_0859=84.3466; kms_s_0446r_0859=1.09208; kmp_s_0537r_0859=1.34278; kms_s_0539r_0859=0.104555Reaction: s_0446 + s_0539 => s_0400 + s_0537 + s_0763_b; s_0446, s_0446, s_0539, s_0400, s_0537, s_0763_b, Rate Law: intracellular*Vmax_r_0859*(1/kms_s_0446r_0859)^1*(1/kms_s_0539r_0859)^1*(s_0446^1*s_0539^1-s_0400^1*s_0537^1*s_0763_b^1/Keq_r_0859)/((1+s_0446/kms_s_0446r_0859)*(1+s_0539/kms_s_0539r_0859)+(1+s_0400/kmp_s_0400r_0859)*(1+s_0537/kmp_s_0537r_0859)*(1+s_0763_b/kmp_s_0763_br_0859)+((1+s_0446/kmI_s_0446mr_0859)-1))/intracellular
kms_s_0118r_0661=0.549; Keq_r_0661=63.2537; kmp_s_1082r_0661=1.50326; Vmax_r_0661=3.30332; kmp_s_1379r_0661=0.549; kms_s_0763_br_0661=0.549; kms_s_1087r_0661=0.0867353Reaction: s_0118 + s_0763_b + s_1087 => s_1082 + s_1379; s_0118, s_0763_b, s_1082, s_1087, s_1379, Rate Law: intracellular*Vmax_r_0661*(1/kms_s_0118r_0661)^1*(1/kms_s_0763_br_0661)^2*(1/kms_s_1087r_0661)^1*(s_0118^1*s_0763_b^2*s_1087^1-s_1082^1*s_1379^1/Keq_r_0661)/(((1+s_0118/kms_s_0118r_0661)*(1+s_0763_b/kms_s_0763_br_0661)*(1+s_1087/kms_s_1087r_0661)+(1+s_1082/kmp_s_1082r_0661)*(1+s_1379/kmp_s_1379r_0661))-1)/intracellular
kmp_s_0007r_0640=0.549; kms_s_0763_br_0640=0.549; Keq_r_0640=2.00364; kms_s_1096r_0640=0.549; kms_s_0042r_0640=0.549; Vmax_r_0640=1.15192; kmp_s_1091r_0640=0.549Reaction: s_0042 + s_0763_b + s_1096 => s_0007 + s_1091; s_0007, s_0042, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0640*(1/kms_s_0042r_0640)^1*(1/kms_s_0763_br_0640)^1*(1/kms_s_1096r_0640)^1*(s_0042^1*s_0763_b^1*s_1096^1-s_0007^1*s_1091^1/Keq_r_0640)/(((1+s_0042/kms_s_0042r_0640)*(1+s_0763_b/kms_s_0763_br_0640)*(1+s_1096/kms_s_1096r_0640)+(1+s_0007/kmp_s_0007r_0640)*(1+s_1091/kmp_s_1091r_0640))-1)/intracellular
kmp_s_1517r_0959=0.549; kms_s_0446r_0959=1.09208; Keq_r_0959=0.303587; kms_s_1521r_0959=0.549; kmp_s_1434_br_0959=0.549; kmp_s_0566r_0959=0.549; Vmax_r_0959=0.0120516Reaction: s_0446 + s_1521 => s_0566 + s_1434_b + s_1517; s_0446, s_0566, s_1434_b, s_1517, s_1521, Rate Law: intracellular*Vmax_r_0959*(1/kms_s_0446r_0959)^1*(1/kms_s_1521r_0959)^1*(s_0446^1*s_1521^1-s_0566^1*s_1434_b^1*s_1517^1/Keq_r_0959)/(((1+s_0446/kms_s_0446r_0959)*(1+s_1521/kms_s_1521r_0959)+(1+s_0566/kmp_s_0566r_0959)*(1+s_1434_b/kmp_s_1434_br_0959)*(1+s_1517/kmp_s_1517r_0959))-1)/intracellular
kms_s_0881r_0232=0.549; kmp_s_0763_br_0232=0.549; kmp_s_1073r_0232=0.549; kmp_s_1207r_0232=0.549; Vmax_r_0232=0.826427; kms_s_0469r_0232=0.549; Keq_r_0232=0.6039Reaction: s_0469 + s_0881 => s_0763_b + s_1073 + s_1207; s_0469, s_0763_b, s_0881, s_1073, s_1207, Rate Law: intracellular*Vmax_r_0232*(1/kms_s_0469r_0232)^1*(1/kms_s_0881r_0232)^1*(s_0469^1*s_0881^1-s_0763_b^1*s_1073^1*s_1207^1/Keq_r_0232)/(((1+s_0469/kms_s_0469r_0232)*(1+s_0881/kms_s_0881r_0232)+(1+s_0763_b/kmp_s_0763_br_0232)*(1+s_1073/kmp_s_1073r_0232)*(1+s_1207/kmp_s_1207r_0232))-1)/intracellular
kmp_s_1087r_0940=0.0867353; kms_s_0514r_0940=0.549; Vmax_r_0940=9.4545; kmp_s_0470r_0940=1.0; kmp_s_0380r_0940=0.549; kms_s_1277r_0940=0.0605905; Keq_r_0940=1.04749; kms_s_1082r_0940=1.50326Reaction: s_0514 + s_1082 + s_1277 => s_0380 + s_0470 + s_1087; s_0380, s_0470, s_0514, s_1082, s_1087, s_1277, Rate Law: intracellular*Vmax_r_0940*(1/kms_s_0514r_0940)^1*(1/kms_s_1082r_0940)^1*(1/kms_s_1277r_0940)^1*(s_0514^1*s_1082^1*s_1277^1-s_0380^1*s_0470^1*s_1087^1/Keq_r_0940)/(((1+s_0514/kms_s_0514r_0940)*(1+s_1082/kms_s_1082r_0940)*(1+s_1277/kms_s_1277r_0940)+(1+s_0380/kmp_s_0380r_0940)*(1+s_0470/kmp_s_0470r_0940)*(1+s_1087/kmp_s_1087r_0940))-1)/intracellular
kmp_s_1290r_0874=0.549; Vmax_r_0874=0.0193599; kms_s_1293r_0874=0.549; Keq_r_0874=0.6039; kmp_s_1225r_0874=0.549; kmp_s_0763_br_0874=0.549; kms_s_1226r_0874=0.549Reaction: s_1226 + s_1293 => s_0763_b + s_1225 + s_1290; s_0763_b, s_1225, s_1226, s_1290, s_1293, Rate Law: intracellular*Vmax_r_0874*(1/kms_s_1226r_0874)^1*(1/kms_s_1293r_0874)^1*(s_1226^1*s_1293^1-s_0763_b^1*s_1225^1*s_1290^1/Keq_r_0874)/(((1+s_1226/kms_s_1226r_0874)*(1+s_1293/kms_s_1293r_0874)+(1+s_0763_b/kmp_s_0763_br_0874)*(1+s_1225/kmp_s_1225r_0874)*(1+s_1290/kmp_s_1290r_0874))-1)/intracellular
kmp_s_0766_br_1503=0.1; kmp_s_1339_br_1503=1.0; Keq_r_1503=1.0; kms_s_0763_br_1503=0.549; Vmax_r_1503=0.840147; kms_s_1338r_1503=0.549Reaction: s_0763_b + s_1338 => s_0766_b + s_1339_b; s_0763_b, s_0766_b, s_1338, s_1339_b, Rate Law: Vmax_r_1503*(1/kms_s_0763_br_1503)^1*(1/kms_s_1338r_1503)^1*(s_0763_b^1*s_1338^1-s_0766_b^1*s_1339_b^1/Keq_r_1503)/(((1+s_0763_b/kms_s_0763_br_1503)*(1+s_1338/kms_s_1338r_1503)+(1+s_0766_b/kmp_s_0766_br_1503)*(1+s_1339_b/kmp_s_1339_br_1503))-1)
kmp_s_0763_br_0040=0.549; kms_s_0557r_0040=0.549; kmp_s_0163r_0040=0.549; Vmax_r_0040=0.00989001; kmp_s_0689r_0040=0.549; Keq_r_0040=0.331541Reaction: s_0557 => s_0163 + s_0689 + s_0763_b; s_0163, s_0557, s_0689, s_0763_b, Rate Law: intracellular*Vmax_r_0040*(1/kms_s_0557r_0040)^1*(s_0557^1-s_0163^1*s_0689^1*s_0763_b^1/Keq_r_0040)/((1+s_0557/kms_s_0557r_0040+(1+s_0163/kmp_s_0163r_0040)*(1+s_0689/kmp_s_0689r_0040)*(1+s_0763_b/kmp_s_0763_br_0040))-1)/intracellular
Vmax_r_0951=0.0120515; Keq_r_0951=0.192861; kmp_s_1517r_0951=0.549; kmp_s_0562r_0951=0.549; kmp_s_1434_br_0951=0.549; kms_s_1521r_0951=0.549; kms_s_0400r_0951=1.71907Reaction: s_0400 + s_1521 => s_0562 + s_1434_b + s_1517; s_0400, s_0562, s_1434_b, s_1517, s_1521, Rate Law: intracellular*Vmax_r_0951*(1/kms_s_0400r_0951)^1*(1/kms_s_1521r_0951)^1*(s_0400^1*s_1521^1-s_0562^1*s_1434_b^1*s_1517^1/Keq_r_0951)/(((1+s_0400/kms_s_0400r_0951)*(1+s_1521/kms_s_1521r_0951)+(1+s_0562/kmp_s_0562r_0951)*(1+s_1434_b/kmp_s_1434_br_0951)*(1+s_1517/kmp_s_1517r_0951))-1)/intracellular
kmp_s_1306r_0976=0.549; kms_s_1096r_0976=0.549; kms_s_0217r_0976=0.549; kms_s_0763_br_0976=0.549; Vmax_r_0976=1.60931; Keq_r_0976=2.00364; kmp_s_1091r_0976=0.549Reaction: s_0217 + s_0763_b + s_1096 => s_1091 + s_1306; s_0217, s_0763_b, s_1091, s_1096, s_1306, Rate Law: intracellular*Vmax_r_0976*(1/kms_s_0217r_0976)^1*(1/kms_s_0763_br_0976)^1*(1/kms_s_1096r_0976)^1*(s_0217^1*s_0763_b^1*s_1096^1-s_1091^1*s_1306^1/Keq_r_0976)/(((1+s_0217/kms_s_0217r_0976)*(1+s_0763_b/kms_s_0763_br_0976)*(1+s_1096/kms_s_1096r_0976)+(1+s_1091/kmp_s_1091r_0976)*(1+s_1306/kmp_s_1306r_0976))-1)/intracellular
kms_s_0470r_0883=1.0; kmp_s_0318r_0883=0.549; kms_s_0316r_0883=0.549; Vmax_r_0883=0.46739; Keq_r_0883=0.6039; kmp_s_0763_br_0883=0.549Reaction: s_0316 + s_0470 => s_0318 + s_0763_b; s_0316, s_0318, s_0470, s_0763_b, Rate Law: intracellular*Vmax_r_0883*(1/kms_s_0316r_0883)^1*(1/kms_s_0470r_0883)^1*(s_0316^1*s_0470^1-s_0318^1*s_0763_b^1/Keq_r_0883)/(((1+s_0316/kms_s_0316r_0883)*(1+s_0470/kms_s_0470r_0883)+(1+s_0318/kmp_s_0318r_0883)*(1+s_0763_b/kmp_s_0763_br_0883))-1)/intracellular
kms_s_0218r_0044=0.549; kms_s_1096r_0044=0.549; Vmax_r_0044=0.00279511; kms_s_0763_br_0044=0.549; kmp_s_1325r_0044=0.549; Keq_r_0044=3.64962; kmp_s_1091r_0044=0.549Reaction: s_0218 + s_0763_b + s_1096 => s_1091 + s_1325; s_0218, s_0763_b, s_1091, s_1096, s_1325, Rate Law: intracellular*Vmax_r_0044*(1/kms_s_0218r_0044)^1*(1/kms_s_0763_br_0044)^2*(1/kms_s_1096r_0044)^1*(s_0218^1*s_0763_b^2*s_1096^1-s_1091^1*s_1325^1/Keq_r_0044)/(((1+s_0218/kms_s_0218r_0044)*(1+s_0763_b/kms_s_0763_br_0044)*(1+s_1096/kms_s_1096r_0044)+(1+s_1091/kmp_s_1091r_0044)*(1+s_1325/kmp_s_1325r_0044))-1)/intracellular
Keq_r_0660=0.331541; kmp_s_0118r_0660=0.549; kms_s_1091r_0660=0.549; kmp_s_1096r_0660=0.549; Vmax_r_0660=3.30329; kmp_s_0763_br_0660=0.549; kms_s_1379r_0660=0.549Reaction: s_1091 + s_1379 => s_0118 + s_0763_b + s_1096; s_0118, s_0763_b, s_1091, s_1096, s_1379, Rate Law: intracellular*Vmax_r_0660*(1/kms_s_1091r_0660)^1*(1/kms_s_1379r_0660)^1*(s_1091^1*s_1379^1-s_0118^1*s_0763_b^2*s_1096^1/Keq_r_0660)/(((1+s_1091/kms_s_1091r_0660)*(1+s_1379/kms_s_1379r_0660)+(1+s_0118/kmp_s_0118r_0660)*(1+s_0763_b/kmp_s_0763_br_0660)*(1+s_1096/kmp_s_1096r_0660))-1)/intracellular
kms_s_1096r_0464=0.549; kmp_s_0514r_0464=0.549; kmp_s_1091r_0464=0.549; kms_s_0763_br_0464=0.549; Keq_r_0464=3.64962; kmp_s_1434_br_0464=0.549; kms_s_0582r_0464=0.549; kms_s_1005r_0464=0.549; kmp_s_0470r_0464=1.0; Vmax_r_0464=0.0179399; kmp_s_0977r_0464=0.549Reaction: s_0582 + s_0763_b + s_1005 + s_1096 => s_0470 + s_0514 + s_0977 + s_1091 + s_1434_b; s_0470, s_0514, s_0582, s_0763_b, s_0977, s_1005, s_1091, s_1096, s_1434_b, Rate Law: intracellular*Vmax_r_0464*(1/kms_s_0582r_0464)^1*(1/kms_s_0763_br_0464)^3*(1/kms_s_1005r_0464)^1*(1/kms_s_1096r_0464)^2*(s_0582^1*s_0763_b^3*s_1005^1*s_1096^2-s_0470^1*s_0514^1*s_0977^1*s_1091^2*s_1434_b^1/Keq_r_0464)/(((1+s_0582/kms_s_0582r_0464)*(1+s_0763_b/kms_s_0763_br_0464)*(1+s_1005/kms_s_1005r_0464)*(1+s_1096/kms_s_1096r_0464)+(1+s_0470/kmp_s_0470r_0464)*(1+s_0514/kmp_s_0514r_0464)*(1+s_0977/kmp_s_0977r_0464)*(1+s_1091/kmp_s_1091r_0464)*(1+s_1434_b/kmp_s_1434_br_0464))-1)/intracellular
Keq_r_0698=5.77591; Vmax_r_0698=1.5048; kmp_s_0554r_0698=0.549; kms_s_0539r_0698=0.104555Reaction: s_0539 => s_0554; s_0539, s_0554, Rate Law: intracellular*Vmax_r_0698*(1/kms_s_0539r_0698)^1*(s_0539^1-s_0554^1/Keq_r_0698)/((1+s_0539/kms_s_0539r_0698+1+s_0554/kmp_s_0554r_0698)-1)/intracellular
kms_s_0446r_0977=1.09208; kmp_s_0267r_0977=0.549; kms_s_1306r_0977=0.549; Vmax_r_0977=1.60929; kmp_s_0400r_0977=1.71907; Keq_r_0977=0.950614; kmp_s_0763_br_0977=0.549Reaction: s_0446 + s_1306 => s_0267 + s_0400 + s_0763_b; s_0267, s_0400, s_0446, s_0763_b, s_1306, Rate Law: intracellular*Vmax_r_0977*(1/kms_s_0446r_0977)^1*(1/kms_s_1306r_0977)^1*(s_0446^1*s_1306^1-s_0267^1*s_0400^1*s_0763_b^1/Keq_r_0977)/(((1+s_0446/kms_s_0446r_0977)*(1+s_1306/kms_s_1306r_0977)+(1+s_0267/kmp_s_0267r_0977)*(1+s_0400/kmp_s_0400r_0977)*(1+s_0763_b/kmp_s_0763_br_0977))-1)/intracellular
Keq_r_0707=1.1; Vmax_r_0707=1.2166; kms_s_1091r_0707=0.549; kms_s_0307r_0707=0.549; kmp_s_1096r_0707=0.549; kmp_s_0015r_0707=0.549Reaction: s_0307 + s_1091 => s_0015 + s_1096; s_0015, s_0307, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0707*(1/kms_s_0307r_0707)^1*(1/kms_s_1091r_0707)^1*(s_0307^1*s_1091^1-s_0015^1*s_1096^1/Keq_r_0707)/(((1+s_0307/kms_s_0307r_0707)*(1+s_1091/kms_s_1091r_0707)+(1+s_0015/kmp_s_0015r_0707)*(1+s_1096/kmp_s_1096r_0707))-1)/intracellular
a_s_0873r_1812=0.13579; a_s_0949r_1812=0.19653; s_0743_or_1812=0.549; s_0960_or_1812=1.0; s_1283_or_1812=0.549; a_s_0434r_1812=0.051; a_s_0593r_1812=0.002432; s_0936_or_1812=0.549; s_1011_or_1812=0.549; a_s_0564r_1812=0.003587; s_0929_or_1812=0.549; a_s_0943r_1812=0.25371; a_s_0960r_1812=0.25728; a_s_1000r_1812=1.0; a_s_0933r_1812=0.050027; s_0619_or_1812=0.549; a_s_0416r_1812=0.023371; s_0511_or_1812=0.549; s_0920_or_1812=0.549; a_s_0955r_1812=0.096481; s_0889_or_1812=0.549; a_s_0743r_1812=0.51852; a_s_0752r_1812=0.051; a_s_0925r_1812=0.25014; s_0949_or_1812=1.0; s_0863_or_1812=0.549; s_0907_or_1812=0.549; a_s_0907r_1812=0.268; s_0939_or_1812=0.549; a_s_0001r_1812=1.1358; a_s_1011r_1812=0.82099; s_0899_or_1812=0.549; s_0955_or_1812=0.549; s_1347_or_1812=0.549; a_s_0446r_1812=59.276; s_0952_or_1812=1.0; s_0416_or_1812=0.549; a_s_0929r_1812=0.23942; s_0881_or_1812=0.549; a_s_0899r_1812=0.268; s_0873_or_1812=0.549; s_0569_or_1812=0.549; s_0593_or_1812=0.549; V_o=0.0555; a_s_0939r_1812=0.12864; a_s_0881r_1812=0.17152; a_s_0569r_1812=0.002432; s_0877_or_1812=0.549; a_s_0863r_1812=0.35734; a_s_0952r_1812=0.028; a_s_1347r_1812=0.02; s_0933_or_1812=0.549; s_0564_or_1812=0.549; s_0925_or_1812=0.549; a_s_0877r_1812=0.17152; s_1000_or_1812=0.549; a_s_0911r_1812=0.075041; s_0740_or_1812=0.549; s_0752_or_1812=0.549; zero_flux=0.0; a_s_1417r_1812=0.067; s_0001_or_1812=0.549; s_0943_or_1812=0.549; a_s_0920r_1812=0.17152; a_s_0619r_1812=0.003587; a_s_0740r_1812=0.32518; a_s_0511r_1812=0.05; s_1417_or_1812=0.549; a_s_0889r_1812=0.04288; a_s_0936r_1812=0.11435; s_0434_or_1812=1.25956; a_s_1283r_1812=9.0E-4; s_0446_or_1812=1.09208; s_0911_or_1812=0.549Reaction: s_0001 + s_0416 + s_0434 + s_0446 + s_0511 + s_0564 + s_0569 + s_0593 + s_0619 + s_0740 + s_0743 + s_0752 + s_0863 + s_0873 + s_0877 + s_0881 + s_0889 + s_0899 + s_0907 + s_0911 + s_0920 + s_0925 + s_0929 + s_0933 + s_0936 + s_0939 + s_0943 + s_0949 + s_0952 + s_0955 + s_0960 + s_1000 + s_1011 + s_1347 + s_1417 + s_1283 => s_0400 + s_0463 + s_1207; s_0547_b, s_0001, s_0416, s_0434, s_0446, s_0511, s_0564, s_0569, s_0593, s_0619, s_0740, s_0743, s_0752, s_0863, s_0873, s_0877, s_0881, s_0889, s_0899, s_0907, s_0911, s_0920, s_0925, s_0929, s_0933, s_0936, s_0939, s_0943, s_0949, s_0952, s_0955, s_0960, s_1000, s_1011, s_1283, s_1347, s_1417, Rate Law: intracellular*piecewise(V_o*(1+a_s_0001r_1812*ln(s_0001/s_0001_or_1812)+a_s_0416r_1812*ln(s_0416/s_0416_or_1812)+a_s_0434r_1812*ln(s_0434/s_0434_or_1812)+a_s_0446r_1812*ln(s_0446/s_0446_or_1812)+a_s_0511r_1812*ln(s_0511/s_0511_or_1812)+a_s_0564r_1812*ln(s_0564/s_0564_or_1812)+a_s_0569r_1812*ln(s_0569/s_0569_or_1812)+a_s_0593r_1812*ln(s_0593/s_0593_or_1812)+a_s_0619r_1812*ln(s_0619/s_0619_or_1812)+a_s_0740r_1812*ln(s_0740/s_0740_or_1812)+a_s_0743r_1812*ln(s_0743/s_0743_or_1812)+a_s_0752r_1812*ln(s_0752/s_0752_or_1812)+a_s_0863r_1812*ln(s_0863/s_0863_or_1812)+a_s_0873r_1812*ln(s_0873/s_0873_or_1812)+a_s_0877r_1812*ln(s_0877/s_0877_or_1812)+a_s_0881r_1812*ln(s_0881/s_0881_or_1812)+a_s_0889r_1812*ln(s_0889/s_0889_or_1812)+a_s_0899r_1812*ln(s_0899/s_0899_or_1812)+a_s_0907r_1812*ln(s_0907/s_0907_or_1812)+a_s_0911r_1812*ln(s_0911/s_0911_or_1812)+a_s_0920r_1812*ln(s_0920/s_0920_or_1812)+a_s_0925r_1812*ln(s_0925/s_0925_or_1812)+a_s_0929r_1812*ln(s_0929/s_0929_or_1812)+a_s_0933r_1812*ln(s_0933/s_0933_or_1812)+a_s_0936r_1812*ln(s_0936/s_0936_or_1812)+a_s_0939r_1812*ln(s_0939/s_0939_or_1812)+a_s_0943r_1812*ln(s_0943/s_0943_or_1812)+a_s_0949r_1812*ln(s_0949/s_0949_or_1812)+a_s_0952r_1812*ln(s_0952/s_0952_or_1812)+a_s_0955r_1812*ln(s_0955/s_0955_or_1812)+a_s_0960r_1812*ln(s_0960/s_0960_or_1812)+a_s_1000r_1812*ln(s_1000/s_1000_or_1812)+a_s_1011r_1812*ln(s_1011/s_1011_or_1812)+a_s_1347r_1812*ln(s_1347/s_1347_or_1812)+a_s_1417r_1812*ln(s_1417/s_1417_or_1812)+a_s_1283r_1812*ln(s_1283/s_1283_or_1812)), (V_o*(1+a_s_0001r_1812*ln(s_0001/s_0001_or_1812)+a_s_0416r_1812*ln(s_0416/s_0416_or_1812)+a_s_0434r_1812*ln(s_0434/s_0434_or_1812)+a_s_0446r_1812*ln(s_0446/s_0446_or_1812)+a_s_0511r_1812*ln(s_0511/s_0511_or_1812)+a_s_0564r_1812*ln(s_0564/s_0564_or_1812)+a_s_0569r_1812*ln(s_0569/s_0569_or_1812)+a_s_0593r_1812*ln(s_0593/s_0593_or_1812)+a_s_0619r_1812*ln(s_0619/s_0619_or_1812)+a_s_0740r_1812*ln(s_0740/s_0740_or_1812)+a_s_0743r_1812*ln(s_0743/s_0743_or_1812)+a_s_0752r_1812*ln(s_0752/s_0752_or_1812)+a_s_0863r_1812*ln(s_0863/s_0863_or_1812)+a_s_0873r_1812*ln(s_0873/s_0873_or_1812)+a_s_0877r_1812*ln(s_0877/s_0877_or_1812)+a_s_0881r_1812*ln(s_0881/s_0881_or_1812)+a_s_0889r_1812*ln(s_0889/s_0889_or_1812)+a_s_0899r_1812*ln(s_0899/s_0899_or_1812)+a_s_0907r_1812*ln(s_0907/s_0907_or_1812)+a_s_0911r_1812*ln(s_0911/s_0911_or_1812)+a_s_0920r_1812*ln(s_0920/s_0920_or_1812)+a_s_0925r_1812*ln(s_0925/s_0925_or_1812)+a_s_0929r_1812*ln(s_0929/s_0929_or_1812)+a_s_0933r_1812*ln(s_0933/s_0933_or_1812)+a_s_0936r_1812*ln(s_0936/s_0936_or_1812)+a_s_0939r_1812*ln(s_0939/s_0939_or_1812)+a_s_0943r_1812*ln(s_0943/s_0943_or_1812)+a_s_0949r_1812*ln(s_0949/s_0949_or_1812)+a_s_0952r_1812*ln(s_0952/s_0952_or_1812)+a_s_0955r_1812*ln(s_0955/s_0955_or_1812)+a_s_0960r_1812*ln(s_0960/s_0960_or_1812)+a_s_1000r_1812*ln(s_1000/s_1000_or_1812)+a_s_1011r_1812*ln(s_1011/s_1011_or_1812)+a_s_1347r_1812*ln(s_1347/s_1347_or_1812)+a_s_1417r_1812*ln(s_1417/s_1417_or_1812)+a_s_1283r_1812*ln(s_1283/s_1283_or_1812))) >= zero_flux, zero_flux)/intracellular
Vmax_r_0568=0.0076692; kmp_s_0706r_0568=0.549; Keq_r_0568=1.1; kms_s_0566r_0568=0.549; kms_s_0752r_0568=0.549; kmp_s_0562r_0568=0.549Reaction: s_0566 + s_0752 => s_0562 + s_0706; s_0562, s_0566, s_0706, s_0752, Rate Law: intracellular*Vmax_r_0568*(1/kms_s_0566r_0568)^1*(1/kms_s_0752r_0568)^1*(s_0566^1*s_0752^1-s_0562^1*s_0706^1/Keq_r_0568)/(((1+s_0566/kms_s_0566r_0568)*(1+s_0752/kms_s_0752r_0568)+(1+s_0562/kmp_s_0562r_0568)*(1+s_0706/kmp_s_0706r_0568))-1)/intracellular
kmp_s_1187r_0466=0.549; Keq_r_0466=3.64962; Vmax_r_0466=0.0179399; kmp_s_1434_br_0466=0.549; kms_s_1096r_0466=0.549; kmp_s_1091r_0466=0.549; kmp_s_0514r_0466=0.549; kms_s_0763_br_0466=0.549; kms_s_1005r_0466=0.549; kmp_s_0470r_0466=1.0; kms_s_1044r_0466=0.549Reaction: s_0763_b + s_1005 + s_1044 + s_1096 => s_0470 + s_0514 + s_1091 + s_1187 + s_1434_b; s_0470, s_0514, s_0763_b, s_1005, s_1044, s_1091, s_1096, s_1187, s_1434_b, Rate Law: intracellular*Vmax_r_0466*(1/kms_s_0763_br_0466)^3*(1/kms_s_1005r_0466)^1*(1/kms_s_1044r_0466)^1*(1/kms_s_1096r_0466)^2*(s_0763_b^3*s_1005^1*s_1044^1*s_1096^2-s_0470^1*s_0514^1*s_1091^2*s_1187^1*s_1434_b^1/Keq_r_0466)/(((1+s_0763_b/kms_s_0763_br_0466)*(1+s_1005/kms_s_1005r_0466)*(1+s_1044/kms_s_1044r_0466)*(1+s_1096/kms_s_1096r_0466)+(1+s_0470/kmp_s_0470r_0466)*(1+s_0514/kmp_s_0514r_0466)*(1+s_1091/kmp_s_1091r_0466)*(1+s_1187/kmp_s_1187r_0466)*(1+s_1434_b/kmp_s_1434_br_0466))-1)/intracellular

States:

NameDescription
s 0315[5-[(5-phospho-1-deoxy-D-ribulos-1-ylimino)methylamino]-1-(5-phospho-beta-D-ribosyl)imidazole-4-carboxamide]
s 0009[SAICAR]
s 0881[L-aspartate(1-)]
s 0562[dADP]
s 0650[ethanol]
s 0514[coenzyme A]
s 1325[sphinganine]
s 0015[(6R)-5,10-methenyltetrahydrofolic acid]
s 0566[dATP]
s 1355[tetracosanoyl-CoA]
s 0316[5-amino-1-(5-phospho-D-ribosyl)imidazole]
s 0564[dAMP]
s 1070[N-acetyl-L-gamma-glutamyl phosphate]
s 0641[ergosteryl ester]
s 0416[alpha,alpha-trehalose]
s 1342[succinyl-CoA]
s 0419[alpha,alpha-trehalose 6-phosphate]
s 1082[NAD(+)]
s 0366[acetaldehyde]
s 1434 b[water]
s 0434[AMP]
s 0008[(2R,3S)-3-isopropylmalate(2-)]
s 0539[keto-D-fructose 6-phosphate]
s 0446[ATP(4-)]
s 0554[D-mannose 6-phosphate]
s 1306[shikimate]
s 0706[GDP]
s 1338[succinate(2-)]
s 0763 b[proton]
s 0410[aldehydo-D-glucose 6-phosphate]
s 1347[sulfate]
s 0533[D-erythrose 4-phosphate(2-)]
s 1379[trans-4-hydroxy-L-proline]
s 0557[D-ribulose 5-phosphate]
s 0569[dCMP]
s 1096[NADPH]
s 0309[5,6,7,8-tetrahydrofolic acid]
s 0561[D-xylulose 5-phosphate]
s 1349[sulfite]
s 0537[keto-D-fructose 1,6-bisphosphate]
s 0427[alpha-D-ribofuranose 5-phosphate]
s 0317[AICA ribonucleotide]
s 0007[(2R,3R)-2,3-dihydroxy-3-methylpentanoate]
s 0010[(2S)-2-isopropyl-3-oxosuccinate(2-)]
s 1315[sn-glycerol 3-phosphate]
s 0692[fumarate(2-)]

Stanford2013 - Kinetic model of yeast metabolic network (standard): BIOMD0000000496v0.0.1

Stanford2013 - Kinetic model of yeast metabolic network (standard)Large-scale model construction based on a logical laye…

Details

The quantitative effects of environmental and genetic perturbations on metabolism can be studied in silico using kinetic models. We present a strategy for large-scale model construction based on a logical layering of data such as reaction fluxes, metabolite concentrations, and kinetic constants. The resulting models contain realistic standard rate laws and plausible parameters, adhere to the laws of thermodynamics, and reproduce a predefined steady state. These features have not been simultaneously achieved by previous workflows. We demonstrate the advantages and limitations of the workflow by translating the yeast consensus metabolic network into a kinetic model. Despite crudely selected data, the model shows realistic control behaviour, a stable dynamic, and realistic response to perturbations in extracellular glucose concentrations. The paper concludes by outlining how new data can continuously be fed into the workflow and how iterative model building can assist in directing experiments. link: http://identifiers.org/pubmed/24324546

Parameters:

NameDescription
kms_s_1091r_0722=0.549; kmp_s_1096r_0722=0.549; kms_s_0055r_0722=0.549; Keq_r_0722=0.6039; kmp_s_0261r_0722=0.549; Vmax_r_0722=3.30329; kmp_s_0763_br_0722=0.549Reaction: s_0055 + s_1091 => s_0261 + s_0763_b + s_1096; s_0055, s_0261, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0722*(1/kms_s_0055r_0722)^1*(1/kms_s_1091r_0722)^1*(s_0055^1*s_1091^1-s_0261^1*s_0763_b^1*s_1096^1/Keq_r_0722)/(((1+s_0055/kms_s_0055r_0722)*(1+s_1091/kms_s_1091r_0722)+(1+s_0261/kmp_s_0261r_0722)*(1+s_0763_b/kmp_s_0763_br_0722)*(1+s_1096/kmp_s_1096r_0722))-1)/intracellular
Vmax_r_1435=0.0232306; kmp_s_1160r_1435=0.549; Keq_r_1435=1.0; kms_s_1162_br_1435=24.5Reaction: s_1162_b => s_1160; s_1160, s_1162_b, Rate Law: Vmax_r_1435*(1/kms_s_1162_br_1435)^1*(s_1162_b^1-s_1160^1/Keq_r_1435)/((1+s_1162_b/kms_s_1162_br_1435+1+s_1160/kmp_s_1160r_1435)-1)
Keq_r_0370=0.0999269; Vmax_r_0370=0.0120878; kmp_s_0514r_0370=0.549; kms_s_0386r_0370=0.549; kmp_s_1399r_0370=0.549; kms_s_0596r_0370=0.549; kmp_s_0763_br_0370=0.549Reaction: s_0386 + s_0596 => s_0514 + s_0763_b + s_1399; s_0386, s_0514, s_0596, s_0763_b, s_1399, Rate Law: intracellular*Vmax_r_0370*(1/kms_s_0386r_0370)^1*(1/kms_s_0596r_0370)^1*(s_0386^1*s_0596^1-s_0514^1*s_0763_b^4*s_1399^1/Keq_r_0370)/(((1+s_0386/kms_s_0386r_0370)*(1+s_0596/kms_s_0596r_0370)+(1+s_0514/kmp_s_0514r_0370)*(1+s_0763_b/kmp_s_0763_br_0370)*(1+s_1399/kmp_s_1399r_0370))-1)/intracellular
kmp_s_0731r_1042=0.0436363; kms_s_0088r_1042=0.549; Keq_r_1042=0.0874316; kmp_s_1434_br_1042=0.549; kmp_s_0952r_1042=1.0; Vmax_r_1042=0.187549; kms_s_0943r_1042=0.549Reaction: s_0088 + s_0943 => s_0731 + s_0952 + s_1434_b; s_0088, s_0731, s_0943, s_0952, s_1434_b, Rate Law: intracellular*Vmax_r_1042*(1/kms_s_0088r_1042)^1*(1/kms_s_0943r_1042)^1*(s_0088^1*s_0943^1-s_0731^1*s_0952^1*s_1434_b^1/Keq_r_1042)/(((1+s_0088/kms_s_0088r_1042)*(1+s_0943/kms_s_0943r_1042)+(1+s_0731/kmp_s_0731r_1042)*(1+s_0952/kmp_s_0952r_1042)*(1+s_1434_b/kmp_s_1434_br_1042))-1)/intracellular
Vmax_r_1038=0.1001; kms_s_1434_br_1038=0.549; Keq_r_1038=1.1; kms_s_0419r_1038=0.549; kmp_s_0416r_1038=0.549; kmp_s_1207r_1038=0.549Reaction: s_0419 + s_1434_b => s_0416 + s_1207; s_0416, s_0419, s_1207, s_1434_b, Rate Law: intracellular*Vmax_r_1038*(1/kms_s_0419r_1038)^1*(1/kms_s_1434_br_1038)^1*(s_0419^1*s_1434_b^1-s_0416^1*s_1207^1/Keq_r_1038)/(((1+s_0419/kms_s_0419r_1038)*(1+s_1434_b/kms_s_1434_br_1038)+(1+s_0416/kmp_s_0416r_1038)*(1+s_1207/kmp_s_1207r_1038))-1)/intracellular
kms_s_0438r_0006=0.549; Vmax_r_0006=1.58399; kmp_s_0743r_0006=0.549; kmp_s_1434_br_0006=0.549; Keq_r_0006=0.6039Reaction: s_0438 => s_0743 + s_1434_b; s_0438, s_0743, s_1434_b, Rate Law: intracellular*Vmax_r_0006*(1/kms_s_0438r_0006)^1*(s_0438^1-s_0743^1*s_1434_b^1/Keq_r_0006)/((1+s_0438/kms_s_0438r_0006+(1+s_0743/kmp_s_0743r_0006)*(1+s_1434_b/kmp_s_1434_br_0006))-1)/intracellular
Keq_r_0721=0.6039; kmp_s_0763_br_0721=0.549; kms_s_1091r_0721=0.549; kms_s_0234r_0721=0.549; Vmax_r_0721=3.30329; kmp_s_0254r_0721=0.549; kmp_s_1096r_0721=0.549Reaction: s_0234 + s_1091 => s_0254 + s_0763_b + s_1096; s_0234, s_0254, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0721*(1/kms_s_0234r_0721)^1*(1/kms_s_1091r_0721)^1*(s_0234^1*s_1091^1-s_0254^1*s_0763_b^1*s_1096^1/Keq_r_0721)/(((1+s_0234/kms_s_0234r_0721)*(1+s_1091/kms_s_1091r_0721)+(1+s_0254/kmp_s_0254r_0721)*(1+s_0763_b/kmp_s_0763_br_0721)*(1+s_1096/kmp_s_1096r_0721))-1)/intracellular
Keq_r_0118=1.1; kmp_s_0374r_0118=0.549; kms_s_0380r_0118=0.549; Vmax_r_0118=0.125399; kmp_s_0514r_0118=0.549Reaction: s_0380 => s_0374 + s_0514; s_0374, s_0380, s_0514, Rate Law: intracellular*Vmax_r_0118*(1/kms_s_0380r_0118)^2*(s_0380^2-s_0374^1*s_0514^1/Keq_r_0118)/((1+s_0380/kms_s_0380r_0118+(1+s_0374/kmp_s_0374r_0118)*(1+s_0514/kmp_s_0514r_0118))-1)/intracellular
kms_s_0446r_0499=1.09208; Keq_r_0499=4.77829; kmp_s_0763_br_0499=0.549; kms_s_0545r_0499=0.0987587; kmp_s_0400r_0499=1.71907; kmp_s_0455r_0499=0.496414; Vmax_r_0499=72.4789Reaction: s_0446 + s_0545 => s_0400 + s_0455 + s_0763_b; s_0400, s_0446, s_0455, s_0545, s_0763_b, Rate Law: intracellular*Vmax_r_0499*(1/kms_s_0446r_0499)^1*(1/kms_s_0545r_0499)^1*(s_0446^1*s_0545^1-s_0400^1*s_0455^1*s_0763_b^1/Keq_r_0499)/(((1+s_0446/kms_s_0446r_0499)*(1+s_0545/kms_s_0545r_0499)+(1+s_0400/kmp_s_0400r_0499)*(1+s_0455/kmp_s_0455r_0499)*(1+s_0763_b/kmp_s_0763_br_0499))-1)/intracellular
Vmax_r_0509=38.2031; kmp_s_0899r_0509=0.549; kms_s_0763_br_0509=0.549; kms_s_1096r_0509=0.549; kmp_s_1091r_0509=0.549; kms_s_0185r_0509=0.549; Keq_r_0509=2.00364; kmp_s_1434_br_0509=0.549; kms_s_0430r_0509=0.549Reaction: s_0185 + s_0430 + s_0763_b + s_1096 => s_0899 + s_1091 + s_1434_b; s_0185, s_0430, s_0763_b, s_0899, s_1091, s_1096, s_1434_b, Rate Law: intracellular*Vmax_r_0509*(1/kms_s_0185r_0509)^1*(1/kms_s_0430r_0509)^1*(1/kms_s_0763_br_0509)^1*(1/kms_s_1096r_0509)^1*(s_0185^1*s_0430^1*s_0763_b^1*s_1096^1-s_0899^1*s_1091^1*s_1434_b^1/Keq_r_0509)/(((1+s_0185/kms_s_0185r_0509)*(1+s_0430/kms_s_0430r_0509)*(1+s_0763_b/kms_s_0763_br_0509)*(1+s_1096/kms_s_1096r_0509)+(1+s_0899/kmp_s_0899r_0509)*(1+s_1091/kmp_s_1091r_0509)*(1+s_1434_b/kmp_s_1434_br_0509))-1)/intracellular
Vmax_r_0890=1.53571; kmp_s_0400r_0890=1.71907; kmp_s_0763_br_0890=0.549; kmp_s_1048r_0890=0.549; kmp_s_1207r_0890=0.549; Keq_r_0890=0.950614; kms_s_0333r_0890=0.549; kms_s_0446r_0890=1.09208; kms_s_0740r_0890=0.549Reaction: s_0333 + s_0446 + s_0740 => s_0400 + s_0763_b + s_1048 + s_1207; s_0333, s_0400, s_0446, s_0740, s_0763_b, s_1048, s_1207, Rate Law: intracellular*Vmax_r_0890*(1/kms_s_0333r_0890)^1*(1/kms_s_0446r_0890)^1*(1/kms_s_0740r_0890)^1*(s_0333^1*s_0446^1*s_0740^1-s_0400^1*s_0763_b^1*s_1048^1*s_1207^1/Keq_r_0890)/(((1+s_0333/kms_s_0333r_0890)*(1+s_0446/kms_s_0446r_0890)*(1+s_0740/kms_s_0740r_0890)+(1+s_0400/kmp_s_0400r_0890)*(1+s_0763_b/kmp_s_0763_br_0890)*(1+s_1048/kmp_s_1048r_0890)*(1+s_1207/kmp_s_1207r_0890))-1)/intracellular
Keq_r_0720=0.6039; kmp_s_1096r_0720=0.549; Vmax_r_0720=3.30329; kms_s_0052r_0720=0.549; kms_s_1091r_0720=0.549; kmp_s_0763_br_0720=0.549; kmp_s_0257r_0720=0.549Reaction: s_0052 + s_1091 => s_0257 + s_0763_b + s_1096; s_0052, s_0257, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0720*(1/kms_s_0052r_0720)^1*(1/kms_s_1091r_0720)^1*(s_0052^1*s_1091^1-s_0257^1*s_0763_b^1*s_1096^1/Keq_r_0720)/(((1+s_0052/kms_s_0052r_0720)*(1+s_1091/kms_s_1091r_0720)+(1+s_0257/kmp_s_0257r_0720)*(1+s_0763_b/kmp_s_0763_br_0720)*(1+s_1096/kmp_s_1096r_0720))-1)/intracellular
kmp_s_0215r_0262=0.549; kmp_s_0470r_0262=1.0; kms_s_0303r_0262=0.549; kmp_s_1087r_0262=0.0867353; Vmax_r_0262=0.0785834; kms_s_1082r_0262=1.50326; kmp_s_0763_br_0262=0.549; Keq_r_0262=0.0348439Reaction: s_0303 + s_1082 => s_0215 + s_0470 + s_0763_b + s_1087; s_0215, s_0303, s_0470, s_0763_b, s_1082, s_1087, Rate Law: intracellular*Vmax_r_0262*(1/kms_s_0303r_0262)^1*(1/kms_s_1082r_0262)^1*(s_0303^1*s_1082^1-s_0215^1*s_0470^1*s_0763_b^1*s_1087^1/Keq_r_0262)/(((1+s_0303/kms_s_0303r_0262)*(1+s_1082/kms_s_1082r_0262)+(1+s_0215/kmp_s_0215r_0262)*(1+s_0470/kmp_s_0470r_0262)*(1+s_0763_b/kmp_s_0763_br_0262)*(1+s_1087/kmp_s_1087r_0262))-1)/intracellular
kms_s_0315r_0604=0.549; kms_s_0907r_0604=0.549; kmp_s_0763_br_0604=0.549; Vmax_r_0604=0.871524; kmp_s_0899r_0604=0.549; kmp_s_0317r_0604=0.549; Keq_r_0604=0.331541; kmp_s_0532r_0604=0.549Reaction: s_0315 + s_0907 => s_0317 + s_0532 + s_0763_b + s_0899; s_0315, s_0317, s_0532, s_0763_b, s_0899, s_0907, Rate Law: intracellular*Vmax_r_0604*(1/kms_s_0315r_0604)^1*(1/kms_s_0907r_0604)^1*(s_0315^1*s_0907^1-s_0317^1*s_0532^1*s_0763_b^1*s_0899^1/Keq_r_0604)/(((1+s_0315/kms_s_0315r_0604)*(1+s_0907/kms_s_0907r_0604)+(1+s_0317/kmp_s_0317r_0604)*(1+s_0532/kmp_s_0532r_0604)*(1+s_0763_b/kmp_s_0763_br_0604)*(1+s_0899/kmp_s_0899r_0604))-1)/intracellular
Keq_r_0064=0.0348439; kms_s_1082r_0064=1.50326; Vmax_r_0064=1.68189; kmp_s_0763_br_0064=0.549; kmp_s_1087r_0064=0.0867353; kms_s_0008r_0064=0.549; kmp_s_0010r_0064=0.549Reaction: s_0008 + s_1082 => s_0010 + s_0763_b + s_1087; s_0008, s_0010, s_0763_b, s_1082, s_1087, Rate Law: intracellular*Vmax_r_0064*(1/kms_s_0008r_0064)^1*(1/kms_s_1082r_0064)^1*(s_0008^1*s_1082^1-s_0010^1*s_0763_b^1*s_1087^1/Keq_r_0064)/(((1+s_0008/kms_s_0008r_0064)*(1+s_1082/kms_s_1082r_0064)+(1+s_0010/kmp_s_0010r_0064)*(1+s_0763_b/kmp_s_0763_br_0064)*(1+s_1087/kmp_s_1087r_0064))-1)/intracellular
Vmax_r_0970=3.3649; kmp_s_0942r_0970=0.549; kms_s_0763_br_0970=0.549; kms_s_0899r_0970=0.549; kmp_s_1434_br_0970=0.549; kms_s_0867r_0970=0.549; kms_s_1096r_0970=0.549; Keq_r_0970=2.00364; kmp_s_1091r_0970=0.549Reaction: s_0763_b + s_0867 + s_0899 + s_1096 => s_0942 + s_1091 + s_1434_b; s_0763_b, s_0867, s_0899, s_0942, s_1091, s_1096, s_1434_b, Rate Law: intracellular*Vmax_r_0970*(1/kms_s_0763_br_0970)^1*(1/kms_s_0867r_0970)^1*(1/kms_s_0899r_0970)^1*(1/kms_s_1096r_0970)^1*(s_0763_b^1*s_0867^1*s_0899^1*s_1096^1-s_0942^1*s_1091^1*s_1434_b^1/Keq_r_0970)/(((1+s_0763_b/kms_s_0763_br_0970)*(1+s_0867/kms_s_0867r_0970)*(1+s_0899/kms_s_0899r_0970)*(1+s_1096/kms_s_1096r_0970)+(1+s_0942/kmp_s_0942r_0970)*(1+s_1091/kmp_s_1091r_0970)*(1+s_1434_b/kmp_s_1434_br_0970))-1)/intracellular
kms_s_1258r_0913=0.549; kmp_s_0209r_0913=0.549; kmp_s_0470r_0913=1.0; Keq_r_0913=1.1; kms_s_1091r_0913=0.549; kmp_s_1096r_0913=0.549; Vmax_r_0913=0.648558Reaction: s_1091 + s_1258 => s_0209 + s_0470 + s_1096; s_0209, s_0470, s_1091, s_1096, s_1258, Rate Law: intracellular*Vmax_r_0913*(1/kms_s_1091r_0913)^1*(1/kms_s_1258r_0913)^1*(s_1091^1*s_1258^1-s_0209^1*s_0470^1*s_1096^1/Keq_r_0913)/(((1+s_1091/kms_s_1091r_0913)*(1+s_1258/kms_s_1258r_0913)+(1+s_0209/kmp_s_0209r_0913)*(1+s_0470/kmp_s_0470r_0913)*(1+s_1096/kmp_s_1096r_0913))-1)/intracellular
kmp_s_0763_br_0526=0.549; Keq_r_0526=2.21027; kmp_s_1096r_0526=0.549; kms_s_1091r_0526=0.549; Vmax_r_0526=5.48128; kmp_s_0734r_0526=0.549; kms_s_0732r_0526=0.15Reaction: s_0732 + s_1091 => s_0734 + s_0763_b + s_1096; s_0732, s_0734, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0526*(1/kms_s_0732r_0526)^1*(1/kms_s_1091r_0526)^1*(s_0732^1*s_1091^1-s_0734^1*s_0763_b^1*s_1096^1/Keq_r_0526)/(((1+s_0732/kms_s_0732r_0526)*(1+s_1091/kms_s_1091r_0526)+(1+s_0734/kmp_s_0734r_0526)*(1+s_0763_b/kmp_s_0763_br_0526)*(1+s_1096/kmp_s_1096r_0526))-1)/intracellular
kms_s_0455r_0504=0.496414; Keq_r_0504=0.29; kmp_s_0539r_0504=0.104555; Vmax_r_0504=6.56505Reaction: s_0455 => s_0539; s_0455, s_0539, Rate Law: intracellular*Vmax_r_0504*(1/kms_s_0455r_0504)^1*(s_0455^1-s_0539^1/Keq_r_0504)/((1+s_0455/kms_s_0455r_0504+1+s_0539/kmp_s_0539r_0504)-1)/intracellular
Vmax_r_0936=0.863944; kmp_s_1091r_0936=0.549; kms_s_0763_br_0936=0.549; Keq_r_0936=3.64962; kms_s_0120r_0936=0.549; kmp_s_0939r_0936=0.549; kms_s_1096r_0936=0.549Reaction: s_0120 + s_0763_b + s_1096 => s_0939 + s_1091; s_0120, s_0763_b, s_0939, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0936*(1/kms_s_0120r_0936)^1*(1/kms_s_0763_br_0936)^2*(1/kms_s_1096r_0936)^1*(s_0120^1*s_0763_b^2*s_1096^1-s_0939^1*s_1091^1/Keq_r_0936)/(((1+s_0120/kms_s_0120r_0936)*(1+s_0763_b/kms_s_0763_br_0936)*(1+s_1096/kms_s_1096r_0936)+(1+s_0939/kmp_s_0939r_0936)*(1+s_1091/kmp_s_1091r_0936))-1)/intracellular
kms_s_0514r_0437=0.549; kmp_s_0434r_0437=1.25956; Keq_r_0437=1.26869; kmp_s_1355r_0437=0.549; kms_s_0987r_0437=0.549; Vmax_r_0437=0.0038115; kms_s_0446r_0437=1.09208; kmp_s_0605r_0437=0.549Reaction: s_0446 + s_0514 + s_0987 => s_0434 + s_0605 + s_1355; s_0434, s_0446, s_0514, s_0605, s_0987, s_1355, Rate Law: intracellular*Vmax_r_0437*(1/kms_s_0446r_0437)^1*(1/kms_s_0514r_0437)^1*(1/kms_s_0987r_0437)^1*(s_0446^1*s_0514^1*s_0987^1-s_0434^1*s_0605^1*s_1355^1/Keq_r_0437)/(((1+s_0446/kms_s_0446r_0437)*(1+s_0514/kms_s_0514r_0437)*(1+s_0987/kms_s_0987r_0437)+(1+s_0434/kmp_s_0434r_0437)*(1+s_0605/kmp_s_0605r_0437)*(1+s_1355/kmp_s_1355r_0437))-1)/intracellular
kms_s_1455r_0266=0.549; kmp_s_1091r_0266=0.549; Keq_r_0266=1.1; kmp_s_1456r_0266=0.549; kmp_s_1434_br_0266=0.549; Vmax_r_0266=0.0951282; kms_s_1160r_0266=0.549; kms_s_1096r_0266=0.549; kms_s_0763_br_0266=0.549Reaction: s_0763_b + s_1096 + s_1160 + s_1455 => s_1091 + s_1434_b + s_1456; s_0763_b, s_1091, s_1096, s_1160, s_1434_b, s_1455, s_1456, Rate Law: intracellular*Vmax_r_0266*(1/kms_s_0763_br_0266)^1*(1/kms_s_1096r_0266)^1*(1/kms_s_1160r_0266)^1*(1/kms_s_1455r_0266)^1*(s_0763_b^1*s_1096^1*s_1160^1*s_1455^1-s_1091^1*s_1434_b^2*s_1456^1/Keq_r_0266)/(((1+s_0763_b/kms_s_0763_br_0266)*(1+s_1096/kms_s_1096r_0266)*(1+s_1160/kms_s_1160r_0266)*(1+s_1455/kms_s_1455r_0266)+(1+s_1091/kmp_s_1091r_0266)*(1+s_1434_b/kmp_s_1434_br_0266)*(1+s_1456/kmp_s_1456r_0266))-1)/intracellular
kmp_s_1207r_0728=0.549; kmp_s_0149r_0728=0.549; kms_s_1070r_0728=0.549; kms_s_0763_br_0728=0.549; Vmax_r_0728=1.2441; Keq_r_0728=1.1; kms_s_1096r_0728=0.549; kmp_s_1091r_0728=0.549Reaction: s_0763_b + s_1070 + s_1096 => s_0149 + s_1091 + s_1207; s_0149, s_0763_b, s_1070, s_1091, s_1096, s_1207, Rate Law: intracellular*Vmax_r_0728*(1/kms_s_0763_br_0728)^1*(1/kms_s_1070r_0728)^1*(1/kms_s_1096r_0728)^1*(s_0763_b^1*s_1070^1*s_1096^1-s_0149^1*s_1091^1*s_1207^1/Keq_r_0728)/(((1+s_0763_b/kms_s_0763_br_0728)*(1+s_1070/kms_s_1070r_0728)*(1+s_1096/kms_s_1096r_0728)+(1+s_0149/kmp_s_0149r_0728)*(1+s_1091/kmp_s_1091r_0728)*(1+s_1207/kmp_s_1207r_0728))-1)/intracellular
kms_s_1209_br_1461=24.5; Keq_r_1461=1.0; kms_s_0766_br_1461=0.1; kmp_s_1207r_1461=0.549; kmp_s_0763_br_1461=0.549; Vmax_r_1461=0.0925906Reaction: s_0766_b + s_1209_b => s_0763_b + s_1207; s_0763_b, s_0766_b, s_1207, s_1209_b, Rate Law: Vmax_r_1461*(1/kms_s_0766_br_1461)^1*(1/kms_s_1209_br_1461)^1*(s_0766_b^1*s_1209_b^1-s_0763_b^1*s_1207^1/Keq_r_1461)/(((1+s_0766_b/kms_s_0766_br_1461)*(1+s_1209_b/kms_s_1209_br_1461)+(1+s_0763_b/kmp_s_0763_br_1461)*(1+s_1207/kmp_s_1207r_1461))-1)
kms_s_0079r_0008=0.549; kmp_s_0315r_0008=0.549; Vmax_r_0008=0.13761; Keq_r_0008=1.1Reaction: s_0079 => s_0315; s_0079, s_0315, Rate Law: intracellular*Vmax_r_0008*(1/kms_s_0079r_0008)^1*(s_0079^1-s_0315^1/Keq_r_0008)/((1+s_0079/kms_s_0079r_0008+1+s_0315/kmp_s_0315r_0008)-1)/intracellular
kms_s_1132r_0417=0.549; kmp_s_0470r_0417=1.0; kmp_s_0574r_0417=0.549; Vmax_r_0417=0.00599719; kmp_s_0514r_0417=0.549; kmp_s_1091r_0417=0.549; kms_s_1005r_0417=0.549; kmp_s_1434_br_0417=0.549; kms_s_0763_br_0417=0.549; kms_s_1096r_0417=0.549; Keq_r_0417=3.64962Reaction: s_0763_b + s_1005 + s_1096 + s_1132 => s_0470 + s_0514 + s_0574 + s_1091 + s_1434_b; s_0470, s_0514, s_0574, s_0763_b, s_1005, s_1091, s_1096, s_1132, s_1434_b, Rate Law: intracellular*Vmax_r_0417*(1/kms_s_0763_br_0417)^3*(1/kms_s_1005r_0417)^1*(1/kms_s_1096r_0417)^2*(1/kms_s_1132r_0417)^1*(s_0763_b^3*s_1005^1*s_1096^2*s_1132^1-s_0470^1*s_0514^1*s_0574^1*s_1091^2*s_1434_b^1/Keq_r_0417)/(((1+s_0763_b/kms_s_0763_br_0417)*(1+s_1005/kms_s_1005r_0417)*(1+s_1096/kms_s_1096r_0417)*(1+s_1132/kms_s_1132r_0417)+(1+s_0470/kmp_s_0470r_0417)*(1+s_0514/kmp_s_0514r_0417)*(1+s_0574/kmp_s_0574r_0417)*(1+s_1091/kmp_s_1091r_0417)*(1+s_1434_b/kmp_s_1434_br_0417))-1)/intracellular
Vmax_r_0352=3.30329; kmp_s_1096r_0352=0.549; kmp_s_0529r_0352=0.549; Keq_r_0352=0.6039; kmp_s_0763_br_0352=0.549; kms_s_0530r_0352=0.549; kms_s_1091r_0352=0.549Reaction: s_0530 + s_1091 => s_0529 + s_0763_b + s_1096; s_0529, s_0530, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0352*(1/kms_s_0530r_0352)^1*(1/kms_s_1091r_0352)^1*(s_0530^1*s_1091^1-s_0529^1*s_0763_b^1*s_1096^1/Keq_r_0352)/(((1+s_0530/kms_s_0530r_0352)*(1+s_1091/kms_s_1091r_0352)+(1+s_0529/kmp_s_0529r_0352)*(1+s_0763_b/kmp_s_0763_br_0352)*(1+s_1096/kmp_s_1096r_0352))-1)/intracellular
kmp_s_1342r_1003=0.549; kms_s_0514r_1003=0.549; kmp_s_1207r_1003=0.549; Keq_r_1003=1.73154; kms_s_0446r_1003=1.09208; Vmax_r_1003=0.13134; kms_s_1338r_1003=0.549; kmp_s_0400r_1003=1.71907Reaction: s_0446 + s_0514 + s_1338 => s_0400 + s_1207 + s_1342; s_0400, s_0446, s_0514, s_1207, s_1338, s_1342, Rate Law: intracellular*Vmax_r_1003*(1/kms_s_0446r_1003)^1*(1/kms_s_0514r_1003)^1*(1/kms_s_1338r_1003)^1*(s_0446^1*s_0514^1*s_1338^1-s_0400^1*s_1207^1*s_1342^1/Keq_r_1003)/(((1+s_0446/kms_s_0446r_1003)*(1+s_0514/kms_s_0514r_1003)*(1+s_1338/kms_s_1338r_1003)+(1+s_0400/kmp_s_0400r_1003)*(1+s_1207/kmp_s_1207r_1003)*(1+s_1342/kmp_s_1342r_1003))-1)/intracellular
kms_s_1434_br_0562=0.549; Vmax_r_0562=0.0104499; kmp_s_0145r_0562=0.549; kmp_s_0689r_0562=0.549; kmp_s_0763_br_0562=0.549; Keq_r_0562=0.6039; kms_s_0755r_0562=0.549; kmp_s_0605r_0562=0.549Reaction: s_0755 + s_1434_b => s_0145 + s_0605 + s_0689 + s_0763_b; s_0145, s_0605, s_0689, s_0755, s_0763_b, s_1434_b, Rate Law: intracellular*Vmax_r_0562*(1/kms_s_0755r_0562)^1*(1/kms_s_1434_br_0562)^3*(s_0755^1*s_1434_b^3-s_0145^1*s_0605^1*s_0689^1*s_0763_b^2/Keq_r_0562)/(((1+s_0755/kms_s_0755r_0562)*(1+s_1434_b/kms_s_1434_br_0562)+(1+s_0145/kmp_s_0145r_0562)*(1+s_0605/kmp_s_0605r_0562)*(1+s_0689/kmp_s_0689r_0562)*(1+s_0763_b/kmp_s_0763_br_0562))-1)/intracellular
kms_s_1434_br_0014=0.549; Vmax_r_0014=0.00605002; Keq_r_0014=2.00364; kmp_s_0430r_0014=0.549; kmp_s_0319r_0014=0.549; kms_s_0763_br_0014=0.549; kms_s_0146r_0014=0.549Reaction: s_0146 + s_0763_b + s_1434_b => s_0319 + s_0430; s_0146, s_0319, s_0430, s_0763_b, s_1434_b, Rate Law: intracellular*Vmax_r_0014*(1/kms_s_0146r_0014)^1*(1/kms_s_0763_br_0014)^1*(1/kms_s_1434_br_0014)^1*(s_0146^1*s_0763_b^1*s_1434_b^1-s_0319^1*s_0430^1/Keq_r_0014)/(((1+s_0146/kms_s_0146r_0014)*(1+s_0763_b/kms_s_0763_br_0014)*(1+s_1434_b/kms_s_1434_br_0014)+(1+s_0319/kmp_s_0319r_0014)*(1+s_0430/kmp_s_0430r_0014))-1)/intracellular
kms_s_0763_br_0060=0.549; Vmax_r_0060=3.30332; kms_s_1087r_0060=0.0867353; kmp_s_0055r_0060=0.549; kms_s_0261r_0060=0.549; Keq_r_0060=34.7263; kmp_s_1082r_0060=1.50326Reaction: s_0261 + s_0763_b + s_1087 => s_0055 + s_1082; s_0055, s_0261, s_0763_b, s_1082, s_1087, Rate Law: intracellular*Vmax_r_0060*(1/kms_s_0261r_0060)^1*(1/kms_s_0763_br_0060)^1*(1/kms_s_1087r_0060)^1*(s_0261^1*s_0763_b^1*s_1087^1-s_0055^1*s_1082^1/Keq_r_0060)/(((1+s_0261/kms_s_0261r_0060)*(1+s_0763_b/kms_s_0763_br_0060)*(1+s_1087/kms_s_1087r_0060)+(1+s_0055/kmp_s_0055r_0060)*(1+s_1082/kmp_s_1082r_0060))-1)/intracellular
kmp_s_0511r_0847=0.549; kms_s_0485r_0847=0.549; kmp_s_0763_br_0847=0.549; kmp_s_0090r_0847=0.549; Vmax_r_0847=0.010285; kms_s_1020r_0847=0.549; Keq_r_0847=0.331541Reaction: s_0485 + s_1020 => s_0090 + s_0511 + s_0763_b; s_0090, s_0485, s_0511, s_0763_b, s_1020, Rate Law: intracellular*Vmax_r_0847*(1/kms_s_0485r_0847)^1*(1/kms_s_1020r_0847)^1*(s_0485^1*s_1020^1-s_0090^1*s_0511^1*s_0763_b^2/Keq_r_0847)/(((1+s_0485/kms_s_0485r_0847)*(1+s_1020/kms_s_1020r_0847)+(1+s_0090/kmp_s_0090r_0847)*(1+s_0511/kmp_s_0511r_0847)*(1+s_0763_b/kmp_s_0763_br_0847))-1)/intracellular
Vmax_r_0290=0.00279509; kms_s_1325r_0290=0.549; kmp_s_0763_br_0290=0.549; kmp_s_0514r_0290=0.549; kmp_s_1080r_0290=0.549; Keq_r_0290=0.6039; kms_s_1355r_0290=0.549Reaction: s_1325 + s_1355 => s_0514 + s_0763_b + s_1080; s_0514, s_0763_b, s_1080, s_1325, s_1355, Rate Law: intracellular*Vmax_r_0290*(1/kms_s_1325r_0290)^1*(1/kms_s_1355r_0290)^1*(s_1325^1*s_1355^1-s_0514^1*s_0763_b^1*s_1080^1/Keq_r_0290)/(((1+s_1325/kms_s_1325r_0290)*(1+s_1355/kms_s_1355r_0290)+(1+s_0514/kmp_s_0514r_0290)*(1+s_0763_b/kmp_s_0763_br_0290)*(1+s_1080/kmp_s_1080r_0290))-1)/intracellular
Keq_r_1293=1.0; kms_s_0547_br_1293=11.1; Vmax_r_1293=2.36101; kmp_s_0545r_1293=0.0987587Reaction: s_0547_b => s_0545; s_0545, s_0547_b, Rate Law: Vmax_r_1293*(1/kms_s_0547_br_1293)^1*(s_0547_b^1-s_0545^1/Keq_r_1293)/((1+s_0547_b/kms_s_0547_br_1293+1+s_0545/kmp_s_0545r_1293)-1)
kmp_s_0763_br_0853=0.549; kms_s_0943r_0853=0.549; Keq_r_0853=0.331541; kmp_s_1219r_0853=0.549; kmp_s_0511r_0853=0.549; kms_s_0485r_0853=0.549; Vmax_r_0853=0.0266199Reaction: s_0485 + s_0943 => s_0511 + s_0763_b + s_1219; s_0485, s_0511, s_0763_b, s_0943, s_1219, Rate Law: intracellular*Vmax_r_0853*(1/kms_s_0485r_0853)^1*(1/kms_s_0943r_0853)^1*(s_0485^1*s_0943^1-s_0511^1*s_0763_b^2*s_1219^1/Keq_r_0853)/(((1+s_0485/kms_s_0485r_0853)*(1+s_0943/kms_s_0943r_0853)+(1+s_0511/kmp_s_0511r_0853)*(1+s_0763_b/kmp_s_0763_br_0853)*(1+s_1219/kmp_s_1219r_0853))-1)/intracellular
kms_s_1096r_0598=0.549; kmp_s_1091r_0598=0.549; kmp_s_0031r_0598=0.549; kmp_s_0514r_0598=0.549; kms_s_0225r_0598=0.549; Keq_r_0598=2.00364; Vmax_r_0598=0.3762; kms_s_0763_br_0598=0.549Reaction: s_0225 + s_0763_b + s_1096 => s_0031 + s_0514 + s_1091; s_0031, s_0225, s_0514, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0598*(1/kms_s_0225r_0598)^1*(1/kms_s_0763_br_0598)^2*(1/kms_s_1096r_0598)^2*(s_0225^1*s_0763_b^2*s_1096^2-s_0031^1*s_0514^1*s_1091^2/Keq_r_0598)/(((1+s_0225/kms_s_0225r_0598)*(1+s_0763_b/kms_s_0763_br_0598)*(1+s_1096/kms_s_1096r_0598)+(1+s_0031/kmp_s_0031r_0598)*(1+s_0514/kmp_s_0514r_0598)*(1+s_1091/kmp_s_1091r_0598))-1)/intracellular
kmp_s_1087r_0940=0.0867353; kms_s_0514r_0940=0.549; Vmax_r_0940=9.4545; kmp_s_0470r_0940=1.0; kmp_s_0380r_0940=0.549; kms_s_1277r_0940=0.0605905; Keq_r_0940=1.04749; kms_s_1082r_0940=1.50326Reaction: s_0514 + s_1082 + s_1277 => s_0380 + s_0470 + s_1087; s_0380, s_0470, s_0514, s_1082, s_1087, s_1277, Rate Law: intracellular*Vmax_r_0940*(1/kms_s_0514r_0940)^1*(1/kms_s_1082r_0940)^1*(1/kms_s_1277r_0940)^1*(s_0514^1*s_1082^1*s_1277^1-s_0380^1*s_0470^1*s_1087^1/Keq_r_0940)/(((1+s_0514/kms_s_0514r_0940)*(1+s_1082/kms_s_1082r_0940)*(1+s_1277/kms_s_1277r_0940)+(1+s_0380/kmp_s_0380r_0940)*(1+s_0470/kmp_s_0470r_0940)*(1+s_1087/kmp_s_1087r_0940))-1)/intracellular
kmp_s_1306r_0976=0.549; kms_s_1096r_0976=0.549; kms_s_0217r_0976=0.549; kms_s_0763_br_0976=0.549; Vmax_r_0976=1.60931; Keq_r_0976=2.00364; kmp_s_1091r_0976=0.549Reaction: s_0217 + s_0763_b + s_1096 => s_1091 + s_1306; s_0217, s_0763_b, s_1091, s_1096, s_1306, Rate Law: intracellular*Vmax_r_0976*(1/kms_s_0217r_0976)^1*(1/kms_s_0763_br_0976)^1*(1/kms_s_1096r_0976)^1*(s_0217^1*s_0763_b^1*s_1096^1-s_1091^1*s_1306^1/Keq_r_0976)/(((1+s_0217/kms_s_0217r_0976)*(1+s_0763_b/kms_s_0763_br_0976)*(1+s_1096/kms_s_1096r_0976)+(1+s_1091/kmp_s_1091r_0976)*(1+s_1306/kmp_s_1306r_0976))-1)/intracellular
kms_s_0380r_0127=0.549; kmp_s_0514r_0127=0.549; kms_s_0434r_0127=1.25956; Vmax_r_0127=25.905; Keq_r_0127=0.953736; kmp_s_0369r_0127=0.549; kmp_s_0446r_0127=1.09208; kms_s_0605r_0127=0.549Reaction: s_0380 + s_0434 + s_0605 => s_0369 + s_0446 + s_0514; s_0369, s_0380, s_0434, s_0446, s_0514, s_0605, Rate Law: intracellular*Vmax_r_0127*(1/kms_s_0380r_0127)^1*(1/kms_s_0434r_0127)^1*(1/kms_s_0605r_0127)^1*(s_0380^1*s_0434^1*s_0605^1-s_0369^1*s_0446^1*s_0514^1/Keq_r_0127)/(((1+s_0380/kms_s_0380r_0127)*(1+s_0434/kms_s_0434r_0127)*(1+s_0605/kms_s_0605r_0127)+(1+s_0369/kmp_s_0369r_0127)*(1+s_0446/kmp_s_0446r_0127)*(1+s_0514/kmp_s_0514r_0127))-1)/intracellular
kms_s_0763_br_0125=0.549; Keq_r_0125=2.00364; kms_s_0369r_0125=0.549; kms_s_0514r_0125=0.549; Vmax_r_0125=26.9831; kmp_s_0380r_0125=0.549; kmp_s_1434_br_0125=0.549Reaction: s_0369 + s_0514 + s_0763_b => s_0380 + s_1434_b; s_0369, s_0380, s_0514, s_0763_b, s_1434_b, Rate Law: intracellular*Vmax_r_0125*(1/kms_s_0369r_0125)^1*(1/kms_s_0514r_0125)^1*(1/kms_s_0763_br_0125)^1*(s_0369^1*s_0514^1*s_0763_b^1-s_0380^1*s_1434_b^1/Keq_r_0125)/(((1+s_0369/kms_s_0369r_0125)*(1+s_0514/kms_s_0514r_0125)*(1+s_0763_b/kms_s_0763_br_0125)+(1+s_0380/kmp_s_0380r_0125)*(1+s_1434_b/kmp_s_1434_br_0125))-1)/intracellular
kmp_s_1207r_0934=0.549; kms_s_0319r_0934=0.549; Vmax_r_0934=0.00385; kmp_s_0320r_0934=0.549; kms_s_1434_br_0934=0.549; Keq_r_0934=1.1Reaction: s_0319 + s_1434_b => s_0320 + s_1207; s_0319, s_0320, s_1207, s_1434_b, Rate Law: intracellular*Vmax_r_0934*(1/kms_s_0319r_0934)^1*(1/kms_s_1434_br_0934)^1*(s_0319^1*s_1434_b^1-s_0320^1*s_1207^1/Keq_r_0934)/(((1+s_0319/kms_s_0319r_0934)*(1+s_1434_b/kms_s_1434_br_0934)+(1+s_0320/kmp_s_0320r_0934)*(1+s_1207/kmp_s_1207r_0934))-1)/intracellular
Vmax_r_0264=0.0454962; kms_s_1458r_0264=0.549; Keq_r_0264=2.00364; kmp_s_1091r_0264=0.549; kms_s_0763_br_0264=0.549; kms_s_1096r_0264=0.549; kmp_s_1447r_0264=0.549Reaction: s_0763_b + s_1096 + s_1458 => s_1091 + s_1447; s_0763_b, s_1091, s_1096, s_1447, s_1458, Rate Law: intracellular*Vmax_r_0264*(1/kms_s_0763_br_0264)^1*(1/kms_s_1096r_0264)^1*(1/kms_s_1458r_0264)^1*(s_0763_b^1*s_1096^1*s_1458^1-s_1091^1*s_1447^1/Keq_r_0264)/(((1+s_0763_b/kms_s_0763_br_0264)*(1+s_1096/kms_s_1096r_0264)*(1+s_1458/kms_s_1458r_0264)+(1+s_1091/kmp_s_1091r_0264)*(1+s_1447/kmp_s_1447r_0264))-1)/intracellular
kms_s_0400r_0362=1.71907; kmp_s_0446r_0362=1.09208; Keq_r_0362=0.698801; Vmax_r_0362=0.010395; kms_s_0591r_0362=0.549; kmp_s_0593r_0362=0.549Reaction: s_0400 + s_0591 => s_0446 + s_0593; s_0400, s_0446, s_0591, s_0593, Rate Law: intracellular*Vmax_r_0362*(1/kms_s_0400r_0362)^1*(1/kms_s_0591r_0362)^1*(s_0400^1*s_0591^1-s_0446^1*s_0593^1/Keq_r_0362)/(((1+s_0400/kms_s_0400r_0362)*(1+s_0591/kms_s_0591r_0362)+(1+s_0446/kmp_s_0446r_0362)*(1+s_0593/kmp_s_0593r_0362))-1)/intracellular
Keq_r_0213=0.6039; kmp_s_0763_br_0213=0.549; kms_s_0410r_0213=0.549; kmp_s_0419r_0213=0.549; kmp_s_1411r_0213=0.549; kms_s_1415r_0213=0.549; Vmax_r_0213=0.157299Reaction: s_0410 + s_1415 => s_0419 + s_0763_b + s_1411; s_0410, s_0419, s_0763_b, s_1411, s_1415, Rate Law: intracellular*Vmax_r_0213*(1/kms_s_0410r_0213)^1*(1/kms_s_1415r_0213)^1*(s_0410^1*s_1415^1-s_0419^1*s_0763_b^1*s_1411^1/Keq_r_0213)/(((1+s_0410/kms_s_0410r_0213)*(1+s_1415/kms_s_1415r_0213)+(1+s_0419/kmp_s_0419r_0213)*(1+s_0763_b/kmp_s_0763_br_0213)*(1+s_1411/kmp_s_1411r_0213))-1)/intracellular
Vmax_r_0328=13.2165; kms_s_1434_br_0328=0.549; Keq_r_0328=1.1; kmp_s_0507r_0328=0.549; kmp_s_0763_br_0328=0.549; kms_s_1156r_0328=0.549; kmp_s_0514r_0328=0.549; kms_s_0380r_0328=0.549Reaction: s_0380 + s_1156 + s_1434_b => s_0507 + s_0514 + s_0763_b; s_0380, s_0507, s_0514, s_0763_b, s_1156, s_1434_b, Rate Law: intracellular*Vmax_r_0328*(1/kms_s_0380r_0328)^1*(1/kms_s_1156r_0328)^1*(1/kms_s_1434_br_0328)^1*(s_0380^1*s_1156^1*s_1434_b^1-s_0507^1*s_0514^1*s_0763_b^1/Keq_r_0328)/(((1+s_0380/kms_s_0380r_0328)*(1+s_1156/kms_s_1156r_0328)*(1+s_1434_b/kms_s_1434_br_0328)+(1+s_0507/kmp_s_0507r_0328)*(1+s_0514/kmp_s_0514r_0328)*(1+s_0763_b/kmp_s_0763_br_0328))-1)/intracellular
Keq_r_0111=2.00364; Vmax_r_0111=3.41221; kms_s_0763_br_0111=0.549; kmp_s_1091r_0111=0.549; kms_s_0150r_0111=0.549; kms_s_1096r_0111=0.549; kmp_s_0018r_0111=0.549Reaction: s_0150 + s_0763_b + s_1096 => s_0018 + s_1091; s_0018, s_0150, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0111*(1/kms_s_0150r_0111)^1*(1/kms_s_0763_br_0111)^1*(1/kms_s_1096r_0111)^1*(s_0150^1*s_0763_b^1*s_1096^1-s_0018^1*s_1091^1/Keq_r_0111)/(((1+s_0150/kms_s_0150r_0111)*(1+s_0763_b/kms_s_0763_br_0111)*(1+s_1096/kms_s_1096r_0111)+(1+s_0018/kmp_s_0018r_0111)*(1+s_1091/kmp_s_1091r_0111))-1)/intracellular
a_s_0873r_1812=0.13579; a_s_0949r_1812=0.19653; s_0743_or_1812=0.549; s_0960_or_1812=1.0; s_1283_or_1812=0.549; a_s_0434r_1812=0.051; a_s_0593r_1812=0.002432; s_0936_or_1812=0.549; s_1011_or_1812=0.549; a_s_0564r_1812=0.003587; s_0929_or_1812=0.549; a_s_0943r_1812=0.25371; a_s_0960r_1812=0.25728; a_s_1000r_1812=1.0; a_s_0933r_1812=0.050027; s_0619_or_1812=0.549; a_s_0416r_1812=0.023371; s_0511_or_1812=0.549; s_0920_or_1812=0.549; a_s_0955r_1812=0.096481; s_0889_or_1812=0.549; a_s_0743r_1812=0.51852; a_s_0752r_1812=0.051; a_s_0925r_1812=0.25014; s_0949_or_1812=1.0; s_0863_or_1812=0.549; s_0907_or_1812=0.549; a_s_0907r_1812=0.268; s_0939_or_1812=0.549; a_s_0001r_1812=1.1358; a_s_1011r_1812=0.82099; s_0899_or_1812=0.549; s_0955_or_1812=0.549; s_1347_or_1812=0.549; a_s_0446r_1812=59.276; s_0952_or_1812=1.0; s_0416_or_1812=0.549; a_s_0929r_1812=0.23942; s_0881_or_1812=0.549; a_s_0899r_1812=0.268; s_0873_or_1812=0.549; s_0569_or_1812=0.549; s_0593_or_1812=0.549; V_o=0.0555; a_s_0939r_1812=0.12864; a_s_0881r_1812=0.17152; a_s_0569r_1812=0.002432; s_0877_or_1812=0.549; a_s_0863r_1812=0.35734; a_s_0952r_1812=0.028; a_s_1347r_1812=0.02; s_0933_or_1812=0.549; s_0564_or_1812=0.549; s_0925_or_1812=0.549; a_s_0877r_1812=0.17152; s_1000_or_1812=0.549; a_s_0911r_1812=0.075041; s_0740_or_1812=0.549; s_0752_or_1812=0.549; zero_flux=0.0; a_s_1417r_1812=0.067; s_0001_or_1812=0.549; s_0943_or_1812=0.549; a_s_0920r_1812=0.17152; a_s_0619r_1812=0.003587; a_s_0740r_1812=0.32518; a_s_0511r_1812=0.05; s_1417_or_1812=0.549; a_s_0889r_1812=0.04288; a_s_0936r_1812=0.11435; s_0434_or_1812=1.25956; a_s_1283r_1812=9.0E-4; s_0446_or_1812=1.09208; s_0911_or_1812=0.549Reaction: s_0001 + s_0416 + s_0434 + s_0446 + s_0511 + s_0564 + s_0569 + s_0593 + s_0619 + s_0740 + s_0743 + s_0752 + s_0863 + s_0873 + s_0877 + s_0881 + s_0889 + s_0899 + s_0907 + s_0911 + s_0920 + s_0925 + s_0929 + s_0933 + s_0936 + s_0939 + s_0943 + s_0949 + s_0952 + s_0955 + s_0960 + s_1000 + s_1011 + s_1347 + s_1417 + s_1283 => s_0400 + s_0463 + s_1207; s_0547_b, s_0001, s_0416, s_0434, s_0446, s_0511, s_0564, s_0569, s_0593, s_0619, s_0740, s_0743, s_0752, s_0863, s_0873, s_0877, s_0881, s_0889, s_0899, s_0907, s_0911, s_0920, s_0925, s_0929, s_0933, s_0936, s_0939, s_0943, s_0949, s_0952, s_0955, s_0960, s_1000, s_1011, s_1283, s_1347, s_1417, Rate Law: intracellular*piecewise(V_o*(1+a_s_0001r_1812*ln(s_0001/s_0001_or_1812)+a_s_0416r_1812*ln(s_0416/s_0416_or_1812)+a_s_0434r_1812*ln(s_0434/s_0434_or_1812)+a_s_0446r_1812*ln(s_0446/s_0446_or_1812)+a_s_0511r_1812*ln(s_0511/s_0511_or_1812)+a_s_0564r_1812*ln(s_0564/s_0564_or_1812)+a_s_0569r_1812*ln(s_0569/s_0569_or_1812)+a_s_0593r_1812*ln(s_0593/s_0593_or_1812)+a_s_0619r_1812*ln(s_0619/s_0619_or_1812)+a_s_0740r_1812*ln(s_0740/s_0740_or_1812)+a_s_0743r_1812*ln(s_0743/s_0743_or_1812)+a_s_0752r_1812*ln(s_0752/s_0752_or_1812)+a_s_0863r_1812*ln(s_0863/s_0863_or_1812)+a_s_0873r_1812*ln(s_0873/s_0873_or_1812)+a_s_0877r_1812*ln(s_0877/s_0877_or_1812)+a_s_0881r_1812*ln(s_0881/s_0881_or_1812)+a_s_0889r_1812*ln(s_0889/s_0889_or_1812)+a_s_0899r_1812*ln(s_0899/s_0899_or_1812)+a_s_0907r_1812*ln(s_0907/s_0907_or_1812)+a_s_0911r_1812*ln(s_0911/s_0911_or_1812)+a_s_0920r_1812*ln(s_0920/s_0920_or_1812)+a_s_0925r_1812*ln(s_0925/s_0925_or_1812)+a_s_0929r_1812*ln(s_0929/s_0929_or_1812)+a_s_0933r_1812*ln(s_0933/s_0933_or_1812)+a_s_0936r_1812*ln(s_0936/s_0936_or_1812)+a_s_0939r_1812*ln(s_0939/s_0939_or_1812)+a_s_0943r_1812*ln(s_0943/s_0943_or_1812)+a_s_0949r_1812*ln(s_0949/s_0949_or_1812)+a_s_0952r_1812*ln(s_0952/s_0952_or_1812)+a_s_0955r_1812*ln(s_0955/s_0955_or_1812)+a_s_0960r_1812*ln(s_0960/s_0960_or_1812)+a_s_1000r_1812*ln(s_1000/s_1000_or_1812)+a_s_1011r_1812*ln(s_1011/s_1011_or_1812)+a_s_1347r_1812*ln(s_1347/s_1347_or_1812)+a_s_1417r_1812*ln(s_1417/s_1417_or_1812)+a_s_1283r_1812*ln(s_1283/s_1283_or_1812)), (V_o*(1+a_s_0001r_1812*ln(s_0001/s_0001_or_1812)+a_s_0416r_1812*ln(s_0416/s_0416_or_1812)+a_s_0434r_1812*ln(s_0434/s_0434_or_1812)+a_s_0446r_1812*ln(s_0446/s_0446_or_1812)+a_s_0511r_1812*ln(s_0511/s_0511_or_1812)+a_s_0564r_1812*ln(s_0564/s_0564_or_1812)+a_s_0569r_1812*ln(s_0569/s_0569_or_1812)+a_s_0593r_1812*ln(s_0593/s_0593_or_1812)+a_s_0619r_1812*ln(s_0619/s_0619_or_1812)+a_s_0740r_1812*ln(s_0740/s_0740_or_1812)+a_s_0743r_1812*ln(s_0743/s_0743_or_1812)+a_s_0752r_1812*ln(s_0752/s_0752_or_1812)+a_s_0863r_1812*ln(s_0863/s_0863_or_1812)+a_s_0873r_1812*ln(s_0873/s_0873_or_1812)+a_s_0877r_1812*ln(s_0877/s_0877_or_1812)+a_s_0881r_1812*ln(s_0881/s_0881_or_1812)+a_s_0889r_1812*ln(s_0889/s_0889_or_1812)+a_s_0899r_1812*ln(s_0899/s_0899_or_1812)+a_s_0907r_1812*ln(s_0907/s_0907_or_1812)+a_s_0911r_1812*ln(s_0911/s_0911_or_1812)+a_s_0920r_1812*ln(s_0920/s_0920_or_1812)+a_s_0925r_1812*ln(s_0925/s_0925_or_1812)+a_s_0929r_1812*ln(s_0929/s_0929_or_1812)+a_s_0933r_1812*ln(s_0933/s_0933_or_1812)+a_s_0936r_1812*ln(s_0936/s_0936_or_1812)+a_s_0939r_1812*ln(s_0939/s_0939_or_1812)+a_s_0943r_1812*ln(s_0943/s_0943_or_1812)+a_s_0949r_1812*ln(s_0949/s_0949_or_1812)+a_s_0952r_1812*ln(s_0952/s_0952_or_1812)+a_s_0955r_1812*ln(s_0955/s_0955_or_1812)+a_s_0960r_1812*ln(s_0960/s_0960_or_1812)+a_s_1000r_1812*ln(s_1000/s_1000_or_1812)+a_s_1011r_1812*ln(s_1011/s_1011_or_1812)+a_s_1347r_1812*ln(s_1347/s_1347_or_1812)+a_s_1417r_1812*ln(s_1417/s_1417_or_1812)+a_s_1283r_1812*ln(s_1283/s_1283_or_1812))) >= zero_flux, zero_flux)/intracellular
kmp_s_1434_br_0418=0.549; Vmax_r_0418=0.00599719; kmp_s_1091r_0418=0.549; kms_s_0763_br_0418=0.549; kmp_s_0514r_0418=0.549; kms_s_1096r_0418=0.549; Keq_r_0418=3.64962; kmp_s_0470r_0418=1.0; kmp_s_0968r_0418=0.549; kms_s_1005r_0418=0.549; kms_s_0574r_0418=0.549Reaction: s_0574 + s_0763_b + s_1005 + s_1096 => s_0470 + s_0514 + s_0968 + s_1091 + s_1434_b; s_0470, s_0514, s_0574, s_0763_b, s_0968, s_1005, s_1091, s_1096, s_1434_b, Rate Law: intracellular*Vmax_r_0418*(1/kms_s_0574r_0418)^1*(1/kms_s_0763_br_0418)^3*(1/kms_s_1005r_0418)^1*(1/kms_s_1096r_0418)^2*(s_0574^1*s_0763_b^3*s_1005^1*s_1096^2-s_0470^1*s_0514^1*s_0968^1*s_1091^2*s_1434_b^1/Keq_r_0418)/(((1+s_0574/kms_s_0574r_0418)*(1+s_0763_b/kms_s_0763_br_0418)*(1+s_1005/kms_s_1005r_0418)*(1+s_1096/kms_s_1096r_0418)+(1+s_0470/kmp_s_0470r_0418)*(1+s_0514/kmp_s_0514r_0418)*(1+s_0968/kmp_s_0968r_0418)*(1+s_1091/kmp_s_1091r_0418)*(1+s_1434_b/kmp_s_1434_br_0418))-1)/intracellular
Vmax_r_0015=0.00605002; kmp_s_0146r_0015=0.549; kms_s_1096r_0015=0.549; Keq_r_0015=2.00364; kms_s_0145r_0015=0.549; kmp_s_1091r_0015=0.549; kms_s_0763_br_0015=0.549Reaction: s_0145 + s_0763_b + s_1096 => s_0146 + s_1091; s_0145, s_0146, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0015*(1/kms_s_0145r_0015)^1*(1/kms_s_0763_br_0015)^1*(1/kms_s_1096r_0015)^1*(s_0145^1*s_0763_b^1*s_1096^1-s_0146^1*s_1091^1/Keq_r_0015)/(((1+s_0145/kms_s_0145r_0015)*(1+s_0763_b/kms_s_0763_br_0015)*(1+s_1096/kms_s_1096r_0015)+(1+s_0146/kmp_s_0146r_0015)*(1+s_1091/kmp_s_1091r_0015))-1)/intracellular
Keq_r_0794=2.00364; kms_s_0763_br_0794=0.549; kmp_s_1417r_0794=0.549; Vmax_r_0794=0.52591; kmp_s_0470r_0794=1.0; kms_s_1155r_0794=0.549Reaction: s_0763_b + s_1155 => s_0470 + s_1417; s_0470, s_0763_b, s_1155, s_1417, Rate Law: intracellular*Vmax_r_0794*(1/kms_s_0763_br_0794)^1*(1/kms_s_1155r_0794)^1*(s_0763_b^1*s_1155^1-s_0470^1*s_1417^1/Keq_r_0794)/(((1+s_0763_b/kms_s_0763_br_0794)*(1+s_1155/kms_s_1155r_0794)+(1+s_0470/kmp_s_0470r_0794)*(1+s_1417/kmp_s_1417r_0794))-1)/intracellular
Keq_r_0793=1.1; kmp_s_1155r_0793=0.549; kms_s_0331r_0793=0.549; kmp_s_0605r_0793=0.549; kms_s_1154r_0793=0.549; Vmax_r_0793=0.52591Reaction: s_0331 + s_1154 => s_0605 + s_1155; s_0331, s_0605, s_1154, s_1155, Rate Law: intracellular*Vmax_r_0793*(1/kms_s_0331r_0793)^1*(1/kms_s_1154r_0793)^1*(s_0331^1*s_1154^1-s_0605^1*s_1155^1/Keq_r_0793)/(((1+s_0331/kms_s_0331r_0793)*(1+s_1154/kms_s_1154r_0793)+(1+s_0605/kmp_s_0605r_0793)*(1+s_1155/kmp_s_1155r_0793))-1)/intracellular
kms_s_0798r_0029=0.549; Keq_r_0029=0.6039; kmp_s_0468r_0029=0.549; kmp_s_1434_br_0029=0.549; Vmax_r_0029=0.731496Reaction: s_0798 => s_0468 + s_1434_b; s_0468, s_0798, s_1434_b, Rate Law: intracellular*Vmax_r_0029*(1/kms_s_0798r_0029)^1*(s_0798^1-s_0468^1*s_1434_b^1/Keq_r_0029)/((1+s_0798/kms_s_0798r_0029+(1+s_0468/kmp_s_0468r_0029)*(1+s_1434_b/kmp_s_1434_br_0029))-1)/intracellular
kmp_s_0514r_0442=0.549; kmp_s_1132r_0442=0.549; Vmax_r_0442=0.001914; kms_s_0605r_0442=0.549; kmp_s_0446r_0442=1.09208; kms_s_0434r_0442=1.25956; kms_s_1140r_0442=0.549; Keq_r_0442=0.953736Reaction: s_0434 + s_0605 + s_1140 => s_0446 + s_0514 + s_1132; s_0434, s_0446, s_0514, s_0605, s_1132, s_1140, Rate Law: intracellular*Vmax_r_0442*(1/kms_s_0434r_0442)^1*(1/kms_s_0605r_0442)^1*(1/kms_s_1140r_0442)^1*(s_0434^1*s_0605^1*s_1140^1-s_0446^1*s_0514^1*s_1132^1/Keq_r_0442)/(((1+s_0434/kms_s_0434r_0442)*(1+s_0605/kms_s_0605r_0442)*(1+s_1140/kms_s_1140r_0442)+(1+s_0446/kmp_s_0446r_0442)*(1+s_0514/kmp_s_0514r_0442)*(1+s_1132/kmp_s_1132r_0442))-1)/intracellular
Keq_r_0887=1.1; Vmax_r_0887=0.05115; kms_s_1066r_0887=0.549; kmp_s_0078r_0887=0.549Reaction: s_1066 => s_0078; s_0078, s_1066, Rate Law: intracellular*Vmax_r_0887*(1/kms_s_1066r_0887)^1*(s_1066^1-s_0078^1/Keq_r_0887)/((1+s_1066/kms_s_1066r_0887+1+s_0078/kmp_s_0078r_0887)-1)/intracellular
Vmax_r_1194=2.37902; kmp_s_0472_br_1194=1.0E-5; Keq_r_1194=1.0; kms_s_0470r_1194=1.0Reaction: s_0470 => s_0472_b; s_0470, s_0472_b, Rate Law: Vmax_r_1194*(1/kms_s_0470r_1194)^1*(s_0470^1-s_0472_b^1/Keq_r_1194)/((1+s_0470/kms_s_0470r_1194+1+s_0472_b/kmp_s_0472_br_1194)-1)
kmp_s_0400r_1059=1.71907; Vmax_r_1059=0.23947; kmp_s_1411r_1059=0.549; Keq_r_1059=1.73154; kms_s_0446r_1059=1.09208; kms_s_1417r_1059=0.549Reaction: s_0446 + s_1417 => s_0400 + s_1411; s_0400, s_0446, s_1411, s_1417, Rate Law: intracellular*Vmax_r_1059*(1/kms_s_0446r_1059)^1*(1/kms_s_1417r_1059)^1*(s_0446^1*s_1417^1-s_0400^1*s_1411^1/Keq_r_1059)/(((1+s_0446/kms_s_0446r_1059)*(1+s_1417/kms_s_1417r_1059)+(1+s_0400/kmp_s_0400r_1059)*(1+s_1411/kmp_s_1411r_1059))-1)/intracellular
kmp_s_0564r_0360=0.549; kmp_s_0446r_0360=1.09208; Keq_r_0360=0.698801; Vmax_r_0360=0.015323; kms_s_0400r_0360=1.71907; kms_s_0562r_0360=0.549Reaction: s_0400 + s_0562 => s_0446 + s_0564; s_0400, s_0446, s_0562, s_0564, Rate Law: intracellular*Vmax_r_0360*(1/kms_s_0400r_0360)^1*(1/kms_s_0562r_0360)^1*(s_0400^1*s_0562^1-s_0446^1*s_0564^1/Keq_r_0360)/(((1+s_0400/kms_s_0400r_0360)*(1+s_0562/kms_s_0562r_0360)+(1+s_0446/kmp_s_0446r_0360)*(1+s_0564/kmp_s_0564r_0360))-1)/intracellular
kms_s_0763_br_0112=0.549; kmp_s_0470r_0112=1.0; kmp_s_0150r_0112=0.549; Keq_r_0112=299.629; Vmax_r_0112=2.1714; kms_s_1277r_0112=0.0605905Reaction: s_0763_b + s_1277 => s_0150 + s_0470; s_0150, s_0470, s_0763_b, s_1277, Rate Law: intracellular*Vmax_r_0112*(1/kms_s_0763_br_0112)^1*(1/kms_s_1277r_0112)^2*(s_0763_b^1*s_1277^2-s_0150^1*s_0470^1/Keq_r_0112)/(((1+s_0763_b/kms_s_0763_br_0112)*(1+s_1277/kms_s_1277r_0112)+(1+s_0150/kmp_s_0150r_0112)*(1+s_0470/kmp_s_0470r_0112))-1)/intracellular
Keq_r_0271=2.00364; kms_s_1096r_0271=0.549; kmp_s_1091r_0271=0.549; kms_s_0632r_0271=0.549; Vmax_r_0271=0.0430762; kms_s_0763_br_0271=0.549; kmp_s_0635r_0271=0.549Reaction: s_0632 + s_0763_b + s_1096 => s_0635 + s_1091; s_0632, s_0635, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0271*(1/kms_s_0632r_0271)^1*(1/kms_s_0763_br_0271)^1*(1/kms_s_1096r_0271)^1*(s_0632^1*s_0763_b^1*s_1096^1-s_0635^1*s_1091^1/Keq_r_0271)/(((1+s_0632/kms_s_0632r_0271)*(1+s_0763_b/kms_s_0763_br_0271)*(1+s_1096/kms_s_1096r_0271)+(1+s_0635/kmp_s_0635r_0271)*(1+s_1091/kmp_s_1091r_0271))-1)/intracellular
Keq_r_0633=0.6039; Vmax_r_0633=1.22649; kmp_s_1338r_0633=0.549; kmp_s_0749r_0633=0.549; kms_s_0847r_0633=0.549Reaction: s_0847 => s_0749 + s_1338; s_0749, s_0847, s_1338, Rate Law: intracellular*Vmax_r_0633*(1/kms_s_0847r_0633)^1*(s_0847^1-s_0749^1*s_1338^1/Keq_r_0633)/((1+s_0847/kms_s_0847r_0633+(1+s_0749/kmp_s_0749r_0633)*(1+s_1338/kmp_s_1338r_0633))-1)/intracellular
kmp_s_0514r_0009=0.549; Keq_r_0009=0.0999269; kms_s_0083r_0009=0.549; kmp_s_0763_br_0009=0.549; Vmax_r_0009=0.0421078; kmp_s_1215r_0009=0.549; kms_s_0386r_0009=0.549Reaction: s_0083 + s_0386 => s_0514 + s_0763_b + s_1215; s_0083, s_0386, s_0514, s_0763_b, s_1215, Rate Law: intracellular*Vmax_r_0009*(1/kms_s_0083r_0009)^1*(1/kms_s_0386r_0009)^1*(s_0083^1*s_0386^1-s_0514^1*s_0763_b^4*s_1215^1/Keq_r_0009)/(((1+s_0083/kms_s_0083r_0009)*(1+s_0386/kms_s_0386r_0009)+(1+s_0514/kmp_s_0514r_0009)*(1+s_0763_b/kmp_s_0763_br_0009)*(1+s_1215/kmp_s_1215r_0009))-1)/intracellular
kmp_s_0514r_0534=0.549; kms_s_0386r_0534=0.549; kms_s_1315r_0534=12.8511; Vmax_r_0534=0.0421077; kmp_s_0763_br_0534=0.549; Keq_r_0534=0.0141635; kmp_s_0083r_0534=0.549Reaction: s_0386 + s_1315 => s_0083 + s_0514 + s_0763_b; s_0083, s_0386, s_0514, s_0763_b, s_1315, Rate Law: intracellular*Vmax_r_0534*(1/kms_s_0386r_0534)^1*(1/kms_s_1315r_0534)^1*(s_0386^1*s_1315^1-s_0083^1*s_0514^1*s_0763_b^2/Keq_r_0534)/(((1+s_0386/kms_s_0386r_0534)*(1+s_1315/kms_s_1315r_0534)+(1+s_0083/kmp_s_0083r_0534)*(1+s_0514/kmp_s_0514r_0534)*(1+s_0763_b/kmp_s_0763_br_0534))-1)/intracellular
Keq_r_0165=0.805968; kmp_s_0434r_0165=1.25956; kmp_s_0755r_0165=0.549; Vmax_r_0165=4.0656; kms_s_0706r_0165=0.549; kms_s_0400r_0165=1.71907Reaction: s_0400 + s_0706 => s_0434 + s_0755; s_0400, s_0434, s_0706, s_0755, Rate Law: intracellular*Vmax_r_0165*(1/kms_s_0400r_0165)^1*(1/kms_s_0706r_0165)^1*(s_0400^1*s_0706^1-s_0434^1*s_0755^1/Keq_r_0165)/(((1+s_0400/kms_s_0400r_0165)*(1+s_0706/kms_s_0706r_0165)+(1+s_0434/kmp_s_0434r_0165)*(1+s_0755/kmp_s_0755r_0165))-1)/intracellular
kms_s_1160r_0265=0.549; kms_s_1096r_0265=0.549; kmp_s_1434_br_0265=0.549; kms_s_0763_br_0265=0.549; kms_s_0302r_0265=0.549; Vmax_r_0265=0.0951282; kmp_s_1091r_0265=0.549; kmp_s_1455r_0265=0.549; Keq_r_0265=2.00364Reaction: s_0302 + s_0763_b + s_1096 + s_1160 => s_1091 + s_1434_b + s_1455; s_0302, s_0763_b, s_1091, s_1096, s_1160, s_1434_b, s_1455, Rate Law: intracellular*Vmax_r_0265*(1/kms_s_0302r_0265)^1*(1/kms_s_0763_br_0265)^1*(1/kms_s_1096r_0265)^1*(1/kms_s_1160r_0265)^1*(s_0302^1*s_0763_b^1*s_1096^1*s_1160^1-s_1091^1*s_1434_b^1*s_1455^1/Keq_r_0265)/(((1+s_0302/kms_s_0302r_0265)*(1+s_0763_b/kms_s_0763_br_0265)*(1+s_1096/kms_s_1096r_0265)*(1+s_1160/kms_s_1160r_0265)+(1+s_1091/kmp_s_1091r_0265)*(1+s_1434_b/kmp_s_1434_br_0265)*(1+s_1455/kmp_s_1455r_0265))-1)/intracellular
kms_s_0446r_0123=1.09208; kmp_s_0400r_0123=1.71907; kmp_s_0763_br_0123=0.549; kmp_s_1207r_0123=0.549; Keq_r_0123=0.950614; Vmax_r_0123=0.105501; kmp_s_1005r_0123=0.549; kms_s_0458r_0123=0.549; kms_s_0380r_0123=0.549Reaction: s_0380 + s_0446 + s_0458 => s_0400 + s_0763_b + s_1005 + s_1207; s_0380, s_0400, s_0446, s_0458, s_0763_b, s_1005, s_1207, Rate Law: intracellular*Vmax_r_0123*(1/kms_s_0380r_0123)^1*(1/kms_s_0446r_0123)^1*(1/kms_s_0458r_0123)^1*(s_0380^1*s_0446^1*s_0458^1-s_0400^1*s_0763_b^1*s_1005^1*s_1207^1/Keq_r_0123)/(((1+s_0380/kms_s_0380r_0123)*(1+s_0446/kms_s_0446r_0123)*(1+s_0458/kms_s_0458r_0123)+(1+s_0400/kmp_s_0400r_0123)*(1+s_0763_b/kmp_s_0763_br_0123)*(1+s_1005/kmp_s_1005r_0123)*(1+s_1207/kmp_s_1207r_0123))-1)/intracellular
kms_s_1091r_0719=0.549; kmp_s_1096r_0719=0.549; kms_s_0046r_0719=0.549; kmp_s_0247r_0719=0.549; kmp_s_0763_br_0719=0.549; Keq_r_0719=0.6039; Vmax_r_0719=3.30329Reaction: s_0046 + s_1091 => s_0247 + s_0763_b + s_1096; s_0046, s_0247, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0719*(1/kms_s_0046r_0719)^1*(1/kms_s_1091r_0719)^1*(s_0046^1*s_1091^1-s_0247^1*s_0763_b^1*s_1096^1/Keq_r_0719)/(((1+s_0046/kms_s_0046r_0719)*(1+s_1091/kms_s_1091r_0719)+(1+s_0247/kmp_s_0247r_0719)*(1+s_0763_b/kmp_s_0763_br_0719)*(1+s_1096/kmp_s_1096r_0719))-1)/intracellular
Keq_r_0567=1.73154; Vmax_r_0567=0.008393; kmp_s_0400r_0567=1.71907; kmp_s_0706r_0567=0.549; kms_s_0752r_0567=0.549; kms_s_0446r_0567=1.09208Reaction: s_0446 + s_0752 => s_0400 + s_0706; s_0400, s_0446, s_0706, s_0752, Rate Law: intracellular*Vmax_r_0567*(1/kms_s_0446r_0567)^1*(1/kms_s_0752r_0567)^1*(s_0446^1*s_0752^1-s_0400^1*s_0706^1/Keq_r_0567)/(((1+s_0446/kms_s_0446r_0567)*(1+s_0752/kms_s_0752r_0567)+(1+s_0400/kmp_s_0400r_0567)*(1+s_0706/kmp_s_0706r_0567))-1)/intracellular
Vmax_r_0937=62.2377; Keq_r_0937=8.61335; kms_s_0446r_0937=1.09208; kmp_s_0763_br_0937=0.549; kmp_s_1207r_0937=0.549; kms_s_1277r_0937=0.0605905; kmp_s_0400r_0937=1.71907; kms_s_0458r_0937=0.549; kmp_s_1156r_0937=0.549Reaction: s_0446 + s_0458 + s_1277 => s_0400 + s_0763_b + s_1156 + s_1207; s_0400, s_0446, s_0458, s_0763_b, s_1156, s_1207, s_1277, Rate Law: intracellular*Vmax_r_0937*(1/kms_s_0446r_0937)^1*(1/kms_s_0458r_0937)^1*(1/kms_s_1277r_0937)^1*(s_0446^1*s_0458^1*s_1277^1-s_0400^1*s_0763_b^1*s_1156^1*s_1207^1/Keq_r_0937)/(((1+s_0446/kms_s_0446r_0937)*(1+s_0458/kms_s_0458r_0937)*(1+s_1277/kms_s_1277r_0937)+(1+s_0400/kmp_s_0400r_0937)*(1+s_0763_b/kmp_s_0763_br_0937)*(1+s_1156/kmp_s_1156r_0937)*(1+s_1207/kmp_s_1207r_0937))-1)/intracellular
kms_s_0763_br_1007=0.549; kmp_s_0304r_1007=0.549; kmp_s_1207r_1007=0.549; kms_s_1347r_1007=0.549; Vmax_r_1007=0.624362; kms_s_0400r_1007=1.71907; Keq_r_1007=0.639881Reaction: s_0400 + s_0763_b + s_1347 => s_0304 + s_1207; s_0304, s_0400, s_0763_b, s_1207, s_1347, Rate Law: intracellular*Vmax_r_1007*(1/kms_s_0400r_1007)^1*(1/kms_s_0763_br_1007)^1*(1/kms_s_1347r_1007)^1*(s_0400^1*s_0763_b^1*s_1347^1-s_0304^1*s_1207^1/Keq_r_1007)/(((1+s_0400/kms_s_0400r_1007)*(1+s_0763_b/kms_s_0763_br_1007)*(1+s_1347/kms_s_1347r_1007)+(1+s_0304/kmp_s_0304r_1007)*(1+s_1207/kmp_s_1207r_1007))-1)/intracellular
kms_s_0215r_0263=0.549; kms_s_0763_br_0263=0.549; kms_s_1096r_0263=0.549; Vmax_r_0263=0.0454962; kmp_s_0302r_0263=0.549; Keq_r_0263=2.00364; kmp_s_1091r_0263=0.549Reaction: s_0215 + s_0763_b + s_1096 => s_0302 + s_1091; s_0215, s_0302, s_0763_b, s_1091, s_1096, Rate Law: intracellular*Vmax_r_0263*(1/kms_s_0215r_0263)^1*(1/kms_s_0763_br_0263)^1*(1/kms_s_1096r_0263)^1*(s_0215^1*s_0763_b^1*s_1096^1-s_0302^1*s_1091^1/Keq_r_0263)/(((1+s_0215/kms_s_0215r_0263)*(1+s_0763_b/kms_s_0763_br_0263)*(1+s_1096/kms_s_1096r_0263)+(1+s_0302/kmp_s_0302r_0263)*(1+s_1091/kmp_s_1091r_0263))-1)/intracellular
kmp_s_0434r_0551=1.25956; kmp_s_0899r_0551=0.549; kmp_s_0752r_0551=0.549; kmp_s_0605r_0551=0.549; kms_s_0907r_0551=0.549; kms_s_1434_br_0551=0.549; Keq_r_0551=0.382386; kms_s_0446r_0551=1.09208; kms_s_0306r_0551=0.549; kmp_s_0763_br_0551=0.549; Vmax_r_0551=1.57168Reaction: s_0306 + s_0446 + s_0907 + s_1434_b => s_0434 + s_0605 + s_0752 + s_0763_b + s_0899; s_0306, s_0434, s_0446, s_0605, s_0752, s_0763_b, s_0899, s_0907, s_1434_b, Rate Law: intracellular*Vmax_r_0551*(1/kms_s_0306r_0551)^1*(1/kms_s_0446r_0551)^1*(1/kms_s_0907r_0551)^1*(1/kms_s_1434_br_0551)^1*(s_0306^1*s_0446^1*s_0907^1*s_1434_b^1-s_0434^1*s_0605^1*s_0752^1*s_0763_b^2*s_0899^1/Keq_r_0551)/(((1+s_0306/kms_s_0306r_0551)*(1+s_0446/kms_s_0446r_0551)*(1+s_0907/kms_s_0907r_0551)*(1+s_1434_b/kms_s_1434_br_0551)+(1+s_0434/kmp_s_0434r_0551)*(1+s_0605/kmp_s_0605r_0551)*(1+s_0752/kmp_s_0752r_0551)*(1+s_0763_b/kmp_s_0763_br_0551)*(1+s_0899/kmp_s_0899r_0551))-1)/intracellular
kmp_s_0867r_0650=0.549; kms_s_0763_br_0650=0.549; kmp_s_0434r_0650=1.25956; Vmax_r_0650=4.53532; kmp_s_0605r_0650=0.549; kms_s_1087r_0650=0.0867353; kmp_s_1082r_0650=1.50326; kms_s_0446r_0650=1.09208; Keq_r_0650=21.9885; kms_s_0861r_0650=0.549Reaction: s_0446 + s_0763_b + s_0861 + s_1087 => s_0434 + s_0605 + s_0867 + s_1082; s_0434, s_0446, s_0605, s_0763_b, s_0861, s_0867, s_1082, s_1087, Rate Law: intracellular*Vmax_r_0650*(1/kms_s_0446r_0650)^1*(1/kms_s_0763_br_0650)^1*(1/kms_s_0861r_0650)^1*(1/kms_s_1087r_0650)^1*(s_0446^1*s_0763_b^1*s_0861^1*s_1087^1-s_0434^1*s_0605^1*s_0867^1*s_1082^1/Keq_r_0650)/(((1+s_0446/kms_s_0446r_0650)*(1+s_0763_b/kms_s_0763_br_0650)*(1+s_0861/kms_s_0861r_0650)*(1+s_1087/kms_s_1087r_0650)+(1+s_0434/kmp_s_0434r_0650)*(1+s_0605/kmp_s_0605r_0650)*(1+s_0867/kmp_s_0867r_0650)*(1+s_1082/kmp_s_1082r_0650))-1)/intracellular
Vmax_r_1066=0.025718; kmp_s_0446r_1066=1.09208; Keq_r_1066=0.698801; kms_s_0622r_1066=0.549; kms_s_0400r_1066=1.71907; kmp_s_0624r_1066=0.549Reaction: s_0400 + s_0622 => s_0446 + s_0624; s_0400, s_0446, s_0622, s_0624, Rate Law: intracellular*Vmax_r_1066*(1/kms_s_0400r_1066)^1*(1/kms_s_0622r_1066)^1*(s_0400^1*s_0622^1-s_0446^1*s_0624^1/Keq_r_1066)/(((1+s_0400/kms_s_0400r_1066)*(1+s_0622/kms_s_0622r_1066)+(1+s_0446/kmp_s_0446r_1066)*(1+s_0624/kmp_s_0624r_1066))-1)/intracellular
kms_s_0763_br_0765=0.549; Vmax_r_0765=1.0241; Keq_r_0765=2.00364; kms_s_0180r_0765=0.549; kmp_s_0181r_0765=0.549; kmp_s_0470r_0765=1.0Reaction: s_0180 + s_0763_b => s_0181 + s_0470; s_0180, s_0181, s_0470, s_0763_b, Rate Law: intracellular*Vmax_r_0765*(1/kms_s_0180r_0765)^1*(1/kms_s_0763_br_0765)^1*(s_0180^1*s_0763_b^1-s_0181^1*s_0470^1/Keq_r_0765)/(((1+s_0180/kms_s_0180r_0765)*(1+s_0763_b/kms_s_0763_br_0765)+(1+s_0181/kmp_s_0181r_0765)*(1+s_0470/kmp_s_0470r_0765))-1)/intracellular
Vmax_r_0419=0.00599719; kms_s_0968r_0419=0.549; kms_s_1096r_0419=0.549; kmp_s_1028r_0419=0.549; kmp_s_1091r_0419=0.549; kmp_s_0514r_0419=0.549; Keq_r_0419=3.64962; kmp_s_0470r_0419=1.0; kms_s_1005r_0419=0.549; kmp_s_1434_br_0419=0.549; kms_s_0763_br_0419=0.549Reaction: s_0763_b + s_0968 + s_1005 + s_1096 => s_0470 + s_0514 + s_1028 + s_1091 + s_1434_b; s_0470, s_0514, s_0763_b, s_0968, s_1005, s_1028, s_1091, s_1096, s_1434_b, Rate Law: intracellular*Vmax_r_0419*(1/kms_s_0763_br_0419)^3*(1/kms_s_0968r_0419)^1*(1/kms_s_1005r_0419)^1*(1/kms_s_1096r_0419)^2*(s_0763_b^3*s_0968^1*s_1005^1*s_1096^2-s_0470^1*s_0514^1*s_1028^1*s_1091^2*s_1434_b^1/Keq_r_0419)/(((1+s_0763_b/kms_s_0763_br_0419)*(1+s_0968/kms_s_0968r_0419)*(1+s_1005/kms_s_1005r_0419)*(1+s_1096/kms_s_1096r_0419)+(1+s_0470/kmp_s_0470r_0419)*(1+s_0514/kmp_s_0514r_0419)*(1+s_1028/kmp_s_1028r_0419)*(1+s_1091/kmp_s_1091r_0419)*(1+s_1434_b/kmp_s_1434_br_0419))-1)/intracellular
kms_s_0431_br_1157=38.0; Keq_r_1157=1.0; Vmax_r_1157=0.964941; kmp_s_0430r_1157=0.549Reaction: s_0431_b => s_0430; s_0430, s_0431_b, Rate Law: Vmax_r_1157*(1/kms_s_0431_br_1157)^1*(s_0431_b^1-s_0430^1/Keq_r_1157)/((1+s_0431_b/kms_s_0431_br_1157+1+s_0430/kmp_s_0430r_1157)-1)
kms_s_0532r_0605=0.549; kmp_s_1434_br_0605=0.549; Vmax_r_0605=0.229349; kmp_s_0212r_0605=0.549; Keq_r_0605=0.6039Reaction: s_0532 => s_0212 + s_1434_b; s_0212, s_0532, s_1434_b, Rate Law: intracellular*Vmax_r_0605*(1/kms_s_0532r_0605)^1*(s_0532^1-s_0212^1*s_1434_b^1/Keq_r_0605)/((1+s_0532/kms_s_0532r_0605+(1+s_0212/kmp_s_0212r_0605)*(1+s_1434_b/kmp_s_1434_br_0605))-1)/intracellular
kmp_s_1082r_0058=1.50326; Vmax_r_0058=3.30332; kms_s_0763_br_0058=0.549; Keq_r_0058=34.7263; kms_s_0257r_0058=0.549; kmp_s_0052r_0058=0.549; kms_s_1087r_0058=0.0867353Reaction: s_0257 + s_0763_b + s_1087 => s_0052 + s_1082; s_0052, s_0257, s_0763_b, s_1082, s_1087, Rate Law: intracellular*Vmax_r_0058*(1/kms_s_0257r_0058)^1*(1/kms_s_0763_br_0058)^1*(1/kms_s_1087r_0058)^1*(s_0257^1*s_0763_b^1*s_1087^1-s_0052^1*s_1082^1/Keq_r_0058)/(((1+s_0257/kms_s_0257r_0058)*(1+s_0763_b/kms_s_0763_br_0058)*(1+s_1087/kms_s_1087r_0058)+(1+s_0052/kmp_s_0052r_0058)*(1+s_1082/kmp_s_1082r_0058))-1)/intracellular
kmp_s_0766_br_1503=0.1; kmp_s_1339_br_1503=1.0; Keq_r_1503=1.0; kms_s_0763_br_1503=0.549; Vmax_r_1503=0.840147; kms_s_1338r_1503=0.549Reaction: s_0763_b + s_1338 => s_0766_b + s_1339_b; s_0763_b, s_0766_b, s_1338, s_1339_b, Rate Law: Vmax_r_1503*(1/kms_s_0763_br_1503)^1*(1/kms_s_1338r_1503)^1*(s_0763_b^1*s_1338^1-s_0766_b^1*s_1339_b^1/Keq_r_1503)/(((1+s_0763_b/kms_s_0763_br_1503)*(1+s_1338/kms_s_1338r_1503)+(1+s_0766_b/kmp_s_0766_br_1503)*(1+s_1339_b/kmp_s_1339_br_1503))-1)
kms_s_0446r_0249=1.09208; kmp_s_0766_br_0249=0.1; kmp_s_1207r_0249=0.549; kmp_s_0400r_0249=1.71907; Vmax_r_0249=50.4568; Keq_r_0249=0.173154; kms_s_1434_br_0249=0.549Reaction: s_0446 + s_1434_b => s_0400 + s_0766_b + s_1207; s_0400, s_0446, s_0766_b, s_1207, s_1434_b, Rate Law: Vmax_r_0249*(1/kms_s_0446r_0249)^1*(1/kms_s_1434_br_0249)^1*(s_0446^1*s_1434_b^1-s_0400^1*s_0766_b^1*s_1207^1/Keq_r_0249)/(((1+s_0446/kms_s_0446r_0249)*(1+s_1434_b/kms_s_1434_br_0249)+(1+s_0400/kmp_s_0400r_0249)*(1+s_0766_b/kmp_s_0766_br_0249)*(1+s_1207/kmp_s_1207r_0249))-1)
Vmax_r_1247=4.81765; Keq_r_1247=1.0; kms_s_0650r_1247=50.0; kmp_s_0651_br_1247=24.5Reaction: s_0650 => s_0651_b; s_0650, s_0651_b, Rate Law: Vmax_r_1247*(1/kms_s_0650r_1247)^1*(s_0650^1-s_0651_b^1/Keq_r_1247)/((1+s_0650/kms_s_0650r_1247+1+s_0651_b/kmp_s_0651_br_1247)-1)
kmp_s_0740r_0174=0.549; Vmax_r_0174=1.7171; kms_s_0863r_0174=0.549; kms_s_0749r_0174=0.549; Keq_r_0174=0.121402; kmp_s_1277r_0174=0.0605905Reaction: s_0749 + s_0863 => s_0740 + s_1277; s_0740, s_0749, s_0863, s_1277, Rate Law: intracellular*Vmax_r_0174*(1/kms_s_0749r_0174)^1*(1/kms_s_0863r_0174)^1*(s_0749^1*s_0863^1-s_0740^1*s_1277^1/Keq_r_0174)/(((1+s_0749/kms_s_0749r_0174)*(1+s_0863/kms_s_0863r_0174)+(1+s_0740/kmp_s_0740r_0174)*(1+s_1277/kmp_s_1277r_0174))-1)/intracellular
Keq_r_0948=0.331541; kmp_s_1207r_0948=0.549; kms_s_0163r_0948=0.549; Vmax_r_0948=0.0120878; kms_s_0320r_0948=0.549; kmp_s_0335r_0948=0.549; kmp_s_1434_br_0948=0.549Reaction: s_0163 + s_0320 => s_0335 + s_1207 + s_1434_b; s_0163, s_0320, s_0335, s_1207, s_1434_b, Rate Law: intracellular*Vmax_r_0948*(1/kms_s_0163r_0948)^1*(1/kms_s_0320r_0948)^1*(s_0163^1*s_0320^1-s_0335^1*s_1207^1*s_1434_b^2/Keq_r_0948)/(((1+s_0163/kms_s_0163r_0948)*(1+s_0320/kms_s_0320r_0948)+(1+s_0335/kmp_s_0335r_0948)*(1+s_1207/kmp_s_1207r_0948)*(1+s_1434_b/kmp_s_1434_br_0948))-1)/intracellular
kmp_s_0470r_0608=1.0; kms_s_0763_br_0608=0.549; kmp_s_0088r_0608=0.549; Keq_r_0608=1.1; Vmax_r_0608=0.187549; kms_s_0078r_0608=0.549; kmp_s_1434_br_0608=0.549Reaction: s_0078 + s_0763_b => s_0088 + s_0470 + s_1434_b; s_0078, s_0088, s_0470, s_0763_b, s_1434_b, Rate Law: intracellular*Vmax_r_0608*(1/kms_s_0078r_0608)^1*(1/kms_s_0763_br_0608)^1*(s_0078^1*s_0763_b^1-s_0088^1*s_0470^1*s_1434_b^1/Keq_r_0608)/(((1+s_0078/kms_s_0078r_0608)*(1+s_0763_b/kms_s_0763_br_0608)+(1+s_0088/kmp_s_0088r_0608)*(1+s_0470/kmp_s_0470r_0608)*(1+s_1434_b/kmp_s_1434_br_0608))-1)/intracellular
kmp_s_1207r_0789=0.549; kms_s_1151r_0789=0.549; kmp_s_0763_br_0789=0.549; Vmax_r_0789=0.912336; kmp_s_0887r_0789=0.549; Keq_r_0789=0.6039; kms_s_0469r_0789=0.549Reaction: s_0469 + s_1151 => s_0763_b + s_0887 + s_1207; s_0469, s_0763_b, s_0887, s_1151, s_1207, Rate Law: intracellular*Vmax_r_0789*(1/kms_s_0469r_0789)^1*(1/kms_s_1151r_0789)^1*(s_0469^1*s_1151^1-s_0763_b^1*s_0887^1*s_1207^1/Keq_r_0789)/(((1+s_0469/kms_s_0469r_0789)*(1+s_1151/kms_s_1151r_0789)+(1+s_0763_b/kmp_s_0763_br_0789)*(1+s_0887/kmp_s_0887r_0789)*(1+s_1207/kmp_s_1207r_0789))-1)/intracellular
s_0463_or_1814=0.549; zero_flux=0.0; V_o=0.0555; a_s_0463r_1814=1.0Reaction: s_0463 => s_0464_b; s_0547_b, s_0463, Rate Law: piecewise(V_o*(1+a_s_0463r_1814*ln(s_0463/s_0463_or_1814)), (V_o*(1+a_s_0463r_1814*ln(s_0463/s_0463_or_1814))) >= zero_flux, zero_flux)
kms_s_1122r_1027=0.549; Vmax_r_1027=5.5748; kmp_s_0949r_1027=1.0; kmp_s_1207r_1027=0.549; Keq_r_1027=2.00364; kms_s_1434_br_1027=0.549Reaction: s_1122 + s_1434_b => s_0949 + s_1207; s_0949, s_1122, s_1207, s_1434_b, Rate Law: intracellular*Vmax_r_1027*(1/kms_s_1122r_1027)^1*(1/kms_s_1434_br_1027)^1*(s_1122^1*s_1434_b^1-s_0949^1*s_1207^1/Keq_r_1027)/(((1+s_1122/kms_s_1122r_1027)*(1+s_1434_b/kms_s_1434_br_1027)+(1+s_0949/kmp_s_0949r_1027)*(1+s_1207/kmp_s_1207r_1027))-1)/intracellular
Vmax_r_0568=0.0076692; kmp_s_0706r_0568=0.549; Keq_r_0568=1.1; kms_s_0566r_0568=0.549; kms_s_0752r_0568=0.549; kmp_s_0562r_0568=0.549Reaction: s_0566 + s_0752 => s_0562 + s_0706; s_0562, s_0566, s_0706, s_0752, Rate Law: intracellular*Vmax_r_0568*(1/kms_s_0566r_0568)^1*(1/kms_s_0752r_0568)^1*(s_0566^1*s_0752^1-s_0562^1*s_0706^1/Keq_r_0568)/(((1+s_0566/kms_s_0566r_0568)*(1+s_0752/kms_s_0752r_0568)+(1+s_0562/kmp_s_0562r_0568)*(1+s_0706/kmp_s_0706r_0568))-1)/intracellular

States:

NameDescription
s 0455[beta-D-glucose 6-phosphate]
s 0446[ATP(4-)]
s 0752[GMP]
s 1162 b[dioxygen]
s 0380[acetyl-CoA]
s 0146[CHEBI_52957]
s 0532[D-erythro-1-(imidazol-4-yl)glycerol 3-phosphate]
s 1154[orotate]
s 0472 b[carbon dioxide]
s 0514[coenzyme A]
s 0763 b[proton]
s 0755[GTP(4-)]
s 1207[hydrogenphosphate]
s 0547 b[D-glucose]
s 1096[NADPH]
s 0766 b[proton]
s 0386[acyl-CoA]
s 0078[1-(2-carboxyphenylamino)-1-deoxy-D-ribulose 5-phosphate]
s 0987[lignocerate]
s 0749[glyoxylate]
s 0651 b[ethanol]
s 0419[alpha,alpha-trehalose 6-phosphate]
s 0458[hydrogencarbonate]
s 0530[D-arabinose]
s 0464 b[no biological data found]
s 0431 b[ammonium]
s 1339 b[succinate(2-)]
s 1434 b[water]
s 0079[1-(5-phospho-beta-D-ribosyl)-5-[(5-phospho-beta-D-ribosylamino)methylideneamino]imidazole-4-carboxamide]
s 0400[ADP]
s 0434[AMP]
s 0463[no biological data found]
s 0740[glycine]
s 0743[glycogen]
s 0468[but-1-ene-1,2,4-tricarboxylic acid]

Starbuck1990 - EGF binding and trafficking dynamics in Fibroblast: MODEL2003190005v0.0.1

mathematical model, with accompanying quantitative experimental data, for binding and trafficking properties of the epid…

Details

We provide a mathematical model, with accompanying quantitative experimental data, for binding and trafficking properties of the epidermal growth factor (EGF) receptor on B82 fibroblasts, and propose a theoretical dependence of cell proliferation rate on these properties. The signal for cell proliferation is generated by intrinsic EGF-receptor (EGFR) tyrosine kinase activation via EGF binding at the cell surface, and terminated by receptor/growth factor complex internalization and degradation. Our model consists of kinetic equations which describe the binding, internalization, and recycling of EGF and EGFR, along with a simple expression relation the dependence of cell cycle progression on EGFR dynamics. We show that, with key model parameters determined independently from EGF/fibroblast binding and internalization experiments, our model successfully predicts, as a first step, kinetic data for EGF binding to and internalization by B82 cells at 37°C link: http://identifiers.org/doi/10.1016/0009-2509(90)80117-W