SBMLBioModels: C - D

C


Castillo2016 - Whole-genome metabolic model of T.asperellum using CoReCo: MODEL1604280022v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Castillo2016 - Whole-genome metabolic model of T.atroviride using CoReCo: MODEL1604280054v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Castillo2016 - Whole-genome metabolic model of T.citrinoviride using CoReCo: MODEL1604280004v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Castillo2016 - Whole-genome metabolic model of T.harzianum using CoReCo: MODEL1604280000v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Castillo2016 - Whole-genome metabolic model of T.longibrachiatum using CoReCo: MODEL1604280010v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Castillo2016 - Whole-genome metabolic model of T.reesei using CoReCo: MODEL1604280024v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Castillo2016 - Whole-genome metabolic model of T.virens using CoReCo: MODEL1604280038v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Castillo2016 - Whole-genome metabolic model of U.maydis using CoReCo: MODEL1604280020v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Castillo2016 - Whole-genome metabolic model of U.reesii using CoReCo: MODEL1604280050v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Castillo2016 - Whole-genome metabolic model of Y.lipolytica using CoReCo: MODEL1604280017v0.0.1

This model was reconstructed with CoReCo method from protein sequence and phylogeny data. CoReCo is described in Pitkane…

Details

Trichoderma reesei is one of the main sources of biomass-hydrolyzing enzymes for the biotechnology industry. There is a need for improving its enzyme production efficiency. The use of metabolic modeling for the simulation and prediction of this organism's metabolism is potentially a valuable tool for improving its capabilities. An accurate metabolic model is needed to perform metabolic modeling analysis.A whole-genome metabolic model of T. reesei has been reconstructed together with metabolic models of 55 related species using the metabolic model reconstruction algorithm CoReCo. The previously published CoReCo method has been improved to obtain better quality models. The main improvements are the creation of a unified database of reactions and compounds and the use of reaction directions as constraints in the gap-filling step of the algorithm. In addition, the biomass composition of T. reesei has been measured experimentally to build and include a specific biomass equation in the model.The improvements presented in this work on the CoReCo pipeline for metabolic model reconstruction resulted in higher-quality metabolic models compared with previous versions. A metabolic model of T. reesei has been created and is publicly available in the BIOMODELS database. The model contains a biomass equation, reaction boundaries and uptake/export reactions which make it ready for simulation. To validate the model, we dem1onstrate that the model is able to predict biomass production accurately and no stoichiometrically infeasible yields are detected. The new T. reesei model is ready to be used for simulations of protein production processes. link: http://identifiers.org/pubmed/27895706

Caydasi2012 - Inhibition of Tem1 by the GAP complex in Spindle Position Checkpoint: BIOMD0000000701v0.0.1

This model is from the article: A dynamical model of the spindle position checkpoint Ayse Koca Caydasi, Maiko Lohel,…

Details

The orientation of the mitotic spindle with respect to the polarity axis is crucial for the accuracy of asymmetric cell division. In budding yeast, a surveillance mechanism called the spindle position checkpoint (SPOC) prevents exit from mitosis when the mitotic spindle fails to align along the mother-to-daughter polarity axis. SPOC arrest relies upon inhibition of the GTPase Tem1 by the GTPase-activating protein (GAP) complex Bfa1-Bub2. Importantly, reactions signaling mitotic exit take place at yeast centrosomes (named spindle pole bodies, SPBs) and the GAP complex also promotes SPB localization of Tem1. Yet, whether the regulation of Tem1 by Bfa1-Bub2 takes place only at the SPBs remains elusive. Here, we present a quantitative analysis of Bfa1-Bub2 and Tem1 localization at the SPBs. Based on the measured SPB-bound protein levels, we introduce a dynamical model of the SPOC that describes the regulation of Bfa1 and Tem1. Our model suggests that Bfa1 interacts with Tem1 in the cytoplasm as well as at the SPBs to provide efficient Tem1 inhibition. link: http://identifiers.org/pubmed/22580890

Caydasi2012 - Regulation of Tem1 by the GAP complex in spindle position cell cycle checkpoint - Ubiquitous association model: BIOMD0000000699v0.0.1

Caydasi2012 - Regulation of Tem1 by the GAP complex in spindle position cell cycle checkpoint - Ubiquitous association m…

Details

The orientation of the mitotic spindle with respect to the polarity axis is crucial for the accuracy of asymmetric cell division. In budding yeast, a surveillance mechanism called the spindle position checkpoint (SPOC) prevents exit from mitosis when the mitotic spindle fails to align along the mother-to-daughter polarity axis. SPOC arrest relies upon inhibition of the GTPase Tem1 by the GTPase-activating protein (GAP) complex Bfa1-Bub2. Importantly, reactions signaling mitotic exit take place at yeast centrosomes (named spindle pole bodies, SPBs) and the GAP complex also promotes SPB localization of Tem1. Yet, whether the regulation of Tem1 by Bfa1-Bub2 takes place only at the SPBs remains elusive. Here, we present a quantitative analysis of Bfa1-Bub2 and Tem1 localization at the SPBs. Based on the measured SPB-bound protein levels, we introduce a dynamical model of the SPOC that describes the regulation of Bfa1 and Tem1. Our model suggests that Bfa1 interacts with Tem1 in the cytoplasm as well as at the SPBs to provide efficient Tem1 inhibition. link: http://identifiers.org/pubmed/22580890

Parameters:

NameDescription
koffBT = 0.183 1/s; konB5T = 7000000.0 l/(mol*s)Reaction: Tem1GTP + B_Bfa1P5 => B_Bfa1P5_Tem1GTP, Rate Law: c3*(konB5T*B_Bfa1P5*Tem1GTP-koffBT*B_Bfa1P5_Tem1GTP)
konB4T = 3.65E7 l/(mol*s); koffBT = 0.183 1/sReaction: Tem1GDP + B_Bfa1P4 => B_Bfa1P4_Tem1GDP, Rate Law: c3*(konB4T*B_Bfa1P4*Tem1GDP-koffBT*B_Bfa1P4_Tem1GDP)
kfKin4Cyto = 0.09 1/s; u = 1.0 1Reaction: Bfa1_Tem1GDP => Bfa1P4_Tem1GDP, Rate Law: c2*u*kfKin4Cyto*Bfa1_Tem1GDP
krKin4 = 0.0251 1/sReaction: Bfa1P4_Tem1GTP => Bfa1_Tem1GTP, Rate Law: c2*krKin4*Bfa1P4_Tem1GTP
koffB4 = 0.0365 1/s; konB4 = 20000.0 l/(mol*s)Reaction: Bfa1P4_Tem1GDP + SPB_B => B_Bfa1P4_Tem1GDP, Rate Law: c3*(konB4*SPB_B*Bfa1P4_Tem1GDP-koffB4*B_Bfa1P4_Tem1GDP)
khydBT = 2.0 1/sReaction: Bfa1_Tem1GTP => Bfa1_Tem1GDP, Rate Law: c2*khydBT*Bfa1_Tem1GTP
alpha = 1.0 1; koffBT = 0.183 1/s; konBT = 3.65E7 l/(mol*s)Reaction: Bfa1 + Tem1GDP => Bfa1_Tem1GDP, Rate Law: c2*(alpha*konBT*Bfa1*Tem1GDP-koffBT*Bfa1_Tem1GDP)
kfKin4 = 1000.0 1/s; u = 1.0 1Reaction: B_Bfa1 => B_Bfa1P4, Rate Law: c3*u*kfKin4*B_Bfa1
konB4T = 3.65E7 l/(mol*s); alpha = 1.0 1; koffBT = 0.183 1/sReaction: Bfa1P4 + Tem1GTP => Bfa1P4_Tem1GTP, Rate Law: c2*(alpha*konB4T*Bfa1P4*Tem1GTP-koffBT*Bfa1P4_Tem1GTP)
krCdc5 = 0.01 1/s; u = 1.0 1Reaction: Bfa1P5_Tem1GDP => Bfa1_Tem1GDP, Rate Law: c2*u*krCdc5*Bfa1P5_Tem1GDP
avogadro = 6.0221415E23Reaction: Active_Tem1_at_the_SPB = (T_Tem1GTP+B_Bfa1_Tem1GTP+B_Bfa1P4_Tem1GTP+B_Bfa1P5_Tem1GTP)*c3*avogadro, Rate Law: missing
khydB4T = 2.0 1/sReaction: Bfa1P4_Tem1GTP => Bfa1P4_Tem1GDP, Rate Law: c2*khydB4T*Bfa1P4_Tem1GTP
alpha = 1.0 1; koffBT = 0.183 1/s; konB5T = 7000000.0 l/(mol*s)Reaction: Bfa1P5 + Tem1GTP => Bfa1P5_Tem1GTP, Rate Law: c2*(alpha*konB5T*Bfa1P5*Tem1GTP-koffBT*Bfa1P5_Tem1GTP)
khyd = 0.00224 1/sReaction: Bfa1P5_Tem1GTP => Bfa1P5_Tem1GDP, Rate Law: c2*khyd*Bfa1P5_Tem1GTP
konB = 1250000.0 l/(mol*s); koffB = 0.0012 1/sReaction: Bfa1P5_Tem1GTP + SPB_B => B_Bfa1P5_Tem1GTP, Rate Law: c3*(konB*SPB_B*Bfa1P5_Tem1GTP-koffB*B_Bfa1P5_Tem1GTP)
knex = 0.0136 1/sReaction: Tem1GDP => Tem1GTP, Rate Law: c2*knex*Tem1GDP
kfCdc5 = 1.0 1/sReaction: B_Bfa1 => B_Bfa1P5, Rate Law: c3*kfCdc5*B_Bfa1
koffBT = 0.183 1/s; konBT = 3.65E7 l/(mol*s)Reaction: Tem1GTP + B_Bfa1 => B_Bfa1_Tem1GTP, Rate Law: c3*(konBT*B_Bfa1*Tem1GTP-koffBT*B_Bfa1_Tem1GTP)
konT = 1900000.0 l/(mol*s); koffT = 0.183 1/sReaction: Tem1GTP + SPB_T => T_Tem1GTP, Rate Law: c3*(konT*SPB_T*Tem1GTP-koffT*T_Tem1GTP)
avogadro = 6.0221415E23; q = 1.0 1Reaction: Active_Bfa1_at_the_SPB = (q*(B_Bfa1+B_Bfa1_Tem1GTP+B_Bfa1_Tem1GDP)+B_Bfa1P4+B_Bfa1P4_Tem1GTP+B_Bfa1P4_Tem1GDP)*c3*avogadro, Rate Law: missing

States:

NameDescription
B Bfa1[Mitotic check point protein BFA1; binding site]
Active Tem1 at the SPB[Protein TEM1; active]
Bfa1P5 Tem1GDP[GDP; Mitotic check point protein BFA1; Protein TEM1; increased phosphorylation]
Bfa1P5 Tem1GTP[Mitotic check point protein BFA1; GTP; Protein TEM1; increased phosphorylation]
B Bfa1P4 Tem1GDP[binding site; GDP; Protein TEM1; Mitotic check point protein BFA1; phosphorylated]
Inactive Bfa1 at the SPB[Mitotic check point protein BFA1; inactive]
B Bfa1P4[Mitotic check point protein BFA1; binding site; phosphorylated]
B Bfa1P4 Tem1GTP[Protein TEM1; GTP; binding site; Mitotic check point protein BFA1; phosphorylated]
Bfa1 Tem1GDP[Protein TEM1; GDP; Mitotic check point protein BFA1]
Inactive Bfa1 in the cytosol[Mitotic check point protein BFA1; inactive]
Bfa1P5[Mitotic check point protein BFA1; increased phosphorylation]
SPB T[binding site]
Tem1GTP[Protein TEM1; GTP]
SPB B[binding site]
Bfa1P4[Mitotic check point protein BFA1; phosphorylated]
Bfa1[Mitotic check point protein BFA1]
B Bfa1P5 Tem1GDP[Mitotic check point protein BFA1; binding site; GDP; Protein TEM1; phosphorylated]
B Bfa1P5 Tem1GTP[GTP; Mitotic check point protein BFA1; binding site; Protein TEM1; phosphorylated]
Active Bfa1 at the Cytosol[Mitotic check point protein BFA1; active]
Bfa1P4 Tem1GDP[Protein TEM1; Mitotic check point protein BFA1; GDP; phosphorylated]
B Bfa1P5[Mitotic check point protein BFA1; binding site; phosphorylated]
Bfa1P4 Tem1GTP[Mitotic check point protein BFA1; GTP; Protein TEM1; phosphorylated]
Active Tem1 in the Cytosol[Protein TEM1; active]
B Bfa1 Tem1GTP[GTP; binding site; Protein TEM1; Mitotic check point protein BFA1]
Tem1GDP[GDP; Protein TEM1]
Inactive Tem1 in the cytosol[Protein TEM1; inactive]
Total Bfa1 in the Cytosol[Mitotic check point protein BFA1]
Total Tem1 in the Cytosol[Protein TEM1]
Total Bfa1 at the SPB[Mitotic check point protein BFA1]
Active Bfa1 at the SPB[Mitotic check point protein BFA1; active]
Total Tem1 at the SPB[Protein TEM1]
Bfa1 Tem1GTP[GTP; Mitotic check point protein BFA1; Protein TEM1]
Inactive Tem1 at the SPB[Protein TEM1; inactive]

Caydasi2012 - Regulation of Tem1 by the GAP complex in Spindle Position Checkpoint - Ubiquitous inactive model: BIOMD0000000702v0.0.1

Regulation of Tem1 by the GAP complex in Spindle Position Checkpoint - Ubiquitous inactiveThis model is described in the…

Details

The orientation of the mitotic spindle with respect to the polarity axis is crucial for the accuracy of asymmetric cell division. In budding yeast, a surveillance mechanism called the spindle position checkpoint (SPOC) prevents exit from mitosis when the mitotic spindle fails to align along the mother-to-daughter polarity axis. SPOC arrest relies upon inhibition of the GTPase Tem1 by the GTPase-activating protein (GAP) complex Bfa1-Bub2. Importantly, reactions signaling mitotic exit take place at yeast centrosomes (named spindle pole bodies, SPBs) and the GAP complex also promotes SPB localization of Tem1. Yet, whether the regulation of Tem1 by Bfa1-Bub2 takes place only at the SPBs remains elusive. Here, we present a quantitative analysis of Bfa1-Bub2 and Tem1 localization at the SPBs. Based on the measured SPB-bound protein levels, we introduce a dynamical model of the SPOC that describes the regulation of Bfa1 and Tem1. Our model suggests that Bfa1 interacts with Tem1 in the cytoplasm as well as at the SPBs to provide efficient Tem1 inhibition. link: http://identifiers.org/pubmed/22580890

Parameters:

NameDescription
koffBT = 0.183 1/s; konB5T = 7000000.0 l/(mol*s)Reaction: Tem1GDP + B_Bfa1P5 => B_Bfa1P5_Tem1GDP, Rate Law: c3*(konB5T*B_Bfa1P5*Tem1GDP-koffBT*B_Bfa1P5_Tem1GDP)
konB4T = 3.65E7 l/(mol*s); koffBT = 0.183 1/sReaction: Tem1GTP + B_Bfa1P4 => B_Bfa1P4_Tem1GTP, Rate Law: c3*(konB4T*B_Bfa1P4*Tem1GTP-koffBT*B_Bfa1P4_Tem1GTP)
kfKin4Cyto = 0.09 1/s; u = 1.0 1Reaction: Bfa1_Tem1GTP => Bfa1P4_Tem1GTP, Rate Law: c2*u*kfKin4Cyto*Bfa1_Tem1GTP
krKin4 = 0.0251 1/sReaction: Bfa1P4_Tem1GTP => Bfa1_Tem1GTP, Rate Law: c2*krKin4*Bfa1P4_Tem1GTP
koffB4 = 0.0365 1/s; konB4 = 20000.0 l/(mol*s)Reaction: Bfa1P4 + SPB_B => B_Bfa1P4, Rate Law: c3*(konB4*SPB_B*Bfa1P4-koffB4*B_Bfa1P4)
konB4T = 3.65E7 l/(mol*s); alpha = 1.0 1; koffBT = 0.183 1/sReaction: Bfa1P4 + Tem1GDP => Bfa1P4_Tem1GDP, Rate Law: c2*(alpha*konB4T*Bfa1P4*Tem1GDP-koffBT*Bfa1P4_Tem1GDP)
alpha = 1.0 1; koffBT = 0.183 1/s; konBT = 3.65E7 l/(mol*s)Reaction: Bfa1 + Tem1GDP => Bfa1_Tem1GDP, Rate Law: c2*(alpha*konBT*Bfa1*Tem1GDP-koffBT*Bfa1_Tem1GDP)
kfKin4 = 1000.0 1/s; u = 1.0 1Reaction: B_Bfa1 => B_Bfa1P4, Rate Law: c3*u*kfKin4*B_Bfa1
krCdc5 = 0.01 1/s; u = 1.0 1Reaction: Bfa1P5_Tem1GDP => Bfa1_Tem1GDP, Rate Law: c2*u*krCdc5*Bfa1P5_Tem1GDP
avogadro = 6.0221415E23; q = 0.0 1Reaction: Active_Bfa1_at_the_Cytosol = (q*(Bfa1+Bfa1_Tem1GTP+Bfa1_Tem1GDP)+Bfa1P4+Bfa1P4_Tem1GTP+Bfa1P4_Tem1GDP)*c2*avogadro, Rate Law: missing
avogadro = 6.0221415E23Reaction: Active_Tem1_in_the_Cytosol = (Tem1GTP+Bfa1_Tem1GTP+Bfa1P4_Tem1GTP+Bfa1P5_Tem1GTP)*c2*avogadro, Rate Law: missing
khydB4T = 2.0 1/sReaction: Bfa1P4_Tem1GTP => Bfa1P4_Tem1GDP, Rate Law: c2*khydB4T*Bfa1P4_Tem1GTP
alpha = 1.0 1; koffBT = 0.183 1/s; konB5T = 7000000.0 l/(mol*s)Reaction: Bfa1P5 + Tem1GTP => Bfa1P5_Tem1GTP, Rate Law: c2*(alpha*konB5T*Bfa1P5*Tem1GTP-koffBT*Bfa1P5_Tem1GTP)
khyd = 0.00224 1/sReaction: T_Tem1GTP => T_Tem1GDP, Rate Law: c3*khyd*T_Tem1GTP
konB = 1250000.0 l/(mol*s); koffB = 0.0012 1/sReaction: SPB_B + Bfa1 => B_Bfa1, Rate Law: c3*(konB*SPB_B*Bfa1-koffB*B_Bfa1)
khydBT = 0.00224 1/sReaction: Bfa1_Tem1GTP => Bfa1_Tem1GDP, Rate Law: c2*khydBT*Bfa1_Tem1GTP
koffBT = 0.183 1/s; konBT = 3.65E7 l/(mol*s)Reaction: Tem1GDP + B_Bfa1 => B_Bfa1_Tem1GDP, Rate Law: c3*(konBT*B_Bfa1*Tem1GDP-koffBT*B_Bfa1_Tem1GDP)
kfCdc5 = 1.0 1/sReaction: B_Bfa1 => B_Bfa1P5, Rate Law: c3*kfCdc5*B_Bfa1
knex = 0.0136 1/sReaction: T_Tem1GDP => T_Tem1GTP, Rate Law: c3*knex*T_Tem1GDP
konT = 1900000.0 l/(mol*s); koffT = 0.183 1/sReaction: Tem1GTP + SPB_T => T_Tem1GTP, Rate Law: c3*(konT*SPB_T*Tem1GTP-koffT*T_Tem1GTP)

States:

NameDescription
Bfa1 Tem1GTP[Mitotic check point protein BFA1; GTP; Protein TEM1]
Active Tem1 at the SPB[Protein TEM1; active]
Bfa1P5 Tem1GDP[Mitotic check point protein BFA1; increased phosphorylation; Protein TEM1; GDP]
Bfa1P5 Tem1GTP[Protein TEM1; increased phosphorylation; GTP; Mitotic check point protein BFA1]
B Bfa1P4[Mitotic check point protein BFA1; phosphorylated; urn:miriam:sbo:SBO%3A0000494]
Inactive Bfa1 at the SPB[Mitotic check point protein BFA1; inactive]
B Bfa1P4 Tem1GDP[urn:miriam:sbo:SBO%3A0000494; phosphorylated; GDP; Protein TEM1; Mitotic check point protein BFA1]
B Bfa1P4 Tem1GTP[urn:miriam:sbo:SBO%3A0000494; phosphorylated; Protein TEM1; Mitotic check point protein BFA1; GTP]
Bfa1 Tem1GDP[Protein TEM1; GDP; Mitotic check point protein BFA1]
Inactive Bfa1 in the cytosol[Mitotic check point protein BFA1; inactive]
Bfa1P5[Mitotic check point protein BFA1; increased phosphorylation]
SPB T[urn:miriam:sbo:SBO%3A0000494]
B Bfa1 Tem1GDP[urn:miriam:sbo:SBO%3A0000494; Mitotic check point protein BFA1; GDP; Protein TEM1]
Tem1GTP[GTP; Protein TEM1]
SPB B[urn:miriam:sbo:SBO%3A0000494]
Bfa1P4[Mitotic check point protein BFA1; phosphorylated]
Bfa1[Mitotic check point protein BFA1]
B Bfa1P5 Tem1GTP[urn:miriam:sbo:SBO%3A0000494; phosphorylated; GTP; Protein TEM1; Mitotic check point protein BFA1]
B Bfa1P5 Tem1GDP[GDP; phosphorylated; Mitotic check point protein BFA1; urn:miriam:sbo:SBO%3A0000494; Protein TEM1]
Active Bfa1 at the Cytosol[Mitotic check point protein BFA1; active]
Bfa1P4 Tem1GDP[GDP; phosphorylated; Protein TEM1; Mitotic check point protein BFA1]
B Bfa1P5[Mitotic check point protein BFA1; phosphorylated; urn:miriam:sbo:SBO%3A0000494]
Bfa1P4 Tem1GTP[Protein TEM1; phosphorylated; GTP; Mitotic check point protein BFA1]
B Bfa1 Tem1GTP[Mitotic check point protein BFA1; GTP; Protein TEM1; urn:miriam:sbo:SBO%3A0000494]
Tem1GDP[Protein TEM1; GDP]
Active Tem1 in the Cytosol[Protein TEM1; active]
Inactive Tem1 in the cytosol[Protein TEM1; inactive]
T Tem1GDP[Protein TEM1; urn:miriam:sbo:SBO%3A0000494; GDP]
Total Bfa1 in the Cytosol[Mitotic check point protein BFA1]
Total Tem1 in the Cytosol[Protein TEM1]
Total Bfa1 at the SPB[Mitotic check point protein BFA1]
T Tem1GTP[Protein TEM1; GTP; urn:miriam:sbo:SBO%3A0000494]
Active Bfa1 at the SPB[Mitotic check point protein BFA1; active]
Total Tem1 at the SPB[Protein TEM1]
B Bfa1[Mitotic check point protein BFA1; urn:miriam:sbo:SBO%3A0000494]
Inactive Tem1 at the SPB[Protein TEM1; inactive]

Cazzaniga2014 - Blood Coagulation with Platelet Activation: MODEL1807180003v0.0.1

Mathematical model of blood coagulation with platelet activation. Model includes factor XII, factor VIIIa fragments, mei…

Details

The introduction of general-purpose Graphics Processing Units (GPUs) is boosting scientific applications in Bioinformatics, Systems Biology, and Computational Biology. In these fields, the use of high-performance computing solutions is motivated by the need of performing large numbers of in silico analysis to study the behavior of biological systems in different conditions, which necessitate a computing power that usually overtakes the capability of standard desktop computers. In this work we present coagSODA, a CUDA-powered computational tool that was purposely developed for the analysis of a large mechanistic model of the blood coagulation cascade (BCC), defined according to both mass-action kinetics and Hill functions. coagSODA allows the execution of parallel simulations of the dynamics of the BCC by automatically deriving the system of ordinary differential equations and then exploiting the numerical integration algorithm LSODA. We present the biological results achieved with a massive exploration of perturbed conditions of the BCC, carried out with one-dimensional and bi-dimensional parameter sweep analysis, and show that GPU-accelerated parallel simulations of this model can increase the computational performances up to a 181× speedup compared to the corresponding sequential simulations. link: http://identifiers.org/doi/10.1155/2014/863298

Cellière2011 - Plasticity of TGF-β Signalling: BIOMD0000000600v0.0.1

Cellière2011 - Plasticity of TGF-β SignallingTransforming growth factor beta (TGF-β) signalling has been implicated as a…

Details

BACKGROUND: The family of TGF-β ligands is large and its members are involved in many different signaling processes. These signaling processes strongly differ in type with TGF-β ligands eliciting both sustained or transient responses. Members of the TGF-β family can also act as morphogen and cellular responses would then be expected to provide a direct read-out of the extracellular ligand concentration. A number of different models have been proposed to reconcile these different behaviours. We were interested to define the set of minimal modifications that are required to change the type of signal processing in the TGF-β signaling network. RESULTS: To define the key aspects for signaling plasticity we focused on the core of the TGF-β signaling network. With the help of a parameter screen we identified ranges of kinetic parameters and protein concentrations that give rise to transient, sustained, or oscillatory responses to constant stimuli, as well as those parameter ranges that enable a proportional response to time-varying ligand concentrations (as expected in the read-out of morphogens). A combination of a strong negative feedback and fast shuttling to the nucleus biases signaling to a transient rather than a sustained response, while oscillations were obtained if ligand binding to the receptor is weak and the turn-over of the I-Smad is fast. A proportional read-out required inefficient receptor activation in addition to a low affinity of receptor-ligand binding. We find that targeted modification of single parameters suffices to alter the response type. The intensity of a constant signal (i.e. the ligand concentration), on the other hand, affected only the strength but not the type of the response. CONCLUSIONS: The architecture of the TGF-β pathway enables the observed signaling plasticity. The observed range of signaling outputs to TGF-β ligand in different cell types and under different conditions can be explained with differences in cellular protein concentrations and with changes in effective rate constants due to cross-talk with other signaling pathways. It will be interesting to uncover the exact cellular differences as well as the details of the cross-talks in future work. link: http://identifiers.org/pubmed/22051045

Parameters:

NameDescription
k19 = 4.12E-4Reaction: I_Smad =>, Rate Law: c*k19*I_Smad
k13 = 0.00164Reaction: Smad_P_N => Smad_N, Rate Law: n*k13*Smad_P_N
k10 = 5.12E-8Reaction: Smad_P + CoSmad => Smad_P_CoSmad, Rate Law: c*k10*Smad_P*CoSmad
k7 = 9.35E-6Reaction: Smad => Smad_P; TGFb_TGFbR_P, Smad, Rate Law: c*k7*Smad*TGFb_TGFbR_P
k3 = 0.324Reaction: TGFb_TGFbR => TGFb_TGFbR_P, Rate Law: c*k3*TGFb_TGFbR
k5 = 5.49E-4Reaction: TGFb_TGFbR_P + I_Smad => I_Smad_TGFb_TGFbR_P, Rate Law: c*k5*TGFb_TGFbR_P*I_Smad
k11 = 0.00923Reaction: Smad_P_Smad_P => Smad_P, Rate Law: c*k11*Smad_P_Smad_P
k1 = 0.00446Reaction: TGFb_TGFbR => TGFbR, Rate Law: c*k1*TGFb_TGFbR
k8 = 0.0104; k12 = 0.0513Reaction: Smad_P_Smad_P => Smad_P_Smad_P_N, Rate Law: k12*k8*Smad_P_Smad_P
k6 = 1.29E-5Reaction: I_Smad_TGFb_TGFbR_P => TGFb_TGFbR + I_Smad, Rate Law: c*k6*I_Smad_TGFb_TGFbR_P
k2 = 4.39E-6Reaction: TGFbR + TGFb => TGFb_TGFbR, Rate Law: k2*TGFbR*TGFb
k14 = 0.038; k15 = 28.52; h = 2.06Reaction: => I_Smad_mRNA1; Smad_P_CoSmad_N, Rate Law: n*k14*Smad_P_CoSmad_N^h/(Smad_P_CoSmad_N^h+k15^h)
k4 = 0.00192Reaction: TGFb_TGFbR_P => TGFb_TGFbR, Rate Law: c*k4*TGFb_TGFbR_P
k17 = 8.05E-5Reaction: I_Smad_mRNA2 =>, Rate Law: c*k17*I_Smad_mRNA2
k18 = 0.0434Reaction: => I_Smad; I_Smad_mRNA2, Rate Law: c*k18*I_Smad_mRNA2
k9 = 7.5E-4Reaction: Smad_N => Smad, Rate Law: k9*Smad_N
k8 = 0.0104Reaction: Smad => Smad_N, Rate Law: k8*Smad
k16 = 0.0214Reaction: I_Smad_mRNA1 => I_Smad_mRNA2, Rate Law: k16*I_Smad_mRNA1

States:

NameDescription
TGFb[TGF-beta receptor type-1]
Smad P CoSmad[IPR008984; Mothers against decapentaplegic homolog 4]
Smad P Smad P N[IPR008984]
TGFb TGFbR P[IPR000333; IPR016319]
Smad P CoSmad N[IPR008984; Mothers against decapentaplegic homolog 4]
CoSmad[IPR008984]
I Smad mRNA2[IPR008984]
I Smad TGFb TGFbR P[IPR017855; IPR016319; IPR000333]
Smad P[IPR008984]
Smad P N[IPR008984; Mothers against decapentaplegic homolog 4]
I Smad mRNA1[IPR008984]
Smad P Smad P[IPR008984]
TGFbR[IPR000333]
Smad[IPR008984]
Smad N[IPR008984]
CoSmad N[IPR008984]
I Smad[IPR008984]
TGFb TGFbR[IPR000333; IPR016319]

Cetinkaya2017 - Engineering targets for Komagataella phaffii: MODEL1703150000v0.0.1

Cetinkaya2017 - Engineering targets for Komagataella phaffiiThis model is described in the article: [Metabolic modellin…

Details

Genome-scale metabolic models are valuable tools for the design of novel strains of industrial microorganisms, such as Komagataella phaffii (syn. Pichia pastoris). However, as is the case for many industrial microbes, there is no executable metabolic model for K. phaffiii that confirms to current standards by providing the metabolite and reactions IDs, to facilitate model extension and reuse, and gene-reaction associations to enable identification of targets for genetic manipulation. In order to remedy this deficiency, we decided to reconstruct the genome-scale metabolic model of K. phaffii by reconciling the extant models and performing extensive manual curation in order to construct an executable model (Kp.1.0) that conforms to current standards. We then used this model to study the effect of biomass composition on the predictive success of the model. Twelve different biomass compositions obtained from published empirical data obtained under a range of growth conditions were employed in this investigation. We found that the success of Kp1.0 in predicting both gene essentiality and growth characteristics was relatively unaffected by biomass composition. However, we found that biomass composition had a profound effect on the distribution of the fluxes involved in lipid, DNA and steroid biosynthetic processes, cellular alcohol metabolic process and oxidation-reduction process. Further, we investigated the effect of biomass composition on the identification of suitable target genes for strain development. The analyses revealed that around 40% of the predictions of the effect of gene overexpression or deletion changed depending on the representation of biomass composition in the model. Considering the robustness of the in silico flux distributions to the changing biomass representations enables better interpretation of experimental results, reduces the risk of wrong target identification, and so both speeds and improves the process of directed strain development. link: http://identifiers.org/doi/10.1002/bit.26380

Chakrabarty2010 - A control theory approach to cancer remission aided by an optimal therapy: BIOMD0000000777v0.0.1

This is a reinvestigation of a previous model depicting cancer remission. It involves application of mathematical tools…

Details

The mathematical model depicting cancer remission as presented by Banerjee and Sarkar1 is reinvestigated here. Mathematical tools from control theory have been used to analyze and determine how an optimal external treatment of Adaptive Cellular Immunotherapy and interleukin-2 can result in more effective remission of malignant tumors while minimizing any adverse affect on the immune response. link: http://identifiers.org/doi/10.1142/S0218339010003226

Parameters:

NameDescription
d_1 = 0.0412Reaction: N_CTL =>, Rate Law: compartment*d_1*N_CTL
mu_2 = 0.0Reaction: => N_CTL; N_CTL, Z_THL, Rate Law: compartment*mu_2*N_CTL*Z_THL
mu_1 = 0.05Reaction: M_Tumor_Cells =>, Rate Law: compartment*mu_1*M_Tumor_Cells
alpha_2 = 3.422E-10Reaction: N_CTL => ; M_Tumor_Cells, Rate Law: compartment*alpha_2*M_Tumor_Cells*N_CTL
r_2 = 0.0245; k_2 = 1.0E7Reaction: => Z_THL; Z_THL, Rate Law: compartment*r_2*Z_THL*(1-Z_THL/k_2)
k_1 = 5000000.0; r_1 = 0.18Reaction: => M_Tumor_Cells; M_Tumor_Cells, Rate Law: compartment*r_1*M_Tumor_Cells*(1-M_Tumor_Cells/k_1)
alpha_1 = 1.101E-7Reaction: M_Tumor_Cells => ; M_Tumor_Cells, N_CTL, Rate Law: compartment*alpha_1*M_Tumor_Cells*N_CTL

States:

NameDescription
Z THL[helper T cell]
M Tumor Cells[Neoplastic Cell]
N CTL[cytotoxic T cell]

Chan2004_TCell_receptor_activation: BIOMD0000000120v0.0.1

The model reproduces Fig 3a of the paper. Please note that the authors mention that they used a value of 2 for n, n bein…

Details

The specificity and sensitivity of T-cell recognition is vital to the immune response. Ligand engagement with the T-cell receptor (TCR) results in the activation of a complex sequence of signalling events, both on the cell membrane and intracellularly. Feedback is an integral part of these signalling pathways, yet is often ignored in standard accounts of T-cell signalling. Here we show, using a mathematical model, that these feedback loops can explain the ability of the TCR to discriminate between ligands with high specificity and sensitivity, as well as provide a mechanism for sustained signalling. The model also explains the recent counter-intuitive observation that endogenous 'null' ligands can significantly enhance T-cell signalling. Finally, the model may provide an archetype for receptor switching based on kinase-phosphatase switches, and thus be of interest to the wider signalling community. link: http://identifiers.org/pubmed/15255048

Parameters:

NameDescription
k2 = 0.01 sec_invReaction: phosphatase_inactive => phosphatase_active, Rate Law: k2*phosphatase_inactive
d2 = 0.0 sec_invReaction: phosphatase_active =>, Rate Law: d2*phosphatase_active
k1 = 0.01 sec_invReaction: lck_inactive => lck_active, Rate Law: k1*lck_inactive
r_l = 0.0 items_per_timeReaction: => lck_inactive, Rate Law: r_l
n1 = 1.0 per_sec_per_itemReaction: lck_active => lck_inactive; phosphatase_active, Rate Law: n1*lck_active*phosphatase_active
d1 = 0.15 sec_invReaction: lck_active =>, Rate Law: d1*lck_active
m2 = 1.0 per_sec_per_itemReaction: phosphatase_inactive => phosphatase_active; lck_active, Rate Law: m2*lck_active*phosphatase_inactive
n2 = 0.02 sec_invReaction: phosphatase_active => phosphatase_inactive, Rate Law: n2*phosphatase_active
d0 = 0.15 sec_invReaction: lck_inactive =>, Rate Law: d0*lck_inactive
m1 = 1.0; n = 1.95 dimensionlessReaction: lck_inactive => lck_active, Rate Law: m1*lck_active^n*lck_inactive

States:

NameDescription
lck total[Tyrosine-protein kinase Lck]
phosphatase active[Tyrosine-protein phosphatase non-receptor type 6]
lck active[Tyrosine-protein kinase Lck]
lck inactive[Tyrosine-protein kinase Lck]
phosphatase inactive[Tyrosine-protein phosphatase non-receptor type 6]

Chance1943_Peroxidase_ES_Kinetics: BIOMD0000000283v0.0.1

Default parameter values are those in the right hand panel of Fig 12. The other panels may be obtained by setting X to 1…

Details

Under the narrow range of experimental conditions, and at a temperature of approximately 25 degrees, the following data were obtained. 1. The equilibrium constant of peroxidase and hydrogen peroxide has a minimum value of 2 x 10(-8). 2. The velocity constant for the formation of peroxidase-H2O2 Complex I is 1.2 x 10(7) liter mole-1 sec.-1, +/- 0.4 x 10(7). 3. The velocity constant for the reversible breakdown of peroxidase-H2O2 Complex I is a negligible factor in the enzyme-substrate kinetics and is calculated to be less than 0.2 sec.-1. 4. The velocity constant, k3, for the enzymatic breakdown of peroxidase-H2O2 Complex I varies from nearly zero to higher than 5 sec.-1, depending upon the acceptor and its concentration. The quotient of k3 and the leucomalachite green concentration is 3.0 x 10(4) liter mole-1 sec.-1. For ascorbic acid this has a value of 1.8 x 10(5) liter mole-1 sec.-1. 5. For a particular acceptor concentration, k3 is determined solely from the enzyme-substrate kinetics and is found to be 4.2 sec.-1. 6. For the same conditions, k3 is determined from a simple relationship derived from mathematical solutions of the Michaelis theory and is found to be 5.2 sec.-1. 7. For the same conditions, k3 is determined from the over-all enzyme action and is found to be 5.1 sec.-1. 8. The Michaelis constant determined from kinetic data alone is found to be 0.44 x 10(-6). 9. The Michaelis constant determined from steady state measurements is found to be 0.41 x 10(-6). 10. The Michaelis constant determined from measurement of the overall enzyme reaction is found to be 0.50 x 10(-6). 11. The kinetics of the enzyme-substrate compound closely agree with mathematical solutions of an extension of the Michaelis theory obtained for experimental values of concentrations and reaction velocity constants. 12. The adequacy of the criteria by which experiment and theory were correlated has been examined critically and the mathematical solutions have been found to be sensitive to variations in the experimental conditions. 13. The critical features of the enzyme-substrate kinetics are Pmax, and curve shape, rather than t1/2. t1/2 serves as a simple measure of dx/dt. 14. A second order combination of enzyme and substrate to form the enzyme-substrate compound, followed by a first order breakdown of the compound, describes the activity of peroxidase for a particular acceptor concentration. 15. The kinetic data indicate a bimolecular combination of acceptor and enzyme-substrate compound. link: http://identifiers.org/pubmed/10218104

Parameters:

NameDescription
K3 = 0.5 dimensionlessReaction: P => E + Q, Rate Law: cell*K3*P
K2 = 0.0 dimensionlessReaction: X + E => P, Rate Law: cell*(E*X-K2*P)

States:

NameDescription
Q[water]
X[hydrogen peroxide; Hydrogen peroxide]
P[hydrogen peroxide; IPR000823]
E[Peroxidase C1A; Peroxidase C1C; Peroxidase C1B; IPR000823]

Chance1952_Catalase_Mechanism: BIOMD0000000282v0.0.1

This model is described in the article: **The mechanism of catalase action. II. Electric analog computer studies.** Br…

Details

link: http://identifiers.org/pubmed/14953444

Parameters:

NameDescription
k2 = 0.0 per second; k1 = 11.0 per micromolar per secondReaction: e + x => p, Rate Law: cell*(k1*e*x-k2*p)
k4_prime = 16.6 per micromolar per secondReaction: p + x => e + p1, Rate Law: cell*k4_prime*p*x
k4 = 0.72 per micromolar per secondReaction: p + a => e + p2, Rate Law: cell*k4*p*a

States:

NameDescription
e[catalase activity; Catalase; IPR002226; Catalase]
x[hydrogen peroxide; Hydrogen peroxide]
p2[water; carbonyl compound]
p1[water; dioxygen]
a[primary alcohol; secondary alcohol]
p[hydrogen peroxide; IPR002226]

Chance1960_Glycolysis_Respiration: BIOMD0000000281v0.0.1

This model is described inthe article: **Metabolic control mechanisms. 5. A solution for the equations representing in…

Details

link: http://identifiers.org/pubmed/13692276

Parameters:

NameDescription
k=5.0E9 per molar per secondReaction: PGA + ADP => TP1 + PYR, Rate Law: cell*1E-6*k*PGA*ADP
k=1.0E7 per molar per secondReaction: LAC + DPN => PYR + DPH, Rate Law: cell*1E-6*k*LAC*DPN
k=1.2E8 per molar per secondReaction: XSI => XI; DBP, Rate Law: cell*1E-6*k*XSI*DBP
k=4000000.0 per molar per secondReaction: TP2 => TP1; DBP, Rate Law: cell*1E-6*k*TP2*DBP
k=2000000.0 per secondReaction: PPP => ADP + PUE + PID, Rate Law: cell*1E-6*k*PPP
k=1.0E10 per molar per secondReaction: ENG + TP1 => ADP + GLP + ENZ, Rate Law: cell*1E-6*k*ENG*TP1
k=2.0E9 per molar per secondReaction: DHA + DPH => AGP + DPN, Rate Law: cell*1E-6*k*DHA*DPH
k=6.0E11 per molar per secondReaction: GAP + MOD => MOB + DPH, Rate Law: cell*1E-6*k*GAP*MOD
k=8.0E7 per molar per secondReaction: AGP + DPN => DHA + DPH, Rate Law: cell*1E-6*k*AGP*DPN
k=5.0E8 per molar per secondReaction: PYR + DPH => LAC + DPN, Rate Law: cell*1E-6*k*PYR*DPH
k=6.0E9 per molar per secondReaction: MOX + DPN => MOD, Rate Law: cell*1E-6*k*MOX*DPN
k=100000.0 per secondReaction: GPP => GAP + DHA, Rate Law: cell*1E-6*k*GPP
k=7.5E12 per molar squared per secondReaction: DIH + XI + OXY => XSI + DIN, Rate Law: cell*1E-6*k*DIH*XI*OXY
k=1.5E10 per molar per secondReaction: XSP + ADP => TP2 + XI, Rate Law: cell*1E-6*k*XSP*ADP
k=4.0E10 per molar per secondReaction: ETG + TP1 => GPP + ETZ + ADP, Rate Law: cell*1E-6*k*ETG*TP1
k=2.0E7 per molar per secondReaction: PYR + DIN => DIH, Rate Law: cell*1E-6*k*PYR*DIN
k=4.0E8 per molar per secondReaction: MOB + PID => DGA + MOX, Rate Law: cell*1E-6*k*MOB*PID
k=3.0E9 per molar per secondReaction: GLU + ENZ => ENG, Rate Law: cell*1E-6*k*GLU*ENZ

States:

NameDescription
ENG[glucose; IPR001312]
DPN[NAD(+); NAD+]
PID[phosphate(3-); Orthophosphate]
TP1[ATP; ATP]
MOB[D-glyceraldehyde 3-phosphate; IPR006424; glyceraldehyde-3-phosphate dehydrogenase (NAD+) (phosphorylating) activity]
GPP[beta-D-Fructose 1,6-bisphosphate; beta-D-fructofuranose 1,6-bisphosphate]
DGA[3-Phospho-D-glyceroyl phosphate; 3-phospho-D-glyceroyl dihydrogen phosphate]
PGA[3-Phospho-D-glycerate; 3-phospho-D-glyceric acid]
GLP[D-glucopyranose 6-phosphate; D-Glucose 6-phosphate]
ETZ[IPR022463; 1-phosphofructokinase activity]
MOX[Glyceraldehyde-3-phosphate dehydrogenase; Glyceraldehyde-3-phosphate dehydrogenase; IPR006424; glyceraldehyde-3-phosphate dehydrogenase (NAD+) (phosphorylating) activity]
DHA[Glycerone phosphate; dihydroxyacetone phosphate]
TP2[ATP; ATP]
MOD[NAD(+); IPR006424]
XSP[mitochondrial respiratory chain; proton-transporting ATP synthase complex]
PYR[Pyruvate; pyruvic acid]
DPH[NADH; NADH]
PPP[ATP; protein polypeptide chain; ATPase activity]
DIN[NAD+; NAD(+)]
AGP[sn-Glycerol 1-phosphate; sn-glycerol 1-phosphate]
DIH[NADH; NADH]
XSI[proton-transporting ATP synthase complex; mitochondrial respiratory chain]
OXY[Oxygen; dioxygen]
GAP[D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]
ETG[D-glucopyranose 6-phosphate; IPR022463; 1-phosphofructokinase activity]
LAC[(S)-Lactate; (S)-lactic acid]
ENZ[IPR001312; hexokinase activity]
XI[proton-transporting ATP synthase complex; mitochondrial respiratory chain]
ADP[ADP; ADP]
GLU[C00293; glucose]
PUE[Protein; protein polypeptide chain]

Chang2008 - ERK activation, hallucinogenic drugs mediated signalling through serotonin receptors: MODEL0975191032v0.0.1

Chang2008 - ERK activation, hallucinogenic drugs mediated signalling through serotonin receptorsThis model is described…

Details

Through a multidisciplinary approach involving experimental and computational studies, we address quantitative aspects of signaling mechanisms triggered in the cell by the receptor targets of hallucinogenic drugs, the serotonin 5-HT2A receptors. To reveal the properties of the signaling pathways, and the way in which responses elicited through these receptors alone and in combination with other serotonin receptors' subtypes (the 5-HT1AR), we developed a detailed mathematical model of receptor-mediated ERK1/2 activation in cells expressing the 5-HT1A and 5-HT2A subtypes individually, and together. In parallel, we measured experimentally the activation of ERK1/2 by the action of selective agonists on these receptors expressed in HEK293 cells. We show here that the 5-HT1AR agonist Xaliproden HCl elicited transient activation of ERK1/2 by phosphorylation, whereas 5-HT2AR activation by TCB-2 led to higher, and more sustained responses. The 5-HT2AR response dominated the MAPK signaling pathway when co-expressed with 5-HT1AR, and diminution of the response by the 5-HT2AR antagonist Ketanserin could not be rescued by the 5-HT1AR agonist. Computational simulations produced qualitative results in good agreement with these experimental data, and parameter optimization made this agreement quantitative. In silico simulation experiments suggest that the deletion of the positive regulators PKC in the 5-HT2AR pathway, or PLA2 in the combined 5-HT1A/2AR model greatly decreased the basal level of active ERK1/2. Deletion of negative regulators of MKP and PP2A in 5-HT1AR and 5-HT2AR models was found to have even stronger effects. Under various parameter sets, simulation results implied that the extent of constitutive activity in a particular tissue and the specific drug efficacy properties may determine the distinct dynamics of the 5-HT receptor-mediated ERK1/2 activation pathways. Thus, the mathematical models are useful exploratory tools in the ongoing efforts to establish a mechanistic understanding and an experimentally testable representation of hallucinogen-specific signaling in the cellular machinery, and can be refined with quantitative, function-related information. link: http://identifiers.org/pubmed/18762202

Chang2010_Reduced_Kidney_FBA: MODEL1011080004v0.0.1

This is the reduced kidney metabolic network described in the article Drug off-target effects predicted using structur…

Details

Recent advances in structural bioinformatics have enabled the prediction of protein-drug off-targets based on their ligand binding sites. Concurrent developments in systems biology allow for prediction of the functional effects of system perturbations using large-scale network models. Integration of these two capabilities provides a framework for evaluating metabolic drug response phenotypes in silico. This combined approach was applied to investigate the hypertensive side effect of the cholesteryl ester transfer protein inhibitor torcetrapib in the context of human renal function. A metabolic kidney model was generated in which to simulate drug treatment. Causal drug off-targets were predicted that have previously been observed to impact renal function in gene-deficient patients and may play a role in the adverse side effects observed in clinical trials. Genetic risk factors for drug treatment were also predicted that correspond to both characterized and unknown renal metabolic disorders as well as cryptic genetic deficiencies that are not expected to exhibit a renal disorder phenotype except under drug treatment. This study represents a novel integration of structural and systems biology and a first step towards computational systems medicine. The methodology introduced herein has important implications for drug development and personalized medicine. link: http://identifiers.org/pubmed/20957118

Chang2011_MetabolicNetworkReconstruction_ChlamydomonasReinhardtii: MODEL1106200000v0.0.1

This model is from the article: Metabolic network reconstruction of Chlamydomonas offers insight into light-driven alg…

Details

Metabolic network reconstruction encompasses existing knowledge about an organism's metabolism and genome annotation, providing a platform for omics data analysis and phenotype prediction. The model alga Chlamydomonas reinhardtii is employed to study diverse biological processes from photosynthesis to phototaxis. Recent heightened interest in this species results from an international movement to develop algal biofuels. Integrating biological and optical data, we reconstructed a genome-scale metabolic network for this alga and devised a novel light-modeling approach that enables quantitative growth prediction for a given light source, resolving wavelength and photon flux. We experimentally verified transcripts accounted for in the network and physiologically validated model function through simulation and generation of new experimental growth data, providing high confidence in network contents and predictive applications. The network offers insight into algal metabolism and potential for genetic engineering and efficient light source design, a pioneering resource for studying light-driven metabolism and quantitative systems biology. link: http://identifiers.org/pubmed/21811229

Chaouiya2013 - EGF and TNFalpha mediated signalling pathway: BIOMD0000000562v0.0.1

Chaouiya2013 - EGF and TNFalpha mediated signalling pathwayThis model is described in the article: [SBML qualitative mo…

Details

Qualitative frameworks, especially those based on the logical discrete formalism, are increasingly used to model regulatory and signalling networks. A major advantage of these frameworks is that they do not require precise quantitative data, and that they are well-suited for studies of large networks. While numerous groups have developed specific computational tools that provide original methods to analyse qualitative models, a standard format to exchange qualitative models has been missing.We present the Systems Biology Markup Language (SBML) Qualitative Models Package ("qual"), an extension of the SBML Level 3 standard designed for computer representation of qualitative models of biological networks. We demonstrate the interoperability of models via SBML qual through the analysis of a specific signalling network by three independent software tools. Furthermore, the collective effort to define the SBML qual format paved the way for the development of LogicalModel, an open-source model library, which will facilitate the adoption of the format as well as the collaborative development of algorithms to analyse qualitative models.SBML qual allows the exchange of qualitative models among a number of complementary software tools. SBML qual has the potential to promote collaborative work on the development of novel computational approaches, as well as on the specification and the analysis of comprehensive qualitative models of regulatory and signalling networks. link: http://identifiers.org/pubmed/24321545

Chareyron2009 - Mixed immunotherapy and chemotherapy of tumors Feedback design and model updating schemes: MODEL2001160002v0.0.1

Mixed immunotherapy and chemotherapy of tumors: feedback design and model updating schemes. Chareyron S1, Alamir M. Auth…

Details

In this paper, a recently developed model governing the cancer growth on a cell population level with combination of immune and chemotherapy is used to develop a reactive (feedback) mixed treatment strategy. The feedback design proposed here is based on nonlinear constrained model predictive control together with an adaptation scheme that enables the effects of unavoidable modeling uncertainties to be compensated. The effectiveness of the proposed strategy is shown under realistic human data showing the advantage of treatment in feedback form as well as the relevance of the adaptation strategy in handling uncertainties and modeling errors. A new treatment strategy defined by an original optimal control problem formulation is also proposed. This new formulation shows particularly interesting possibilities since it may lead to tumor regression under better health indicator profile. link: http://identifiers.org/pubmed/18655792

Chassagnole2001_Threonine Synthesis: BIOMD0000000066v0.0.1

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

Details

A computer simulation of the threonine-synthesis pathway in Escherichia coli Tir-8 has been developed based on our previous measurements of the kinetics of the pathway enzymes under near-physiological conditions. The model successfully simulates the main features of the time courses of threonine synthesis previously observed in a cell-free extract without alteration of the experimentally determined parameters, although improved quantitative fits can be obtained with small parameter adjustments. At the concentrations of enzymes, precursors and products present in cells, the model predicts a threonine-synthesis flux close to that required to support cell growth. Furthermore, the first two enzymes operate close to equilibrium, providing an example of a near-equilibrium feedback-inhibited enzyme. The predicted flux control coefficients of the pathway enzymes under physiological conditions show that the control of flux is shared between the first three enzymes: aspartate kinase, aspartate semialdehyde dehydrogenase and homoserine dehydrogenase, with no single activity dominating the control. The response of the model to the external metabolites shows that the sharing of control between the three enzymes holds across a wide range of conditions, but that the pathway flux is sensitive to the aspartate concentration. When the model was embedded in a larger model to simulate the variable demands for threonine at different growth rates, it showed the accumulation of free threonine that is typical of the Tir-8 strain at low growth rates. At low growth rates, the control of threonine flux remains largely with the pathway enzymes. As an example of the predictive power of the model, we studied the consequences of over-expressing different enzymes in the pathway. link: http://identifiers.org/pubmed/11368770

Parameters:

NameDescription
vm5=0.0434 mM_per_min; k5hsp=0.31 mMReaction: hsp => thr + phos, Rate Law: compartment*vm5*hsp/(hsp+k5hsp)
katpase=4.1E-5 millimole_per_mg_per_min; prot=202.0 mg_per_litreReaction: atp => adp + phos, Rate Law: compartment*prot*katpase
knadph=5.4E-6 litre_per_mg_per_min; prot=202.0 mg_per_litreReaction: nadph => nadp, Rate Law: compartment*prot*knadph*nadph
k13atp=0.22 mM; k13aspp=0.017 mM; k1lys=0.391 mM; k1aspp=0.017 mM; k1thr=0.167 mM; k13=0.32 mM; k1atp=0.98 mM; keqak=6.4E-4 dimensionless; vm13=0.0722 mM_per_min; nak3=2.8 dimensionless; nak1=4.09 dimensionless; k13adp=0.25 mM; alpha=2.47 dimensionless; k1adp=0.25 mM; lys=0.46 mM; vm11=0.15 mM_per_min; k11=0.97 mMReaction: atp + asp => aspp + adp; thr, Rate Law: compartment*(vm11*(asp*atp-aspp*adp/keqak)/((k11*(1+(thr/k1thr)^nak1)/(1+(thr/(alpha*k1thr))^nak1)+k11*aspp/k1aspp+asp)*(k1atp*(1+adp/k1adp)+atp))+vm13*(asp*atp-aspp*adp/keqak)/((1+(lys/k1lys)^nak3)*(k13*(1+aspp/k13aspp)+asp)*(k13atp*(1+adp/k13adp)+atp)))
k3thr=0.097 mM; k3hs=3.39 mM; alpha3=3.93 dimensionless; k3nadph=0.037 mM; k3eq=3162.27766016838 dimensionless; k3nadp=0.067 mM; vm3f=1.001 mM_per_min; nhdh1=1.41 dimensionless; k3asa=0.24 mMReaction: nadph + asa => hs + nadp; asp, thr, Rate Law: compartment*vm3f*(asa*nadph-hs*nadp/k3eq)/((1+(thr/k3thr)^nhdh1)/(1+(thr/(alpha3*k3thr))^nhdh1)*(k3asa+asa+hs*k3asa/k3hs)*(k3nadph*(1+nadp/k3nadp)+nadph))
k4lys=9.45 mM; k4thr=1.09 mM; k4atp=0.072 mM; k4ihs=4.7 mM; vm4f=0.1 mM_per_min; k4hs=0.11 mM; k4iatp=4.35 mM; lys=0.46 mMReaction: hs + atp => hsp + adp; thr, Rate Law: compartment*vm4f*hs*atp/((1+lys/k4lys)*(atp+k4atp*(1+hs/k4ihs))*(hs+k4hs*(1+thr/k4thr)*(1+atp/k4iatp)))
k2p=10.0 mM; k2nadph=0.029 mM; k2nadp=0.144 mM; k2asa=0.11 mM; k2eq=56.4150334574039 dimensionless; k2aspp=0.022 mM; vm2f=0.1812 mM_per_minReaction: nadph + aspp => nadp + phos + asa, Rate Law: compartment*vm2f*(aspp*nadph-asa*nadp*phos/k2eq)/((k2aspp*(1+asa/k2asa)*(1+phos/k2p)+aspp)*(k2nadph*(1+nadp/k2nadp)+nadph))

States:

NameDescription
nadph[NADPH; NADPH]
hsp[O-phospho-L-homoserine; O-Phospho-L-homoserine]
asa[L-aspartic 4-semialdehyde; L-Aspartate 4-semialdehyde]
hs[L-homoserine; L-Homoserine]
atp[ATP; ATP]
asp[L-aspartic acid; L-Aspartate]
thr[L-threonine; L-Threonine]
adp[ADP; ADP]
aspp[4-phospho-L-aspartic acid; 4-Phospho-L-aspartate]
nadp[NADP(+); NADP+]
phos[phosphate(3-); Orthophosphate]

Chassagnole2002_Carbon_Metabolism: BIOMD0000000051v0.0.1

The model reproduces Figures 4,5 and 6 of the publication. The analytical functions for cometabolites Catp, Camp, Cnadph…

Details

Application of metabolic engineering principles to the rational design of microbial production processes crucially depends on the ability to describe quantitatively the systemic behavior of the central carbon metabolism to redirect carbon fluxes to the product-forming pathways. Despite the importance for several production processes, development of an essential dynamic model for central carbon metabolism of Escherichia coli has been severely hampered by the current lack of kinetic information on the dynamics of the metabolic reactions. Here we present the design and experimental validation of such a dynamic model, which, for the first time, links the sugar transport system (i.e., phosphotransferase system [PTS]) with the reactions of glycolysis and the pentose-phosphate pathway. Experimental observations of intracellular concentrations of metabolites and cometabolites at transient conditions are used to validate the structure of the model and to estimate the kinetic parameters. Further analysis of the detailed characteristics of the system offers the possibility of studying important questions regarding the stability and control of metabolic fluxes. link: http://identifiers.org/pubmed/17590932

Parameters:

NameDescription
KGAPDHgap=0.683 milli Molar; cnad = 1.47 milli Molar; KGAPDHnad=0.252 milli Molar; KGAPDHnadh=1.09 milli Molar; cnadh = 0.1 milli Molar; KGAPDHpgp=1.04E-5 milli Molar; rmaxGAPDH=921.5942861 mM per second; KGAPDHeq=0.63 dimensionlessReaction: cgap => cpgp, Rate Law: cytosol*rmaxGAPDH*(cgap*cnad-cpgp*cnadh/KGAPDHeq)/((KGAPDHgap*(1+cpgp/KGAPDHpgp)+cgap)*(KGAPDHnad*(1+cnadh/KGAPDHnadh)+cnad))
VALDOblf=2.0 dimensionless; kALDOgapinh=0.6 milli Molar; kALDOeq=0.144 milli Molar; kALDOfdp=1.75 milli Molar; rmaxALDO=17.41464425 mM per second; kALDOgap=0.088 milli Molar; kALDOdhap=0.088 milli MolarReaction: cfdp => cdhap + cgap, Rate Law: cytosol*rmaxALDO*(cfdp-cgap*cdhap/kALDOeq)/(kALDOfdp+cfdp+kALDOgap*cdhap/(kALDOeq*VALDOblf)+kALDOdhap*cgap/(kALDOeq*VALDOblf)+cfdp*cgap/kALDOgapinh+cgap*cdhap/(VALDOblf*kALDOeq))
catp = 4.27 milli Molar; rmaxG1PAT=0.007525458026 mM per second; KG1PATg1p=3.2 milli Molar; KG1PATfdp=0.119 milli Molar; KG1PATatp=4.42 milli Molar; nG1PATfdp=1.2 milli MolarReaction: cg1p => ; cfdp, Rate Law: cytosol*rmaxG1PAT*cg1p*catp*(1+(cfdp/KG1PATfdp)^nG1PATfdp)/((KG1PATatp+catp)*(KG1PATg1p+cg1p))
rmaxR5PI=4.83841193 second inverse; KR5PIeq=4.0 dimensionlessReaction: cribu5p => crib5p, Rate Law: cytosol*rmaxR5PI*(cribu5p-crib5p/KR5PIeq)
cnadph = 0.062 milli Molar; KG6PDHnadp=0.0246 milli Molar; rmaxG6PDH=1.380196955 mM per second; KG6PDHg6p=14.4 milli Molar; cnadp = 0.195 milli Molar; KG6PDHnadphg6pinh=6.43 milli Molar; KG6PDHnadphnadpinh=0.01 milli MolarReaction: cg6p => cpg, Rate Law: cytosol*rmaxG6PDH*cg6p*cnadp/((cg6p+KG6PDHg6p)*(1+cnadph/KG6PDHnadphg6pinh)*(KG6PDHnadp*(1+cnadph/KG6PDHnadphnadpinh)+cnadp))
KG3PDHdhap=1.0 milli Molar; rmaxG3PDH=0.01162042696 mM per secondReaction: cdhap =>, Rate Law: cytosol*rmaxG3PDH*cdhap/(KG3PDHdhap+cdhap)
rmaxTA=10.87164108 per mM per second; KTAeq=1.05 dimensionlessReaction: cgap + csed7p => cf6p + ce4p, Rate Law: cytosol*rmaxTA*(cgap*csed7p-ce4p*cf6p/KTAeq)
KENOpep=0.135 milli Molar; KENOpg2=0.1 milli Molar; KENOeq=6.73 milli Molar; rmaxENO=330.4476151 mM per secondReaction: cpg2 => cpep, Rate Law: cytosol*rmaxENO*(cpg2-cpep/KENOeq)/(KENOpg2*(1+cpep/KENOpep)+cpg2)
KPFKf6ps=0.325 milli Molar; KPFKampa=19.1 milli Molar; nPFK=11.1 dimensionless; catp = 4.27 milli Molar; KPFKadpb=3.89 milli Molar; KPFKampb=3.2 milli Molar; KPFKadpc=4.14 milli Molar; KPFKatps=0.123 milli Molar; cadp = 0.595 milli Molar; rmaxPFK=1840.584747 mM per second; camp = 0.955 milli Molar; KPFKpep=3.26 milli Molar; KPFKadpa=128.0 milli Molar; LPFK=5629067.0 dimensionlessReaction: cf6p => cfdp; cpep, Rate Law: cytosol*rmaxPFK*catp*cf6p/((catp+KPFKatps*(1+cadp/KPFKadpc))*(cf6p+KPFKf6ps*(1+cpep/KPFKpep+cadp/KPFKadpb+camp/KPFKampb)/(1+cadp/KPFKadpa+camp/KPFKampa))*(1+LPFK/(1+cf6p*(1+cadp/KPFKadpa+camp/KPFKampa)/(KPFKf6ps*(1+cpep/KPFKpep+cadp/KPFKadpb+camp/KPFKampb)))^nPFK))
kTISeq=1.39 dimensionless; kTISdhap=2.8 milli Molar; rmaxTIS=68.67474392 mM per second; kTISgap=0.3 milli MolarReaction: cdhap => cgap, Rate Law: cytosol*rmaxTIS*(cdhap-cgap/kTISeq)/(kTISdhap*(1+cgap/kTISgap)+cdhap)
KPTSa2=0.01 milli Molar; nPTSg6p=3.66 dimensionless; rmaxPTS=7829.78 mM per second; KPTSa3=245.3 dimensionless; KPTSa1=3082.3 milli Molar; KPTSg6p=2.15 milli MolarReaction: cglcex + cpep => cg6p + cpyr, Rate Law: extracellular*rmaxPTS*cglcex*cpep/cpyr/((KPTSa1+KPTSa2*cpep/cpyr+KPTSa3*cglcex+cglcex*cpep/cpyr)*(1+cg6p^nPTSg6p/KPTSg6p))
KPGKeq=1934.4 dimensionless; KPGKadp=0.185 milli Molar; catp = 4.27 milli Molar; KPGKatp=0.653 milli Molar; cadp = 0.595 milli Molar; rmaxPGK=3021.773771 mM per second; KPGKpg3=0.473 milli Molar; KPGKpgp=0.0468 milli MolarReaction: cpgp => cpg3, Rate Law: cytosol*rmaxPGK*(cadp*cpgp-catp*cpg3/KPGKeq)/((KPGKadp*(1+catp/KPGKatp)+cadp)*(KPGKpgp*(1+cpg3/KPGKpg3)+cpgp))
rmaxMetSynth=0.0022627 mM per secondReaction: => cpyr, Rate Law: cytosol*rmaxMetSynth
rmaxMurSynth=4.3711E-4 mM per secondReaction: cf6p =>, Rate Law: cytosol*rmaxMurSynth
KRu5Peq=1.4 dimensionless; rmaxRu5P=6.739029475 second inverseReaction: cribu5p => cxyl5p, Rate Law: cytosol*rmaxRu5P*(cribu5p-cxyl5p/KRu5Peq)
npepCxylasefdp=4.21 dimensionless; rmaxpepCxylase=0.1070205858 mM per second; KpepCxylasefdp=0.7 milli Molar; KpepCxylasepep=4.07 milli MolarReaction: cpep => ; cfdp, Rate Law: cytosol*rmaxpepCxylase*cpep*(1+(cfdp/KpepCxylasefdp)^npepCxylasefdp)/(KpepCxylasepep+cpep)
rmaxTrpSynth=0.001037 mM per secondReaction: => cpyr + cgap, Rate Law: cytosol*rmaxTrpSynth
Dil=2.78E-5 second inverse; cfeed=110.96 milli MolarReaction: => cglcex, Rate Law: extracellular*Dil*(cfeed-cglcex)
KPGluMupg3=0.2 milli Molar; KPGluMueq=0.188 dimensionless; KPGluMupg2=0.369 milli Molar; rmaxPGluMu=89.04965407 mM per secondReaction: cpg3 => cpg2, Rate Law: cytosol*rmaxPGluMu*(cpg3-cpg2/KPGluMueq)/(KPGluMupg3*(1+cpg2/KPGluMupg2)+cpg3)
mu=2.78E-5 second inverseReaction: cg6p =>, Rate Law: cytosol*mu*cg6p
rmaxTKb=86.55855855 per mM per second; KTKbeq=10.0 dimensionlessReaction: ce4p + cxyl5p => cgap + cf6p, Rate Law: cytosol*rmaxTKb*(cxyl5p*ce4p-cf6p*cgap/KTKbeq)
LPK=1000.0 dimensionless; KPKatp=22.5 milli Molar; catp = 4.27 milli Molar; KPKpep=0.31 milli Molar; cadp = 0.595 milli Molar; rmaxPK=0.06113150238 mM per second; camp = 0.955 milli Molar; KPKfdp=0.19 milli Molar; KPKadp=0.26 milli Molar; KPKamp=0.2 milli Molar; nPK=4.0 dimensionlessReaction: cpep => cpyr; cfdp, Rate Law: cytosol*rmaxPK*cpep*(cpep/KPKpep+1)^(nPK-1)*cadp/(KPKpep*(LPK*((1+catp/KPKatp)/(cfdp/KPKfdp+camp/KPKamp+1))^nPK+(cpep/KPKpep+1)^nPK)*(cadp+KPKadp))
KPGIf6p=0.266 milli Molar; KPGIf6ppginh=0.2 milli Molar; KPGIg6ppginh=0.2 milli Molar; KPGIg6p=2.9 milli Molar; KPGIeq=0.1725 dimensionless; rmaxPGI=650.9878687 mM per secondReaction: cg6p => cf6p; cpg, Rate Law: cytosol*rmaxPGI*(cg6p-cf6p/KPGIeq)/(KPGIg6p*(1+cf6p/(KPGIf6p*(1+cpg/KPGIf6ppginh))+cpg/KPGIg6ppginh)+cg6p)
KSynth1pep=1.0 milli Molar; rmaxSynth1=0.01953897003 mM per secondReaction: cpep =>, Rate Law: cytosol*rmaxSynth1*cpep/(KSynth1pep+cpep)
KDAHPSpep=0.0053 milli Molar; KDAHPSe4p=0.035 milli Molar; nDAHPSpep=2.2 dimensionless; rmaxDAHPS=0.1079531227 mM per second; nDAHPSe4p=2.6 dimensionlessReaction: ce4p + cpep =>, Rate Law: cytosol*rmaxDAHPS*ce4p^nDAHPSe4p*cpep^nDAHPSpep/((KDAHPSe4p+ce4p^nDAHPSe4p)*(KDAHPSpep+cpep^nDAHPSpep))
KPDHpyr=1159.0 milli Molar; rmaxPDH=6.059531017 mM per second; nPDH=3.68 dimensionlessReaction: cpyr =>, Rate Law: cytosol*rmaxPDH*cpyr^nPDH/(KPDHpyr+cpyr^nPDH)
KSynth2pyr=1.0 milli Molar; rmaxSynth2=0.07361855055 mM per secondReaction: cpyr =>, Rate Law: cytosol*rmaxSynth2*cpyr/(KSynth2pyr+cpyr)
rmaxTKa=9.473384783 per mM per second; KTKaeq=1.2 dimensionlessReaction: crib5p + cxyl5p => cgap + csed7p, Rate Law: cytosol*rmaxTKa*(crib5p*cxyl5p-csed7p*cgap/KTKaeq)
rmaxSerSynth=0.025712107 mM per second; KSerSynthpg3=1.0 milli MolarReaction: cpg3 =>, Rate Law: cytosol*rmaxSerSynth*cpg3/(KSerSynthpg3+cpg3)
cnadph = 0.062 milli Molar; KPGDHpg=37.5 milli Molar; KPGDHatpinh=208.0 milli Molar; catp = 4.27 milli Molar; rmaxPGDH=16.23235977 mM per second; KPGDHnadp=0.0506 milli Molar; KPGDHnadphinh=0.0138 milli Molar; cnadp = 0.195 milli MolarReaction: cpg => cribu5p, Rate Law: cytosol*rmaxPGDH*cpg*cnadp/((cpg+KPGDHpg)*(cnadp+KPGDHnadp*(1+cnadph/KPGDHnadphinh)*(1+catp/KPGDHatpinh)))
rmaxRPPK=0.01290045226 mM per second; KRPPKrib5p=0.1 milli MolarReaction: crib5p =>, Rate Law: cytosol*rmaxRPPK*crib5p/(KRPPKrib5p+crib5p)
KPGMeq=0.196 dimensionless; rmaxPGM=0.8398242773 mM per second; KPGMg6p=1.038 milli Molar; KPGMg1p=0.0136 milli MolarReaction: cg6p => cg1p, Rate Law: cytosol*rmaxPGM*(cg6p-cg1p/KPGMeq)/(KPGMg6p*(1+cg1p/KPGMg1p)+cg6p)

States:

NameDescription
crib5p[aldehydo-D-ribose 5-phosphate; D-Ribose 5-phosphate]
cpg2[2-phospho-D-glyceric acid; 2-Phospho-D-glycerate]
cglcex[D-glucopyranose; D-Glucose; D-glucopyranose; glucose; C00293]
cpg[6-phospho-D-gluconate; 6-Phospho-D-gluconate]
cdhap[dihydroxyacetone phosphate; Glycerone phosphate]
cg1p[D-glucopyranose 1-phosphate; D-Glucose 1-phosphate]
cpep[phosphoenolpyruvate; Phosphoenolpyruvate]
cpg3[3-phospho-D-glyceric acid; 3-Phospho-D-glycerate]
cg6p[alpha-D-glucose 6-phosphate; alpha-D-Glucose 6-phosphate]
cpyr[pyruvate; Pyruvate]
ce4p[D-erythrose 4-phosphate; D-Erythrose 4-phosphate]
cribu5p[D-ribulose 5-phosphate; D-Ribulose 5-phosphate]
csed7p[sedoheptulose 7-phosphate; C00281]
cf6p[beta-D-Fructose 6-phosphate; beta-D-fructofuranose 6-phosphate; D-Fructose 6-phosphate; keto-D-fructose 6-phosphate; keto-D-fructose 6-phosphate]
cfdp[keto-D-fructose 1,6-bisphosphate; D-Fructose 1,6-bisphosphate]
cpgp[3-phospho-D-glyceroyl dihydrogen phosphate; 3-Phospho-D-glyceroyl phosphate]
cgap[glyceraldehyde 3-phosphate; Glyceraldehyde 3-phosphate]
cxyl5p[D-xylulose 5-phosphate; D-Xylulose 5-phosphate]

Chatterjee2010_BloodCoagulation: MODEL1108260014v0.0.1

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

Details

Blood function defines bleeding and clotting risks and dictates approaches for clinical intervention. Independent of adding exogenous tissue factor (TF), human blood treated in vitro with corn trypsin inhibitor (CTI, to block Factor XIIa) will generate thrombin after an initiation time (T(i)) of 1 to 2 hours (depending on donor), while activation of platelets with the GPVI-activator convulxin reduces T(i) to ∼20 minutes. Since current kinetic models fail to generate thrombin in the absence of added TF, we implemented a Platelet-Plasma ODE model accounting for: the Hockin-Mann protease reaction network, thrombin-dependent display of platelet phosphatidylserine, VIIa function on activated platelets, XIIa and XIa generation and function, competitive thrombin substrates (fluorogenic detector and fibrinogen), and thrombin consumption during fibrin polymerization. The kinetic model consisting of 76 ordinary differential equations (76 species, 57 reactions, 105 kinetic parameters) predicted the clotting of resting and convulxin-activated human blood as well as predicted T(i) of human blood under 50 different initial conditions that titrated increasing levels of TF, Xa, Va, XIa, IXa, and VIIa. Experiments with combined anti-XI and anti-XII antibodies prevented thrombin production, demonstrating that a leak of XIIa past saturating amounts of CTI (and not "blood-borne TF" alone) was responsible for in vitro initiation without added TF. Clotting was not blocked by antibodies used individually against TF, VII/VIIa, P-selectin, GPIb, protein disulfide isomerase, cathepsin G, nor blocked by the ribosome inhibitor puromycin, the Clk1 kinase inhibitor Tg003, or inhibited VIIa (VIIai). This is the first model to predict the observed behavior of CTI-treated human blood, either resting or stimulated with platelet activators. CTI-treated human blood will clot in vitro due to the combined activity of XIIa and XIa, a process enhanced by platelet activators and which proceeds in the absence of any evidence for kinetically significant blood borne tissue factor. link: http://identifiers.org/pubmed/20941387

Chavali2008 - Genome-scale metabolic network of Leishmania major (iAC560): MODEL1507180059v0.0.1

Chavali2008 - Genome-scale metabolic network of Leishmania major (iAC560)This model is described in the article: [Syste…

Details

Systems analyses have facilitated the characterization of metabolic networks of several organisms. We have reconstructed the metabolic network of Leishmania major, a poorly characterized organism that causes cutaneous leishmaniasis in mammalian hosts. This network reconstruction accounts for 560 genes, 1112 reactions, 1101 metabolites and 8 unique subcellular localizations. Using a systems-based approach, we hypothesized a comprehensive set of lethal single and double gene deletions, some of which were validated using published data with approximately 70% accuracy. Additionally, we generated hypothetical annotations to dozens of previously uncharacterized genes in the L. major genome and proposed a minimal medium for growth. We further demonstrated the utility of a network reconstruction with two proof-of-concept examples that yielded insight into robustness of the network in the presence of enzymatic inhibitors and delineation of promastigote/amastigote stage-specific metabolism. This reconstruction and the associated network analyses of L. major is the first of its kind for a protozoan. It can serve as a tool for clarifying discrepancies between data sources, generating hypotheses that can be experimentally validated and identifying ideal therapeutic targets. link: http://identifiers.org/pubmed/18364711

Chavez2009 - a core regulatory network of OCT4 in human embryonic stem cells: MODEL1305010000v0.0.1

Chavez2009 - a core regulatory network of OCT4 in human embryonic stem cellsA core OCT4-regulated network has been ident…

Details

BACKGROUND: The transcription factor OCT4 is highly expressed in pluripotent embryonic stem cells which are derived from the inner cell mass of mammalian blastocysts. Pluripotency and self renewal are controlled by a transcription regulatory network governed by the transcription factors OCT4, SOX2 and NANOG. Recent studies on reprogramming somatic cells to induced pluripotent stem cells highlight OCT4 as a key regulator of pluripotency. RESULTS: We have carried out an integrated analysis of high-throughput data (ChIP-on-chip and RNAi experiments along with promoter sequence analysis of putative target genes) and identified a core OCT4 regulatory network in human embryonic stem cells consisting of 33 target genes. Enrichment analysis with these target genes revealed that this integrative analysis increases the functional information content by factors of 1.3 - 4.7 compared to the individual studies. In order to identify potential regulatory co-factors of OCT4, we performed a de novo motif analysis. In addition to known validated OCT4 motifs we obtained binding sites similar to motifs recognized by further regulators of pluripotency and development; e.g. the heterodimer of the transcription factors C-MYC and MAX, a prerequisite for C-MYC transcriptional activity that leads to cell growth and proliferation. CONCLUSION: Our analysis shows how heterogeneous functional information can be integrated in order to reconstruct gene regulatory networks. As a test case we identified a core OCT4-regulated network that is important for the analysis of stem cell characteristics and cellular differentiation. Functional information is largely enriched using different experimental results. The de novo motif discovery identified well-known regulators closely connected to the OCT4 network as well as potential new regulators of pluripotency and differentiation. These results provide the basis for further targeted functional studies. link: http://identifiers.org/pubmed/19604364

Chay1997_CalciumConcentration: BIOMD0000000378v0.0.1

This a model from the article: Effects of extracellular calcium on electrical bursting and intracellular and luminal…

Details

The extracellular calcium concentration has interesting effects on bursting of pancreatic beta-cells. The mechanism underlying the extracellular Ca2+ effect is not well understood. By incorporating a low-threshold transient inward current to the store-operated bursting model of Chay, this paper elucidates the role of the extracellular Ca2+ concentration in influencing electrical activity, intracellular Ca2+ concentration, and the luminal Ca2+ concentration in the intracellular Ca2+ store. The possibility that this inward current is a carbachol-sensitive and TTX-insensitive Na+ current discovered by others is discussed. In addition, this paper explains how these three variables respond when various pharmacological agents are applied to the store-operated model. link: http://identifiers.org/pubmed/9284334

Parameters:

NameDescription
d_infinity = 0.00344187186519272; tau_d = 0.0234265674250627Reaction: d = (d_infinity-d)/tau_d, Rate Law: (d_infinity-d)/tau_d
Cm = 1.0; i_Ca = -24.1248530333721; i_NS = -6.24107017458029; i_fast = -96.6401171990526; i_K_dr = 25.014877991785; i_K_Ca = 46.2079655277309; i_NaL = -35.502438; i_K_ATP = 73.31708Reaction: V_membrane = (-(i_K_dr+i_K_Ca+i_K_ATP+i_fast+i_Ca+i_NS+i_NaL))/Cm, Rate Law: (-(i_K_dr+i_K_Ca+i_K_ATP+i_fast+i_Ca+i_NS+i_NaL))/Cm
tau_n = 0.0313553515963197; n_infinity = 0.189546217642834Reaction: n = (n_infinity-n)/tau_n, Rate Law: (n_infinity-n)/tau_n
tau_h = 0.0320623804770684; h_infinity = 0.201042499324815Reaction: h = (h_infinity-h)/tau_h, Rate Law: (h_infinity-h)/tau_h
k_rel = 0.2; k_pump = 30.0Reaction: Ca_lum = (-k_rel)*(Ca_lum-Ca_i_cytosolic_calcium)+k_pump*Ca_i_cytosolic_calcium, Rate Law: (-k_rel)*(Ca_lum-Ca_i_cytosolic_calcium)+k_pump*Ca_i_cytosolic_calcium
k_Ca = 7.0; i_Ca = -24.1248530333721; k_rel = 0.2; k_pump = 30.0; omega = 0.2Reaction: Ca_i_cytosolic_calcium = k_rel*(Ca_lum-Ca_i_cytosolic_calcium)-(omega*i_Ca+k_Ca*Ca_i_cytosolic_calcium+k_pump*Ca_i_cytosolic_calcium), Rate Law: k_rel*(Ca_lum-Ca_i_cytosolic_calcium)-(omega*i_Ca+k_Ca*Ca_i_cytosolic_calcium+k_pump*Ca_i_cytosolic_calcium)

States:

NameDescription
d[gated channel activity]
Ca i cytosolic calcium[calcium(2+)]
Ca lum[calcium(2+)]
V membrane[membrane potential]
h[gated channel activity]
n[delayed rectifier potassium channel activity]

Chen2000_CellCycle: BIOMD0000000675v0.0.1

This a model from the article: Kinetic analysis of a molecular model of the budding yeast cell cycle. Chen KC, Csika…

Details

The molecular machinery of cell cycle control is known in more detail for budding yeast, Saccharomyces cerevisiae, than for any other eukaryotic organism. In recent years, many elegant experiments on budding yeast have dissected the roles of cyclin molecules (Cln1-3 and Clb1-6) in coordinating the events of DNA synthesis, bud emergence, spindle formation, nuclear division, and cell separation. These experimental clues suggest a mechanism for the principal molecular interactions controlling cyclin synthesis and degradation. Using standard techniques of biochemical kinetics, we convert the mechanism into a set of differential equations, which describe the time courses of three major classes of cyclin-dependent kinase activities. Model in hand, we examine the molecular events controlling "Start" (the commitment step to a new round of chromosome replication, bud formation, and mitosis) and "Finish" (the transition from metaphase to anaphase, when sister chromatids are pulled apart and the bud separates from the mother cell) in wild-type cells and 50 mutants. The model accounts for many details of the physiology, biochemistry, and genetics of cell cycle control in budding yeast. link: http://identifiers.org/pubmed/10637314

Parameters:

NameDescription
Vd_b2 = 2.023494; kd1_c1 = 0.01; kas_b2 = 50.0; Jd2_c1 = 0.05; kdi_b2 = 0.05; Vd2_c1 = 0.0306448922911362Reaction: Clb2_Sic1 = kas_b2*Clb2*Sic1-Clb2_Sic1*(kdi_b2+Vd_b2+kd1_c1+Vd2_c1/(Jd2_c1+Sic1_T)), Rate Law: kas_b2*Clb2*Sic1-Clb2_Sic1*(kdi_b2+Vd_b2+kd1_c1+Vd2_c1/(Jd2_c1+Sic1_T))
ka_t1_ = 2.0; Vi_t1 = 0.118613853471055; ka_t1 = 0.04; Ji_t1 = 0.05; Ja_t1 = 0.05Reaction: Hct1 = (ka_t1+ka_t1_*Cdc20)*(Hct1_T-Hct1)/((Ja_t1+Hct1_T)-Hct1)-Vi_t1*Hct1/(Ji_t1+Hct1), Rate Law: (ka_t1+ka_t1_*Cdc20)*(Hct1_T-Hct1)/((Ja_t1+Hct1_T)-Hct1)-Vi_t1*Hct1/(Ji_t1+Hct1)
mass = 0.6608Reaction: Bck2 = Bck2_0*mass, Rate Law: missing
ki_sbf_ = 6.0; Va_sbf = 0.310772953639202; ki_sbf = 0.5; Ji_sbf = 0.01; Ja_sbf = 0.01Reaction: SBF = 2*Va_sbf*Ji_sbf/(((ki_sbf+ki_sbf_*Clb2+Va_sbf*Ji_sbf+(ki_sbf+ki_sbf_*Clb2)*Ja_sbf)-Va_sbf)+(((ki_sbf+ki_sbf_*Clb2+Va_sbf*Ji_sbf+(ki_sbf+ki_sbf_*Clb2)*Ja_sbf)-Va_sbf)^2-4*Va_sbf*Ji_sbf*((ki_sbf+ki_sbf_*Clb2)-Va_sbf))^(1/2)), Rate Law: missing
Ji_mcm = 1.0; ka_mcm = 1.0; Ja_mcm = 1.0; ki_mcm = 0.15Reaction: Mcm1 = 2*ka_mcm*Clb2*Ji_mcm/(((ki_mcm+ka_mcm*Clb2*Ji_mcm+ki_mcm*Ja_mcm)-ka_mcm*Clb2)+(((ki_mcm+ka_mcm*Clb2*Ji_mcm+ki_mcm*Ja_mcm)-ka_mcm*Clb2)^2-4*(ki_mcm-ka_mcm*Clb2)*ka_mcm*Clb2*Ji_mcm)^(1/2)), Rate Law: missing
kd_n2 = 0.1; ks_n2_ = 0.05; ks_n2 = 0.0; mass = 0.6608Reaction: Cln2 = mass*(ks_n2+ks_n2_*SBF)-kd_n2*Cln2, Rate Law: mass*(ks_n2+ks_n2_*SBF)-kd_n2*Cln2
Vd_b5 = 0.2712; kd1_c1 = 0.01; kas_b5 = 50.0; Jd2_c1 = 0.05; kdi_b5 = 0.05; Vd2_c1 = 0.0306448922911362Reaction: Clb5_Sic1 = kas_b5*Clb5*Sic1-Clb5_Sic1*(kdi_b5+Vd_b5+kd1_c1+Vd2_c1/(Jd2_c1+Sic1_T)), Rate Law: kas_b5*Clb5*Sic1-Clb5_Sic1*(kdi_b5+Vd_b5+kd1_c1+Vd2_c1/(Jd2_c1+Sic1_T))
kd1_c1 = 0.01; ks_c1 = 0.02; Jd2_c1 = 0.05; ks_c1_ = 0.1; Vd2_c1 = 0.0306448922911362Reaction: Sic1_T = (ks_c1+ks_c1_*Swi5)-Sic1_T*(kd1_c1+Vd2_c1/(Jd2_c1+Sic1_T)), Rate Law: (ks_c1+ks_c1_*Swi5)-Sic1_T*(kd1_c1+Vd2_c1/(Jd2_c1+Sic1_T))
Vd_b2 = 2.023494; ks_b2 = 0.002; ks_b2_ = 0.05; mass = 0.6608Reaction: Clb2_T = mass*(ks_b2+ks_b2_*Mcm1)-Vd_b2*Clb2_T, Rate Law: mass*(ks_b2+ks_b2_*Mcm1)-Vd_b2*Clb2_T
kd_20 = 0.08; ks_20_ = 0.06; ks_20 = 0.005Reaction: Cdc20_T = (ks_20+ks_20_*Clb2)-kd_20*Cdc20_T, Rate Law: (ks_20+ks_20_*Clb2)-kd_20*Cdc20_T
Jn3 = 6.0; Dn3 = 1.0; mass = 0.6608Reaction: Cln3 = Cln3_max*Dn3*mass/(Jn3+Dn3*mass), Rate Law: missing
ka_swi = 1.0; Ja_swi = 0.1; ki_swi_ = 0.2; Ji_swi = 0.1; ki_swi = 0.3Reaction: Swi5 = 2*ka_swi*Cdc20*Ji_swi/(((ki_swi+ki_swi_*Clb2+ka_swi*Cdc20*Ji_swi+(ki_swi+ki_swi_*Clb2)*Ja_swi)-ka_swi*Cdc20)+(((ki_swi+ki_swi_*Clb2+ka_swi*Cdc20*Ji_swi+(ki_swi+ki_swi_*Clb2)*Ja_swi)-ka_swi*Cdc20)^2-4*((ki_swi+ki_swi_*Clb2)-ka_swi*Cdc20)*ka_swi*Cdc20*Ji_swi)^(1/2)), Rate Law: missing
kd_20 = 0.08; ka_20 = 1.0; Vi_20 = 0.1Reaction: Cdc20 = ka_20*(Cdc20_T-Cdc20)-Cdc20*(Vi_20+kd_20), Rate Law: ka_20*(Cdc20_T-Cdc20)-Cdc20*(Vi_20+kd_20)
ks_b5 = 0.006; ks_b5_ = 0.02; Vd_b5 = 0.2712; mass = 0.6608Reaction: Clb5_T = mass*(ks_b5+ks_b5_*MBF)-Vd_b5*Clb5_T, Rate Law: mass*(ks_b5+ks_b5_*MBF)-Vd_b5*Clb5_T

States:

NameDescription
Clb2 T[G2/mitotic-specific cyclin-2]
Sic1 T[Protein SIC1]
Cln2[G1/S-specific cyclin CLN2]
Cln3[G1/S-specific cyclin CLN3]
Hct1[APC/C activator protein CDH1]
Clb5 T[S-phase entry cyclin-5]
Clb5[S-phase entry cyclin-5]
Mcm1[Pheromone receptor transcription factor]
Swi5[Transcriptional factor SWI5]
Bck2[Protein BCK2]
Sic1[Protein SIC1]
SBF[DNA-binding protein RAP1; SBF transcription complex]
Clb2 Sic1[Protein SIC1; G2/mitotic-specific cyclin-2]
Clb5 Sic1[S-phase entry cyclin-5; Protein SIC1]
Cdc20[APC/C activator protein CDC20]
Clb2[G2/mitotic-specific cyclin-2]
Cdc20 T[APC/C activator protein CDC20]
MBF[Multiprotein-bridging factor 1]

Chen2004 - Cell Cycle Regulation: BIOMD0000000056v0.0.1

Chen2004 - Cell Cycle RegulationThis is a hypothetical model of cell cycle that describes the molecular mechanism for re…

Details

The adaptive responses of a living cell to internal and external signals are controlled by networks of proteins whose interactions are so complex that the functional integration of the network cannot be comprehended by intuitive reasoning alone. Mathematical modeling, based on biochemical rate equations, provides a rigorous and reliable tool for unraveling the complexities of molecular regulatory networks. The budding yeast cell cycle is a challenging test case for this approach, because the control system is known in exquisite detail and its function is constrained by the phenotypic properties of >100 genetically engineered strains. We show that a mathematical model built on a consensus picture of this control system is largely successful in explaining the phenotypes of mutants described so far. A few inconsistencies between the model and experiments indicate aspects of the mechanism that require revision. In addition, the model allows one to frame and critique hypotheses about how the division cycle is regulated in wild-type and mutant cells, to predict the phenotypes of new mutant combinations, and to estimate the effective values of biochemical rate constants that are difficult to measure directly in vivo. link: http://identifiers.org/pubmed/15169868

Parameters:

NameDescription
IET = 1.0Reaction: IE = IET-IEP, Rate Law: missing
Jatem = 0.1Reaction: TEM1GDP => TEM1GTP; LTE1, Rate Law: 1*TEM1GDP*LTE1/(Jatem+TEM1GDP)
Vppc1 = NaNReaction: C5P => C5, Rate Law: Vppc1*C5P
kd14 = 0.1Reaction: RENT => NET1, Rate Law: kd14*RENT
kd3f6 = 1.0Reaction: F2P => CLB2, Rate Law: kd3f6*F2P
Vdb2 = NaNReaction: F2 => CDC6, Rate Law: Vdb2*F2
Jacdh = 0.03; Vacdh = NaNReaction: CDH1i => CDH1, Rate Law: 1*CDH1i*Vacdh/(Jacdh+CDH1i)
Vicdh = NaN; Jicdh = 0.03Reaction: CDH1 => CDH1i, Rate Law: 1*CDH1*Vicdh/(Jicdh+CDH1)
kasrentp = 1.0Reaction: CDC14 + NET1P => RENTP, Rate Law: kasrentp*CDC14*NET1P
ksb5_p_p = 0.005; ksb5_p = 8.0E-4Reaction: => CLB5; SBF, MASS, Rate Law: (ksb5_p+ksb5_p_p*SBF)*MASS
kd3c1 = 1.0Reaction: C5P => CLB5, Rate Law: kd3c1*C5P
kasb5 = 50.0Reaction: CLB5 + SIC1 => C5, Rate Law: kasb5*CLB5*SIC1
kdswi = 0.08Reaction: SWI5 =>, Rate Law: kdswi*SWI5
kdif5 = 0.01Reaction: F5 => CLB5 + CDC6, Rate Law: kdif5*F5
kdif2 = 0.5Reaction: F2 => CLB2 + CDC6, Rate Law: kdif2*F2
Vppnet = NaNReaction: NET1P => NET1, Rate Law: Vppnet*NET1P
kdib2 = 0.05Reaction: C2 => CLB2 + SIC1, Rate Law: kdib2*C2
ksspn = 0.1; Jspn = 0.14Reaction: => SPN; CLB2, Rate Law: ksspn*CLB2/(Jspn+CLB2)
kasb2 = 50.0Reaction: CLB2 + SIC1 => C2, Rate Law: kasb2*CLB2*SIC1
ksswi_p_p = 0.08; ksswi_p = 0.005Reaction: => SWI5; MCM1, Rate Law: ksswi_p+ksswi_p_p*MCM1
ks14 = 0.2Reaction: => CDC14, Rate Law: ks14
kdspn = 0.06Reaction: SPN =>, Rate Law: kdspn*SPN
b0 = 0.054Reaction: BCK2 = b0*MASS, Rate Law: missing
Vdppx = NaNReaction: PPX =>, Rate Law: Vdppx*PPX
kiswi = 0.05Reaction: SWI5 => SWI5P; CLB2, Rate Law: kiswi*CLB2*SWI5
kdib5 = 0.06Reaction: C5 => CLB5 + SIC1, Rate Law: kdib5*C5
ksc1_p = 0.012; ksc1_p_p = 0.12Reaction: => SIC1; SWI5, Rate Law: ksc1_p+ksc1_p_p*SWI5
Vdpds = NaNReaction: PDS1 =>, Rate Law: Vdpds*PDS1
ks2pds_p_p = 0.055; ks1pds_p_p = 0.03; kspds_p = 0.0Reaction: => PDS1; SBF, MCM1, Rate Law: kspds_p+ks1pds_p_p*SBF+ks2pds_p_p*MCM1
Vppf6 = NaNReaction: F5P => F5, Rate Law: Vppf6*F5P
Vkpnet = NaNReaction: NET1 => NET1P, Rate Law: Vkpnet*NET1
ks20_p = 0.006; ks20_p_p = 0.6Reaction: => CDC20i; MCM1, Rate Law: ks20_p+ks20_p_p*MCM1
Vdb5 = NaNReaction: F5P => CDC6P, Rate Law: Vdb5*F5P
kdiesp = 0.5Reaction: PE => PDS1 + ESP1, Rate Law: kdiesp*PE
Vkpc1 = NaNReaction: C5 => C5P, Rate Law: Vkpc1*C5
ka20_p = 0.05; ka20_p_p = 0.2Reaction: CDC20i => CDC20; IEP, Rate Law: (ka20_p+ka20_p_p*IEP)*CDC20i
kdirentp = 2.0Reaction: RENTP => CDC14 + NET1P, Rate Law: kdirentp*RENTP
Vkpf6 = NaNReaction: F5 => F5P, Rate Law: Vkpf6*F5
ki15 = 0.5Reaction: CDC15 => CDC15i, Rate Law: ki15*CDC15
ksf6_p = 0.024; ksf6_p_p_p = 0.004; ksf6_p_p = 0.12Reaction: => CDC6; SWI5, SBF, Rate Law: ksf6_p+ksf6_p_p*SWI5+ksf6_p_p_p*SBF
ksb2_p_p = 0.04; ksb2_p = 0.001Reaction: => CLB2; MCM1, MASS, Rate Law: (ksb2_p+ksb2_p_p*MCM1)*MASS
kdbud = 0.06Reaction: BUD =>, Rate Law: kdbud*BUD
kasf2 = 15.0Reaction: CLB2 + CDC6 => F2, Rate Law: kasf2*CLB2*CDC6
Vaiep = NaN; Jaiep = 0.1Reaction: IE => IEP, Rate Law: 1*IE*Vaiep/(Jaiep+IE)
kd20 = 0.3Reaction: CDC20 =>, Rate Law: kd20*CDC20
kscdh = 0.01Reaction: => CDH1, Rate Law: kscdh
kasf5 = 0.01Reaction: CLB5 + CDC6 => F5, Rate Law: kasf5*CLB5*CDC6
kdcdh = 0.01Reaction: CDH1i =>, Rate Law: kdcdh*CDH1i
Jitem = 0.1Reaction: TEM1GTP => TEM1GDP; BUB2, Rate Law: 1*TEM1GTP*BUB2/(Jitem+TEM1GTP)
kdnet = 0.03Reaction: NET1 =>, Rate Law: kdnet*NET1
kaswi = 2.0Reaction: SWI5P => SWI5; CDC14, Rate Law: kaswi*CDC14*SWI5P
ksnet = 0.084Reaction: => NET1, Rate Law: ksnet
k=1.0Reaction: CDC20 => CDC20i; MAD2, Rate Law: k*MAD2*CDC20
ka15p = 0.001; ka15_p = 0.002; ka15_p_p = 1.0Reaction: CDC15i => CDC15; TEM1GDP, TEM1GTP, CDC14, Rate Law: (ka15_p*TEM1GDP+ka15_p_p*TEM1GTP+ka15p*CDC14)*CDC15i
TEM1T = 1.0Reaction: TEM1GDP = TEM1T-TEM1GTP, Rate Law: missing
Jiiep = 0.1; kiiep = 0.15Reaction: IEP => IE, Rate Law: kiiep*IEP*1/(Jiiep+IEP)

States:

NameDescription
CDC15i[Cell division control protein 15]
TEM1GTP[Protein TEM1]
BCK2[Protein BCK2]
SPN[CCO:P0000392]
CDH1[APC/C activator protein CDH1]
CDH1i[APC/C activator protein CDH1]
CDC15[Cell division control protein 15]
CKIT[Protein SIC1; Cell division control protein 6]
F5P[Cell division control protein 6; S-phase entry cyclin-5; S-phase entry cyclin-6]
NET1P[Nucleolar protein NET1]
CDC20i[APC/C activator protein CDC20]
C5P[Protein SIC1; S-phase entry cyclin-6; S-phase entry cyclin-5]
CLB2[G2/mitotic-specific cyclin-2; G2/mitotic-specific cyclin-1]
CDC20[APC/C activator protein CDC20]
C2[Protein SIC1; G2/mitotic-specific cyclin-2; G2/mitotic-specific cyclin-1]
CDC6[Cell division control protein 6]
CDC6T[Cell division control protein 6]
IEP[anaphase-promoting complex]
C5[Protein SIC1; S-phase entry cyclin-6; S-phase entry cyclin-5]
SWI5[Transcriptional factor SWI5]
F5[S-phase entry cyclin-6; S-phase entry cyclin-5; Cell division control protein 6]
TEM1GDP[Protein TEM1]
CLB2T[G2/mitotic-specific cyclin-2; G2/mitotic-specific cyclin-1]
CDC6P[Cell division control protein 6]
CLB5T[S-phase entry cyclin-6; S-phase entry cyclin-5]
SIC1P[Protein SIC1]
PDS1[Securin]
SIC1T[Protein SIC1]
IE[anaphase-promoting complex]
CDC14[Tyrosine-protein phosphatase CDC14]
BUD[CCO:C0000485]
PPX[Exopolyphosphatase]
RENTP[RENT complex; NAD-dependent histone deacetylase SIR2; Nucleolar protein NET1; Tyrosine-protein phosphatase CDC14]
SIC1[Protein SIC1]
CLB5[S-phase entry cyclin-6; S-phase entry cyclin-5]
SWI5P[Transcriptional factor SWI5]
NET1[Nucleolar protein NET1]
F2[Cell division control protein 6; G2/mitotic-specific cyclin-1; G2/mitotic-specific cyclin-2]

Chen2006 - Nitric Oxide Release from Endothelial Cells: BIOMD0000000676v0.0.1

Chen2006 - Nitric Oxide Release from Endothelial CellsThis model is described in the article: [Theoretical analysis of…

Details

Vascular endothelium expressing endothelial nitric oxide synthase (eNOS) produces nitric oxide (NO), which has a number of important physiological functions in the microvasculature. The rate of NO production by the endothelium is a critical determinant of NO distribution in the vascular wall. We have analyzed the biochemical pathways of NO synthesis and formulated a model to estimate NO production by the microvascular endothelium under physiological conditions. The model quantifies the NO produced by eNOS based on the kinetics of NO synthesis and the availability of eNOS and its intracellular substrates. The predicted NO production from microvessels was in the range of 0.005-0.1 microM/s. This range of predicted values is in agreement with some experimental values but is much lower than other rates previously measured or estimated from experimental data with the help of mathematical modeling. Paradoxical discrepancies between the model predictions and previously reported results based on experimental measurements of NO concentration in the vicinity of the arteriolar wall suggest that NO can also be released through eNOS-independent mechanisms, such as catalysis by neuronal NOS (nNOS). We also used our model to test the sensitivity of NO production to substrate availability, eNOS concentration, and potential rate-limiting factors. The results indicated that the predicted low level of NO production can be attributed primarily to a low expression of eNOS in the microvascular endothelial cells. link: http://identifiers.org/pubmed/16864000

Parameters:

NameDescription
k10_prime = 89.9Reaction: Fe3__O2__NOHA => Fe2__NOHA, Rate Law: Endothelium*k10_prime*Fe3__O2__NOHA
k14 = 53.9Reaction: Fe3__NO => Fe3__enos + NO, Rate Law: Endothelium*k14*Fe3__NO
S = 0.0Reaction: => Arg, Rate Law: Endothelium*S
k11 = 29.4Reaction: Fe3__O2__NOHA => Fe3__NO + Citrulline, Rate Law: Endothelium*k11*Fe3__O2__NOHA
k2 = 0.91Reaction: Fe3__enos => Fe2, Rate Law: Endothelium*k2*Fe3__enos
k8_prime = 0.1; k8 = 0.1Reaction: Fe3__NOHA => Fe3__enos + NOHA, Rate Law: Endothelium*(k8*Fe3__NOHA-k8_prime*Fe3__enos*NOHA)
k5_prime = 98.0Reaction: Fe3__O2__Arg => Fe2__Arg, Rate Law: Endothelium*k5_prime*Fe3__O2__Arg
k9_prime = 1.89; k9 = 11.4Reaction: Fe2__NOHA => Fe2 + NOHA, Rate Law: Endothelium*(k9*Fe2__NOHA-k9_prime*Fe2*NOHA)
k1 = 0.1; k1_prime = 0.1Reaction: Arg + Fe3__enos => Fe3__Arg, Rate Law: Endothelium*(k1*Arg*Fe3__enos-k1_prime*Fe3__Arg)
k10 = 3.33Reaction: Fe2__NOHA => Fe3__O2__NOHA; O2, Rate Law: Endothelium*k10*O2*Fe2__NOHA
k6 = 12.6Reaction: Fe3__O2__Arg => Fe3__NOHA, Rate Law: Endothelium*k6*Fe3__O2__Arg
k7 = 0.91Reaction: Fe3__NOHA => Fe2__NOHA, Rate Law: Endothelium*k7*Fe3__NOHA
k3 = 0.91Reaction: Fe3__Arg => Fe2__Arg, Rate Law: Endothelium*k3*Fe3__Arg
k12 = 0.91Reaction: Fe3__NO => Fe2__NO, Rate Law: Endothelium*k12*Fe3__NO
k13 = 0.033Reaction: Fe2__NO => Fe3__enos; O2, Rate Law: Endothelium*k13*O2*Fe2__NO
k5 = 2.58Reaction: Fe2__Arg => Fe3__O2__Arg; O2, Rate Law: Endothelium*k5*O2*Fe2__Arg
k4_prime = 11.4; k4 = 1.89Reaction: Arg + Fe2 => Fe2__Arg, Rate Law: Endothelium*(k4*Arg*Fe2-k4_prime*Fe2__Arg)

States:

NameDescription
Fe2 NO[iron(2+); nitric oxide]
NO[nitric oxide]
Fe3 enos[iron(3+)]
Citrulline[citrulline]
Fe2 Arg[arginine; iron(2+)]
Fe2 NOHA[iron(2+); hydroxyarginine]
NOHA[hydroxyarginine]
Fe3 O2 Arg[dioxygen; arginine; iron(3+)]
Fe2[iron(2+)]
Fe3 NO[iron(3+); nitric oxide]
Fe3 O2 NOHA[dioxygen; iron(3+); hydroxyarginine]
Fe3 Arg[iron(3+); arginine]
Arg[arginine]
Fe3 NOHA[iron(3+); hydroxyarginine]

Chen2007_NeuronalEndothelialNOS: MODEL0491251823v0.0.1

This a model from the article: Vascular and perivascular nitric oxide release and transport: biochemical pathways of n…

Details

Nitric oxide (NO) derived from nitric oxide synthase (NOS) is an important paracrine effector that maintains vascular tone. The release of NO mediated by NOS isozymes under various O(2) conditions critically determines the NO bioavailability in tissues. Because of experimental difficulties, there has been no direct information on how enzymatic NO production and distribution change around arterioles under various oxygen conditions. In this study, we used computational models based on the analysis of biochemical pathways of enzymatic NO synthesis and the availability of NOS isozymes to quantify the NO production by neuronal NOS (NOS1) and endothelial NOS (NOS3). We compared the catalytic activities of NOS1 and NOS3 and their sensitivities to the concentration of substrate O(2). Based on the NO release rates predicted from kinetic models, the geometric distribution of NO sources, and mass balance analysis, we predicted the NO concentration profiles around an arteriole under various O(2) conditions. The results indicated that NOS1-catalyzed NO production was significantly more sensitive to ambient O(2) concentration than that catalyzed by NOS3. Also, the high sensitivity of NOS1 catalytic activity to O(2) was associated with significantly reduced NO production and therefore NO concentrations, upon hypoxia. Moreover, the major source determining the distribution of NO was NOS1, which was abundantly expressed in the nerve fibers and mast cells close to arterioles, rather than NOS3, which was expressed in the endothelium. Finally, the perivascular NO concentration predicted by the models under conditions of normoxia was paradoxically at least an order of magnitude lower than a number of experimental measurements, suggesting a higher abundance of NOS1 or NOS3 and/or the existence of other enzymatic or nonenzymatic sources of NO in the microvasculature. link: http://identifiers.org/pubmed/17320763

Chen2009 - ErbB Signaling: BIOMD0000000255v0.0.1

This is A431 IERMv1.0 model described in the article Input-output behavior of ErbB signaling pathways as revealed by a…

Details

The ErbB signaling pathways, which regulate diverse physiological responses such as cell survival, proliferation and motility, have been subjected to extensive molecular analysis. Nonetheless, it remains poorly understood how different ligands induce different responses and how this is affected by oncogenic mutations. To quantify signal flow through ErbB-activated pathways we have constructed, trained and analyzed a mass action model of immediate-early signaling involving ErbB1-4 receptors (EGFR, HER2/Neu2, ErbB3 and ErbB4), and the MAPK and PI3K/Akt cascades. We find that parameter sensitivity is strongly dependent on the feature (e.g. ERK or Akt activation) or condition (e.g. EGF or heregulin stimulation) under examination and that this context dependence is informative with respect to mechanisms of signal propagation. Modeling predicts log-linear amplification so that significant ERK and Akt activation is observed at ligand concentrations far below the K(d) for receptor binding. However, MAPK and Akt modules isolated from the ErbB model continue to exhibit switch-like responses. Thus, key system-wide features of ErbB signaling arise from nonlinear interaction among signaling elements, the properties of which appear quite different in context and in isolation. link: http://identifiers.org/pubmed/19156131

Parameters:

NameDescription
k8 = 5.91474E-7; kd8 = 0.2Reaction: c14 + c336 => c344, Rate Law: k8*c14*c336-kd8*c344
k60c = 5.2E-4; kd60 = 0.0Reaction: c349 => c86, Rate Law: k60c*c349-kd60*c86
kd123 = 0.177828; k123 = 0.0Reaction: c336 + c105 => c139, Rate Law: k123*c336*c105-kd123*c139
k18 = 2.5E-5; kd18 = 1.3Reaction: c26 + c317 => c320, Rate Law: k18*c26*c317-kd18*c320
k5 = 0.0; kd5b = 0.0080833Reaction: c9 + c320 => c319, Rate Law: k5*c9*c320-kd5b*c319
k37 = 1.5E-6; kd37 = 0.3Reaction: c341 + c40 => c351, Rate Law: k37*c341*c40-kd37*c351
kd36 = 0.0; k36 = 0.005Reaction: c40 => c31, Rate Law: k36*c40-kd36*c31
k17 = 1.67E-5; kd17 = 0.06Reaction: c24 + c314 => c317, Rate Law: k17*c24*c314-kd17*c317
kd122 = 1.0; k122 = 1.8704E-8Reaction: c550 + c105 => c555, Rate Law: k122*c550*c105-kd122*c555
k2b = 3.73632E-8; kd2b = 0.016Reaction: c499 + c141 => c492, Rate Law: k2b*c499*c141-kd2b*c492
kd33 = 0.2; k33 = 3.5E-5Reaction: c40 + c30 => c38, Rate Law: k33*c40*c30-kd33*c38
kd2 = 0.16; k2 = 7.44622E-6Reaction: c3 + c499 => c500, Rate Law: k2*c3*c499-kd2*c500
kd6b = 0.0; k6b = 0.0Reaction: c347 => c349, Rate Law: k6b*c347-kd6b*c349
k6 = 0.013; kd6 = 5.0E-5Reaction: c32 => c63, Rate Law: k6*c32-kd6*c63
k32 = 4.0E-7; kd32 = 0.1Reaction: c293 + c38 => c305, Rate Law: k32*c293*c38-kd32*c305
k60 = 0.00266742; kd60 = 0.0Reaction: c17 => c86, Rate Law: k60*c17-kd60*c86
kd111 = 6.57; k111 = 0.0Reaction: c83 + c490 => c475, Rate Law: k111*c83*c490-kd111*c475
k34 = 7.5E-6; kd34 = 0.03Reaction: c293 + c30 => c317, Rate Law: k34*c293*c30-kd34*c317
k1 = 0.0; kd1 = 0.033Reaction: c1 + c286 => c499, Rate Law: k1*c1*c286-kd1*c499
kd23 = 0.06; k23 = 6.0Reaction: c32 => c33, Rate Law: k23*c32-kd23*c33
kd103 = 0.016; k103 = 8.36983E-9Reaction: c87 + c332 => c336, Rate Law: k103*c87*c332-kd103*c336
k60b = 0.0471248; kd60 = 0.0Reaction: c323 => c86, Rate Law: k60b*c323-kd60*c86
k16 = 1.67E-5; kd24 = 0.55Reaction: c22 + c299 => c302, Rate Law: k16*c22*c299-kd24*c302
kd1d = 0.1; k1d = 518.0Reaction: c117 + c1 => c336, Rate Law: k1d*c117*c1-kd1d*c336
kd97 = 0.015; k97 = 1000000.0Reaction: c531 + c285 => c286, Rate Law: k97*c531*c285-kd97*c286
kd19 = 0.5; k19 = 1.667E-7Reaction: c69 + c317 => c320, Rate Law: k19*c69*c317-kd19*c320
kd35 = 0.0015; k35 = 7.5E-6Reaction: c24 + c22 => c30, Rate Law: k35*c24*c22-kd35*c30
k65 = 0.0; kd65 = 0.2Reaction: c83 + c420 => c98, Rate Law: k65*c83*c420-kd65*c98
k20 = 1.1068E-5; kd20 = 0.4Reaction: c317 + c71 => c323, Rate Law: k20*c317*c71-kd20*c323
k110 = 3.33E-4; kd110 = 0.1Reaction: c83 + c446 => c437, Rate Law: k110*c83*c446-kd110*c437
kd41 = 0.0429; k41 = 5.0E-5Reaction: c30 + c299 => c305, Rate Law: k41*c30*c299-kd41*c305
k21 = 3.67E-7; kd21 = 0.23Reaction: c317 + c26 => c323, Rate Law: k21*c317*c26-kd21*c323
k64 = 1.67E-5; kd64 = 0.3Reaction: c83 + c24 => c102, Rate Law: k64*c83*c24-kd64*c102
k22 = 1.39338E-7; kd22 = 0.1Reaction: c31 + c17 => c63, Rate Law: k22*c31*c17-kd22*c63
kd123h = 0.1; k123h = 0.0Reaction: c5 + c105 => c556, Rate Law: k123h*c5*c105-kd123h*c556
k22 = 1.39338E-7; kd22b = 0.1Reaction: c31 + c341 => c347, Rate Law: k22*c31*c341-kd22b*c347
kd63 = 0.275; k16 = 1.67E-5Reaction: c293 + c22 => c314, Rate Law: k16*c293*c22-kd63*c314
kd7 = 1.38E-4; k7 = 5.0E-5Reaction: c336 => c338, Rate Law: k7*c336-kd7*c338
kd25 = 0.0214; k25 = 1.67E-5Reaction: c24 + c302 => c305, Rate Law: k25*c24*c302-kd25*c305

States:

NameDescription
c349(ErbB3:ErbB2)_P:GAP:Shc
c286[Epidermal growth factor receptor]
c172(EGF:ErbB1)_P:GAP
c332(EGF:ErbB1)_P:GAP:(Shc_P)
c499[Pro-epidermal growth factor; Epidermal growth factor receptor]
c3022(ErbB2)_P:GAP:(Shc_P):Grb2
c83(ERK_PP)_i
c105ATP 1.2e9
c125[Pro-epidermal growth factor; Epidermal growth factor receptor]
c336[Receptor tyrosine-protein kinase erbB-4; Receptor tyrosine-protein kinase erbB-2]
c3142(ErbB2)_P:GAP:Grb2
c3112(ErbB2)_P:GAP:(Shc_P):Grb2:Sos:(Ras:GTP)
c31[SHC-transforming protein 2; 605217]
c347(ErbB3:ErbB2)_P:GAP:Shc
c353(ErbB3:ErbB2)_P:GAP:(Shc_P)
c351(ErbB3:ErbB2)_P:GAP:(Shc_P)
c117[Receptor tyrosine-protein kinase erbB-2; Receptor tyrosine-protein kinase erbB-4]
c344(ErbB4:ErbB2)_P:GAP
c341(ErbB3:ErbB2)_P:GAP
c642(EGF:ErbB1)_P:GAP:(Shc_P)
c3202(ErbB2)_P:GAP:Grb2:Sos:(Ras:GDP)
c348(ErbB4:ErbB2)_P:GAP:Shc
c40(Shc_P)
c3052(ErbB2)_P:GAP:(Shc_P):Grb2:Sos
c3082(ErbB2)_P:GAP:(Shc_P):Grb2:Sos:(Ras:GDP)
c322(EGF:ErbB1)_P:GAP:Shc
c3232(ErbB2)_P:GAP:Grb2:Sos:(Ras:GTP)
c3172(ErbB2)_P:GAP:Grb2:Sos
c632(EGF:ErbB1)_P:GAP:Shc
c343(ErbB3:ErbB2)_P:GAP
c346(ErbB4:ErbB2)_P:GAP
c22[Growth factor receptor-bound protein 2; 108355]
c123[Pro-epidermal growth factor; Epidermal growth factor receptor]
c124[Pro-epidermal growth factor; Epidermal growth factor receptor]
c350(ErbB4:ErbB2)_P:GAP:Shc

Chen2011/1 - bone marrow invasion absolute model: BIOMD0000000793v0.0.1

The paper describes a model of tumor invasion to bone marrow. Created by COPASI 4.26 (Build 213) This model is des…

Details

The invasion of a new species into an established ecosystem can be directly compared to the steps involved in cancer metastasis. Cancer must grow in a primary site, extravasate and survive in the circulation to then intravasate into target organ (invasive species survival in transport). Cancer cells often lay dormant at their metastatic site for a long period of time (lag period for invasive species) before proliferating (invasive spread). Proliferation in the new site has an impact on the target organ microenvironment (ecological impact) and eventually the human host (biosphere impact).Tilman has described mathematical equations for the competition between invasive species in a structured habitat. These equations were adapted to study the invasion of cancer cells into the bone marrow microenvironment as a structured habitat. A large proportion of solid tumor metastases are bone metastases, known to usurp hematopoietic stem cells (HSC) homing pathways to establish footholds in the bone marrow. This required accounting for the fact that this is the natural home of hematopoietic stem cells and that they already occupy this structured space. The adapted Tilman model of invasion dynamics is especially valuable for modeling the lag period or dormancy of cancer cells.The Tilman equations for modeling the invasion of two species into a defined space have been modified to study the invasion of cancer cells into the bone marrow microenvironment. These modified equations allow a more flexible way to model the space competition between the two cell species. The ability to model initial density, metastatic seeding into the bone marrow and growth once the cells are present, and movement of cells out of the bone marrow niche and apoptosis of cells are all aspects of the adapted equations. These equations are currently being applied to clinical data sets for verification and further refinement of the models. link: http://identifiers.org/pubmed/21967667

Parameters:

NameDescription
b1 = 0.2 1Reaction: => H, Rate Law: bone_marrow*b1*H*(1-H)
u1 = 0.1 1Reaction: H =>, Rate Law: bone_marrow*u1*H
u2 = 0.1 1Reaction: T =>, Rate Law: bone_marrow*u2*T
b2 = 0.8 1Reaction: => T; H, Rate Law: bone_marrow*b2*T*((1-T)-H)

States:

NameDescription
T[malignant cell]
H[hematopoietic stem cell]

Chen2011/2 - bone marrow invasion relative model: BIOMD0000000795v0.0.1

The paper describes a model of tumor invasion to bone marrow. Created by COPASI 4.26 (Build 213) This model is des…

Details

The invasion of a new species into an established ecosystem can be directly compared to the steps involved in cancer metastasis. Cancer must grow in a primary site, extravasate and survive in the circulation to then intravasate into target organ (invasive species survival in transport). Cancer cells often lay dormant at their metastatic site for a long period of time (lag period for invasive species) before proliferating (invasive spread). Proliferation in the new site has an impact on the target organ microenvironment (ecological impact) and eventually the human host (biosphere impact).Tilman has described mathematical equations for the competition between invasive species in a structured habitat. These equations were adapted to study the invasion of cancer cells into the bone marrow microenvironment as a structured habitat. A large proportion of solid tumor metastases are bone metastases, known to usurp hematopoietic stem cells (HSC) homing pathways to establish footholds in the bone marrow. This required accounting for the fact that this is the natural home of hematopoietic stem cells and that they already occupy this structured space. The adapted Tilman model of invasion dynamics is especially valuable for modeling the lag period or dormancy of cancer cells.The Tilman equations for modeling the invasion of two species into a defined space have been modified to study the invasion of cancer cells into the bone marrow microenvironment. These modified equations allow a more flexible way to model the space competition between the two cell species. The ability to model initial density, metastatic seeding into the bone marrow and growth once the cells are present, and movement of cells out of the bone marrow niche and apoptosis of cells are all aspects of the adapted equations. These equations are currently being applied to clinical data sets for verification and further refinement of the models. link: http://identifiers.org/pubmed/21967667

Parameters:

NameDescription
u1 = 0.1 1Reaction: T =>, Rate Law: bone_marrow*u1*T
v = 0.1 1; b2 = 0.8 1Reaction: => H; T, Rate Law: bone_marrow*b2*H*((1-H)-(1-v)*T)
u2 = 0.1 1Reaction: H =>, Rate Law: bone_marrow*u2*H
k = 0.9 1; b1 = 0.2 1Reaction: H => ; T, Rate Law: bone_marrow*b1*H*T*k

States:

NameDescription
T[malignant cell]
H[hematopoietic stem cell]

ChenXF2008_CICR: BIOMD0000000202v0.0.1

The model reproduces the plots in Figures 1 and 2. Note that the units of the time scale "A" are not right in the paper,…

Details

A mathematical model is proposed to illustrate the activation of STIM1 (stromal interaction molecule 1) protein, the assembly and activation of calcium-release activated calcium (CRAC) channels in T cells. In combination with De Young-Keizer-Li-Rinzel model, we successfully reproduce a sustained Ca(2+) oscillation in cytoplasm. Our results reveal that Ca(2+) oscillation dynamics in cytoplasm can be significantly affected by the way how the Orai1 CRAC channel are assembled and activated. A low sustained Ca(2+) influx is observed through the CRAC channels across the plasma membrane. In particular, our model shows that a tetrameric channel complex can effectively regulate the total quantity of the channels and the ratio of the active channels to the total channels, and a period of Ca(2+) oscillation about 29 s is in agreement with published experimental data. The bifurcation analyses illustrate the different dynamic properties between our mixed Ca(2+) feedback model and the single positive or negative feedback models. link: http://identifiers.org/pubmed/18538916

Parameters:

NameDescription
k_a = 4.0 s_1Reaction: => S2a; S2, Rate Law: ER*k_a*S2
kod = 1.0 s_1Reaction: O_o => Oc, Rate Law: PM*kod*O_o
k_i = 6.0 s_1Reaction: S2a =>, Rate Law: ER*k_i*S2a
Ca_ec = 1500.0 uM; k_soc = 2.3 uM_1_s_1; V_PMleak = 5.0E-7 s_1Reaction: => Ca_Cyt; O_o, Rate Law: Cytoplasm*(k_soc*O_o+V_PMleak)*(Ca_ec-Ca_Cyt)
kdo = 0.6 s_1Reaction: O_o =>, Rate Law: PM*kdo*O_o
kop = 0.5 s_1; l_hill = 1.0 dimensionless; Ko = 0.2 uMReaction: Oc => O_o; S2a, Rate Law: PM*kop*S2a^l_hill*Oc/(Ko^l_hill+S2a^l_hill)
K1 = 5.0 uM; St = 0.6 uMReaction: S2 = K1^2/(Ca_ER^2+K1^2)*(St-S2a), Rate Law: missing
kdc = 0.5 s_1Reaction: Oc =>, Rate Law: PM*kdc*Oc
q = 2.0 dimensionless; V_PMCA = 1.0 uM_s_1; K_PMCA = 0.45 uMReaction: Ca_Cyt =>, Rate Law: Cytoplasm*V_PMCA*Ca_Cyt^q/(K_PMCA^q+Ca_Cyt^q)
Vs4 = 0.25 uM_s_1; K2 = 0.14 uMReaction: => S4; S2, Rate Law: ER*Vs4*S2^2/(S2^2+K2^2)
kd_oligo = 0.8 s_1Reaction: S4 =>, Rate Law: ER*kd_oligo*S4
K_PLC = 0.12 uM; V_PLC = 0.5 uM_s_1Reaction: => IP3_Cyt; Ca_Cyt, Rate Law: Cytoplasm*V_PLC*Ca_Cyt^2/(K_PLC^2+Ca_Cyt^2)
K_SERCA = 0.15 uM; V_SERCA = 1.0 uM_s_1; p = 2.0 dimensionlessReaction: Ca_Cyt => Ca_ER, Rate Law: Cytoplasm*V_SERCA*Ca_Cyt^p/(K_SERCA^p+Ca_Cyt^p)
L = 9.3E-4 s_1; Ki = 1.0 uM; Ka = 0.4 uM; h = 0.0 dimensionless; P_IP3R = 66.6 s_1Reaction: Ca_ER => Ca_Cyt; IP3_Cyt, Rate Law: Cytoplasm*(L+P_IP3R*IP3_Cyt^3*Ca_Cyt^3*h^3/((IP3_Cyt+Ki)^3*(Ca_Cyt+Ka)^3))*(Ca_ER-Ca_Cyt)
n_hill = 3.0 dimensionless; Kc = 2.0E-5 uM; Vcp = 1.8E-4 uM_s_1Reaction: => Oc; Orai1, Rate Law: PM*Vcp*Orai1^n_hill/(Kc^n_hill+Orai1^n_hill)
r_hill = 4.0 dimensionless; Orai1_t = 0.001 uMReaction: Orai1 = Orai1_t-(r_hill*Oc+r_hill*O_o), Rate Law: missing
kdeg = 0.5 s_1; K_deg = 0.1 uMReaction: IP3_Cyt => ; Ca_Cyt, Rate Law: Cytoplasm*kdeg*Ca_Cyt^2/(K_deg^2+Ca_Cyt^2)*IP3_Cyt

States:

NameDescription
S2a[Stromal interaction molecule 1]
O oO_o
S4[Stromal interaction molecule 1]
Orai1[Calcium release-activated calcium channel protein 1]
Ca Cyt[calcium(2+); Calcium cation]
S2[Stromal interaction molecule 1]
Ca ER[calcium(2+); Calcium cation]
IP3 Cyt[1D-myo-inositol 1,4,5-trisphosphate; D-myo-Inositol 1,4,5-trisphosphate]
OcOc

Chickarmane2006 - Stem cell switch irreversible: BIOMD0000000204v0.0.1

Chickarmane2006 - Stem cell switch irreversibleKinetic modeling approach of the transcriptional dynamics of the embryoni…

Details

Recent ChIP experiments of human and mouse embryonic stem cells have elucidated the architecture of the transcriptional regulatory circuitry responsible for cell determination, which involves the transcription factors OCT4, SOX2, and NANOG. In addition to regulating each other through feedback loops, these genes also regulate downstream target genes involved in the maintenance and differentiation of embryonic stem cells. A search for the OCT4-SOX2-NANOG network motif in other species reveals that it is unique to mammals. With a kinetic modeling approach, we ascribe function to the observed OCT4-SOX2-NANOG network by making plausible assumptions about the interactions between the transcription factors at the gene promoter binding sites and RNA polymerase (RNAP), at each of the three genes as well as at the target genes. We identify a bistable switch in the network, which arises due to several positive feedback loops, and is switched on/off by input environmental signals. The switch stabilizes the expression levels of the three genes, and through their regulatory roles on the downstream target genes, leads to a binary decision: when OCT4, SOX2, and NANOG are expressed and the switch is on, the self-renewal genes are on and the differentiation genes are off. The opposite holds when the switch is off. The model is extremely robust to parameter changes. In addition to providing a self-consistent picture of the transcriptional circuit, the model generates several predictions. Increasing the binding strength of NANOG to OCT4 and SOX2, or increasing its basal transcriptional rate, leads to an irreversible bistable switch: the switch remains on even when the activating signal is removed. Hence, the stem cell can be manipulated to be self-renewing without the requirement of input signals. We also suggest tests that could discriminate between a variety of feedforward regulation architectures of the target genes by OCT4, SOX2, and NANOG. link: http://identifiers.org/pubmed/16978048

Parameters:

NameDescription
c1 = 1.0; f = 1000.0; d3 = 0.001; eta3 = 1.0E-4; c2 = 0.01; d2 = 0.001; c3 = 0.5; d1 = 0.0011Reaction: SOX2_Gene => SOX2; A, OCT4_SOX2, NANOG, Rate Law: (eta3+c1*A+c2*OCT4_SOX2+c3*OCT4_SOX2*NANOG)/(1+eta3/f+d1*A+d2*OCT4_SOX2+d3*OCT4_SOX2*NANOG)
a2 = 0.01; b2 = 0.001; a3 = 0.5; f = 1000.0; b3 = 0.001; a1 = 1.0; eta1 = 1.0E-4; b1 = 0.0011Reaction: OCT4_Gene => OCT4; A, OCT4_SOX2, NANOG, Rate Law: (eta1+a1*A+a2*OCT4_SOX2+a3*OCT4_SOX2*NANOG)/(1+eta1/f+b1*A+b2*OCT4_SOX2+b3*OCT4_SOX2*NANOG)
gamma4 = 0.01Reaction: Protein => degradation, Rate Law: gamma4*Protein
h1 = 0.0011; g1 = 0.1; f2 = 0.001; h2 = 1.0; eta7 = 1.0E-4Reaction: targetGene => Protein; OCT4_SOX2, NANOG, Rate Law: (g1*OCT4_SOX2+eta7)/(1+eta7/f2+h1*OCT4_SOX2+h2*OCT4_SOX2*NANOG)
f3 = 0.05; f = 1000.0; e1 = 0.01; f1 = 0.001; e2 = 0.1; f2 = 0.001; eta5 = 1.0E-4Reaction: NANOG_Gene => NANOG; OCT4_SOX2, p53, Rate Law: (eta5+e1*OCT4_SOX2+e2*OCT4_SOX2*NANOG)/(1+eta5/f+f2*OCT4_SOX2+f1*OCT4_SOX2*NANOG+f3*p53)
gamma2 = 1.0Reaction: NANOG => degradation, Rate Law: gamma2*NANOG
k2c = 0.001; k1c = 0.05Reaction: OCT4 + SOX2 => OCT4_SOX2, Rate Law: k1c*OCT4*SOX2-k2c*OCT4_SOX2
gamma1 = 1.0Reaction: OCT4 => degradation, Rate Law: gamma1*OCT4
k3c = 5.0Reaction: OCT4_SOX2 => degradation, Rate Law: k3c*OCT4_SOX2
gamma3 = 1.0Reaction: SOX2 => degradation, Rate Law: gamma3*SOX2

States:

NameDescription
OCT4 Gene[POU5F1; POU domain, class 5, transcription factor 1]
SOX2 Gene[QSOX2; Sulfhydryl oxidase 2]
targetGenetargetGene
NANOG[Putative homeobox protein NANOG2]
NANOG Gene[Putative homeobox protein NANOG2; NANOGP1]
SOX2[Sulfhydryl oxidase 2]
OCT4[POU domain, class 5, transcription factor 1]
ProteinProtein
degradationdegradation
OCT4 SOX2[POU domain, class 5, transcription factor 1; Sulfhydryl oxidase 2]

Chickarmane2006 - Stem cell switch reversible: BIOMD0000000203v0.0.1

Chickarmane2006 - Stem cell switch reversibleKinetic modeling approach of the transcriptional dynamics of the embryonic…

Details

Recent ChIP experiments of human and mouse embryonic stem cells have elucidated the architecture of the transcriptional regulatory circuitry responsible for cell determination, which involves the transcription factors OCT4, SOX2, and NANOG. In addition to regulating each other through feedback loops, these genes also regulate downstream target genes involved in the maintenance and differentiation of embryonic stem cells. A search for the OCT4-SOX2-NANOG network motif in other species reveals that it is unique to mammals. With a kinetic modeling approach, we ascribe function to the observed OCT4-SOX2-NANOG network by making plausible assumptions about the interactions between the transcription factors at the gene promoter binding sites and RNA polymerase (RNAP), at each of the three genes as well as at the target genes. We identify a bistable switch in the network, which arises due to several positive feedback loops, and is switched on/off by input environmental signals. The switch stabilizes the expression levels of the three genes, and through their regulatory roles on the downstream target genes, leads to a binary decision: when OCT4, SOX2, and NANOG are expressed and the switch is on, the self-renewal genes are on and the differentiation genes are off. The opposite holds when the switch is off. The model is extremely robust to parameter changes. In addition to providing a self-consistent picture of the transcriptional circuit, the model generates several predictions. Increasing the binding strength of NANOG to OCT4 and SOX2, or increasing its basal transcriptional rate, leads to an irreversible bistable switch: the switch remains on even when the activating signal is removed. Hence, the stem cell can be manipulated to be self-renewing without the requirement of input signals. We also suggest tests that could discriminate between a variety of feedforward regulation architectures of the target genes by OCT4, SOX2, and NANOG. link: http://identifiers.org/pubmed/16978048

Parameters:

NameDescription
f = 1000.0; f2 = 9.95E-4; f1 = 0.001; e1 = 0.005; e2 = 0.1; f3 = 0.01; eta5 = 1.0E-4Reaction: NANOG_Gene => NANOG; OCT4_SOX2, p53, Rate Law: (eta5+e1*OCT4_SOX2+e2*OCT4_SOX2*NANOG)/(1+eta5/f+f2*OCT4_SOX2+f1*OCT4_SOX2*NANOG+f3*p53)
gamma4 = 0.01Reaction: Protein => degradation, Rate Law: gamma4*Protein
a2 = 0.01; b2 = 0.001; b3 = 7.0E-4; a3 = 0.2; f = 1000.0; a1 = 1.0; eta1 = 1.0E-4; b1 = 0.0011Reaction: OCT4_Gene => OCT4; A, OCT4_SOX2, NANOG, Rate Law: (eta1+a1*A+a2*OCT4_SOX2+a3*OCT4_SOX2*NANOG)/(1+eta1/f+b1*A+b2*OCT4_SOX2+b3*OCT4_SOX2*NANOG)
f = 1000.0; g1 = 0.1; h1 = 0.0019; h2 = 0.05; eta7 = 1.0E-4Reaction: targetGene => Protein; OCT4_SOX2, NANOG, Rate Law: (g1*OCT4_SOX2+eta7)/(1+eta7/f+h1*OCT4_SOX2+h2*OCT4_SOX2*NANOG)
c1 = 1.0; f = 1000.0; eta3 = 1.0E-4; c2 = 0.01; c3 = 0.2; d2 = 0.001; d3 = 7.0E-4; d1 = 0.0011Reaction: SOX2_Gene => SOX2; A, OCT4_SOX2, NANOG, Rate Law: (eta3+c1*A+c2*OCT4_SOX2+c3*OCT4_SOX2*NANOG)/(1+eta3/f+d1*A+d2*OCT4_SOX2+d3*OCT4_SOX2*NANOG)
gamma2 = 1.0Reaction: NANOG => degradation, Rate Law: gamma2*NANOG
k2c = 0.001; k1c = 0.05Reaction: OCT4 + SOX2 => OCT4_SOX2, Rate Law: k1c*OCT4*SOX2-k2c*OCT4_SOX2
gamma1 = 1.0Reaction: OCT4 => degradation, Rate Law: gamma1*OCT4
k3c = 5.0Reaction: OCT4_SOX2 => degradation, Rate Law: k3c*OCT4_SOX2
gamma3 = 1.0Reaction: SOX2 => degradation, Rate Law: gamma3*SOX2

States:

NameDescription
OCT4 Gene[POU5F1; POU domain, class 5, transcription factor 1]
SOX2 Gene[QSOX2; Sulfhydryl oxidase 2]
OCT4[POU domain, class 5, transcription factor 1]
targetGenetargetGene
NANOG Gene[NANOGP1; Putative homeobox protein NANOG2]
SOX2[Sulfhydryl oxidase 2]
NANOG[Putative homeobox protein NANOG2]
ProteinProtein
degradationdegradation
OCT4 SOX2[POU domain, class 5, transcription factor 1; Sulfhydryl oxidase 2]

Chickarmane2008 - Stem cell lineage - NANOG GATA-6 switch: BIOMD0000000210v0.0.1

Chickarmane2008 - Stem cell lineage - NANOG GATA-6 switchIn this work, a dynamical model of lineage determination based…

Details

Recent studies have associated the transcription factors, Oct4, Sox2 and Nanog as parts of a self-regulating network which is responsible for maintaining embryonic stem cell properties: self renewal and pluripotency. In addition, mutual antagonism between two of these and other master regulators have been shown to regulate lineage determination. In particular, an excess of Cdx2 over Oct4 determines the trophectoderm lineage whereas an excess of Gata-6 over Nanog determines differentiation into the endoderm lineage. Also, under/over-expression studies of the master regulator Oct4 have revealed that some self-renewal/pluripotency as well as differentiation genes are expressed in a biphasic manner with respect to the concentration of Oct4.We construct a dynamical model of a minimalistic network, extracted from ChIP-on-chip and microarray data as well as literature studies. The model is based upon differential equations and makes two plausible assumptions; activation of Gata-6 by Oct4 and repression of Nanog by an Oct4-Gata-6 heterodimer. With these assumptions, the results of simulations successfully describe the biphasic behavior as well as lineage commitment. The model also predicts that reprogramming the network from a differentiated state, in particular the endoderm state, into a stem cell state, is best achieved by over-expressing Nanog, rather than by suppression of differentiation genes such as Gata-6.The computational model provides a mechanistic understanding of how different lineages arise from the dynamics of the underlying regulatory network. It provides a framework to explore strategies of reprogramming a cell from a differentiated state to a stem cell state through directed perturbations. Such an approach is highly relevant to regenerative medicine since it allows for a rapid search over the host of possibilities for reprogramming to a stem cell state. link: http://identifiers.org/pubmed/18941526

Parameters:

NameDescription
i2 = 0.1; j1 = 0.1; i1 = 0.1; j0 = 0.1; i0 = 0.001Reaction: GCNF_Gene => GCNF; CDX2, GATA6, Rate Law: (i0+i1*CDX2+i2*GATA6)/(1+j0*CDX2+j1*GATA6)
c1 = 0.05; d1 = 0.05; d2 = 0.0125; d3 = 0.05; c2 = 0.0125Reaction: GATA6_Gene => GATA6; OCT4_SOX2, NANOG, Rate Law: (c1*OCT4_SOX2+c2*GATA6)/(1+d1*OCT4_SOX2+d2*GATA6+d3*NANOG)
gamman = 0.01Reaction: NANOG => degradation, Rate Law: gamman*NANOG
gamma1 = 0.1Reaction: OCT4 => degradation, Rate Law: gamma1*OCT4
a0 = 0.001; a2 = 0.0125; b1 = 0.02; a1 = 0.02; b3 = 0.03; a3 = 0.025; b5 = 10.0; b0 = 1.0; b4 = 10.0; b2 = 0.0125Reaction: OCT4_Gene => OCT4; A, SOX2, NANOG, CDX2, GCNF, Rate Law: (a0+a1*A+a2*OCT4*SOX2+a3*OCT4*SOX2*NANOG)/(1+b0*A+b1*OCT4+b2*OCT4*SOX2+b3*OCT4*SOX2*NANOG+b4*CDX2*OCT4+b5*GCNF)
c1 = 0.05; d1 = 0.05; d2 = 0.0125; d0 = 0.001; c2 = 0.0125; c0 = 0.001Reaction: SOX2_Gene => SOX2; OCT4, NANOG, Rate Law: (c0+c1*OCT4*SOX2+c2*OCT4*SOX2*NANOG)/(1+d0*OCT4+d1*OCT4*SOX2+d2*OCT4*SOX2*NANOG)
gammag = 0.01Reaction: GATA6 => degradation, Rate Law: gammag*GATA6
a2 = 0.0125; b1 = 0.02; a1 = 0.02; b3 = 0.03; b2 = 0.0125Reaction: NANOG_Gene => NANOG; OCT4_SOX2, GATA6, Rate Law: (a1*OCT4_SOX2+a2*OCT4_SOX2*NANOG)/(1+b1*OCT4_SOX2+b2*OCT4_SOX2*NANOG+b3*OCT4_SOX2*GATA6)
gamma5 = 0.1Reaction: GCNF => degradation, Rate Law: gamma5*GCNF
gamma4 = 0.1Reaction: CDX2 => degradation, Rate Law: gamma4*CDX2
gamma2 = 0.1Reaction: SOX2 => degradation, Rate Law: gamma2*SOX2
g0 = 0.001; h0 = 2.0; g1 = 2.0; h1 = 5.0Reaction: CDX2_Gene => CDX2; OCT4, Rate Law: (g0+g1*CDX2)/(1+h0*CDX2+h1*CDX2*OCT4)

States:

NameDescription
GCNF[Nuclear receptor subfamily 6 group A member 1]
CDX2 Gene[CDX2; Homeobox protein CDX-2]
GATA6[Transcription factor GATA-6]
SOX2 Gene[QSOX2; Sulfhydryl oxidase 2]
GCNF Gene[NR6A1; Nuclear receptor subfamily 6 group A member 1]
NANOG Gene[NANOGP1; Putative homeobox protein NANOG2]
SOX2[Sulfhydryl oxidase 2]
CDX2[Homeobox protein CDX-2]
OCT4 Gene[POU5F1; POU domain, class 5, transcription factor 1]
GATA6 Gene[GATA6; Transcription factor GATA-6]
OCT4[POU domain, class 5, transcription factor 1]
NANOG[Putative homeobox protein NANOG2]
degradationdegradation

Chickarmane2008 - Stem cell lineage determination: BIOMD0000000209v0.0.1

Chickarmane2008 - Stem cell lineage determinationIn this work, a dynamical model of lineage determination based upon a…

Details

Recent studies have associated the transcription factors, Oct4, Sox2 and Nanog as parts of a self-regulating network which is responsible for maintaining embryonic stem cell properties: self renewal and pluripotency. In addition, mutual antagonism between two of these and other master regulators have been shown to regulate lineage determination. In particular, an excess of Cdx2 over Oct4 determines the trophectoderm lineage whereas an excess of Gata-6 over Nanog determines differentiation into the endoderm lineage. Also, under/over-expression studies of the master regulator Oct4 have revealed that some self-renewal/pluripotency as well as differentiation genes are expressed in a biphasic manner with respect to the concentration of Oct4.We construct a dynamical model of a minimalistic network, extracted from ChIP-on-chip and microarray data as well as literature studies. The model is based upon differential equations and makes two plausible assumptions; activation of Gata-6 by Oct4 and repression of Nanog by an Oct4-Gata-6 heterodimer. With these assumptions, the results of simulations successfully describe the biphasic behavior as well as lineage commitment. The model also predicts that reprogramming the network from a differentiated state, in particular the endoderm state, into a stem cell state, is best achieved by over-expressing Nanog, rather than by suppression of differentiation genes such as Gata-6.The computational model provides a mechanistic understanding of how different lineages arise from the dynamics of the underlying regulatory network. It provides a framework to explore strategies of reprogramming a cell from a differentiated state to a stem cell state through directed perturbations. Such an approach is highly relevant to regenerative medicine since it allows for a rapid search over the host of possibilities for reprogramming to a stem cell state. link: http://identifiers.org/pubmed/18941526

Parameters:

NameDescription
d1 = 0.005; c1 = 0.005; d2 = 0.025; d0 = 0.001; c2 = 0.025; c0 = 0.001Reaction: SOX2_Gene => SOX2; OCT4, NANOG, Rate Law: (c0+c1*OCT4*SOX2+c2*OCT4*SOX2*NANOG)/(1+d0*OCT4+d1*OCT4*SOX2+d2*OCT4*SOX2*NANOG)
i2 = 0.1; j1 = 0.1; i1 = 0.1; j0 = 0.1; i0 = 0.001Reaction: GCNF_Gene => GCNF; CDX2, GATA6, Rate Law: (i0+i1*CDX2+i2*GATA6)/(1+j0*CDX2+j1*GATA6)
gamma1 = 0.1Reaction: OCT4 => degradation, Rate Law: gamma1*OCT4
e1 = 0.1; e3 = 1.0; f0 = 0.001; f1 = 0.1; e2 = 0.1; f2 = 0.1; e0 = 0.001; f3 = 10.0; f4 = 1.0Reaction: NANOG_Gene => NANOG; OCT4, SOX2, GATA6, SN, Rate Law: (e0+e1*OCT4*SOX2+e2*OCT4*SOX2*NANOG+e3*SN)/(1+f0*OCT4+f1*OCT4*SOX2+f2*OCT4*SOX2*NANOG+f3*OCT4*GATA6+f4*SN)
gamma3 = 0.1Reaction: NANOG => degradation, Rate Law: gamma3*NANOG
gammag = 0.1Reaction: GATA6 => degradation, Rate Law: gammag*GATA6
gamma5 = 0.1Reaction: GCNF => degradation, Rate Law: gamma5*GCNF
gamma4 = 0.1Reaction: CDX2 => degradation, Rate Law: gamma4*CDX2
a0 = 0.001; b1 = 0.001; b2 = 0.005; b3 = 0.025; a3 = 0.025; b5 = 10.0; a1 = 1.0; b0 = 1.0; b4 = 10.0; a2 = 0.005Reaction: OCT4_Gene => OCT4; A, SOX2, NANOG, CDX2, GCNF, Rate Law: (a0+a1*A+a2*OCT4*SOX2+a3*OCT4*SOX2*NANOG)/(1+b0*A+b1*OCT4+b2*OCT4*SOX2+b3*OCT4*SOX2*NANOG+b4*CDX2*OCT4+b5*GCNF)
gamma2 = 0.1Reaction: SOX2 => degradation, Rate Law: gamma2*SOX2
q2 = 15.0; p2 = 2.5E-4; p0 = 0.1; q1 = 2.5E-4; q0 = 1.0; p1 = 1.0; q3 = 10.0Reaction: GATA6_Gene => GATA6; OCT4, NANOG, SG, Rate Law: (p0+p1*OCT4+p2*GATA6)/(1+q0*OCT4+q1*GATA6+q2*NANOG+q3*SG)
g0 = 0.001; h0 = 2.0; g1 = 2.0; h1 = 5.0Reaction: CDX2_Gene => CDX2; OCT4, Rate Law: (g0+g1*CDX2)/(1+h0*CDX2+h1*CDX2*OCT4)

States:

NameDescription
GCNF[Nuclear receptor subfamily 6 group A member 1]
CDX2 Gene[CDX2; Homeobox protein CDX-2]
GATA6[Transcription factor GATA-6]
SOX2 Gene[QSOX2; Sulfhydryl oxidase 2]
GCNF Gene[NR6A1; Nuclear receptor subfamily 6 group A member 1]
NANOG Gene[NANOGP1; Putative homeobox protein NANOG2]
SOX2[Sulfhydryl oxidase 2]
CDX2[Homeobox protein CDX-2]
OCT4 Gene[POU5F1; POU domain, class 5, transcription factor 1]
GATA6 Gene[GATA6; Transcription factor GATA-6]
OCT4[POU domain, class 5, transcription factor 1]
NANOG[Putative homeobox protein NANOG2]
degradationdegradation

Chiorino2002 - G1/S transition model: MODEL2003180003v0.0.1

mathematical approach to model the protein interactions regulating the transition from the G1 phase to the phase of DNA…

Details

Cell cycle duration and phase transition times are not fixed, even within homogeneous cell populations growing under optimal environmental conditions. We investigate G(1) phase variability from the molecular point of view and propose a mathematical approach to model the protein interactions regulating the transition from the G(1) phase to the phase of DNA synthesis. The mathematical model has some connections with flow cytometry experimental data. link: http://identifiers.org/pubmed/11965250

Chitnis2008 - Mathematical model of malaria transmission: BIOMD0000000949v0.0.1

Mathematical model of malaria transmission for low and high transmission rates.

Details

We perform sensitivity analyses on a mathematical model of malaria transmission to determine the relative importance of model parameters to disease transmission and prevalence. We compile two sets of baseline parameter values: one for areas of high transmission and one for low transmission. We compute sensitivity indices of the reproductive number (which measures initial disease transmission) and the endemic equilibrium point (which measures disease prevalence) to the parameters at the baseline values. We find that in areas of low transmission, the reproductive number and the equilibrium proportion of infectious humans are most sensitive to the mosquito biting rate. In areas of high transmission, the reproductive number is again most sensitive to the mosquito biting rate, but the equilibrium proportion of infectious humans is most sensitive to the human recovery rate. This suggests strategies that target the mosquito biting rate (such as the use of insecticide-treated bed nets and indoor residual spraying) and those that target the human recovery rate (such as the prompt diagnosis and treatment of infectious individuals) can be successful in controlling malaria. link: http://identifiers.org/pubmed/18293044

Parameters:

NameDescription
v_h = 0.1Reaction: Exposed_Human => Infected_Human, Rate Law: Human*v_h*Exposed_Human
N_h = 623.0; Psi_h = 5.5E-5Reaction: => Susceptible_Human, Rate Law: Human*Psi_h*N_h
lambda_v = 2.93379660870939E-4Reaction: Susceptible_Mosquito => Exposed_Mosquito, Rate Law: Mosquito*lambda_v*Susceptible_Mosquito
v_v = 0.083Reaction: Exposed_Mosquito => Infected_Mosquito, Rate Law: Mosquito*v_v*Exposed_Mosquito
lambda_h = 4.48218926330601E-5Reaction: Susceptible_Human => Exposed_Human, Rate Law: Human*lambda_h*Susceptible_Human
rho_h = 0.0027Reaction: Recovered => Susceptible_Human, Rate Law: Human*rho_h*Recovered
gamma_h = 0.0035Reaction: Infected_Human => Recovered, Rate Law: Human*gamma_h*Infected_Human
f_h = 1.334E-4Reaction: Infected_Human =>, Rate Law: Human*f_h*Infected_Human
Capital_lambda_h = 0.041Reaction: => Susceptible_Human, Rate Law: Human*Capital_lambda_h
delta_h = 1.8E-5Reaction: Infected_Human =>, Rate Law: Human*delta_h*Infected_Human
Psi_v = 0.13; N_v = 2435.0Reaction: => Susceptible_Mosquito, Rate Law: Mosquito*Psi_v*N_v
f_v = 0.1304Reaction: Infected_Mosquito =>, Rate Law: Mosquito*f_v*Infected_Mosquito

States:

NameDescription
Infected Human[Infection; Homo sapiens]
Exposed Mosquito[Disease Transmission; C123547]
Susceptible Human[0005461]
Recovered[Recovery]
Susceptible Mosquito[0005461]
Infected Mosquito[Infection; C123547]
Exposed Human[C156623; Homo sapiens]

Chitnis2012 - Model Rift Valley Fever transmission between cattle and mosquitoes (Model 1): BIOMD0000000950v0.0.1

Mathematical model for Rift Valley Fever transmission between cattle and mosquitoes without infectious eggs.

Details

We present two ordinary differential equation models for Rift Valley fever (RVF) transmission in cattle and mosquitoes. We extend existing models for vector-borne diseases to include an asymptomatic host class and vertical transmission in vectors. We define the basic reproductive number, ℛ(0), and analyse the existence and stability of equilibrium points. We compute sensitivity indices of ℛ(0) and a reactivity index (that measures epidemicity) to parameters for baseline wet and dry season values. ℛ(0) is most sensitive to the mosquito biting and death rates. The reactivity index is most sensitive to the mosquito biting rate and the infectivity of hosts to vectors. Numerical simulations show that even with low equilibrium prevalence, increases in mosquito densities through higher rainfall, in the presence of vertical transmission, can result in large epidemics. This suggests that vertical transmission is an important factor in the size and persistence of RVF epidemics. link: http://identifiers.org/pubmed/23098257

Parameters:

NameDescription
gamma_h = 0.25; u_h = 4.5662100456621E-4; gamma_tilde_h = 0.25Reaction: R_h = (gamma_h*I_h+gamma_tilde_h*A_h)-u_h*R_h, Rate Law: (gamma_h*I_h+gamma_tilde_h*A_h)-u_h*R_h
lambda_h = 5.143359375E-5; theta_h = 0.4; u_h = 4.5662100456621E-4; gamma_tilde_h = 0.25Reaction: A_h = theta_h*lambda_h*S_h-(u_h+gamma_tilde_h)*A_h, Rate Law: theta_h*lambda_h*S_h-(u_h+gamma_tilde_h)*A_h
u_v = 0.05; lambda_v = 0.0; N_v = 20000.0; M0 = 20000.0; psi_v = 0.1Reaction: S_v = ((N_v-psi_v*I_v)/N_v*u_v*M0-lambda_v*S_v)-u_v*S_v, Rate Law: ((N_v-psi_v*I_v)/N_v*u_v*M0-lambda_v*S_v)-u_v*S_v
u_v = 0.05; N_v = 20000.0; M0 = 20000.0; v_v = 0.0714285714285714; psi_v = 0.1Reaction: I_v = (psi_v*I_v/N_v*u_v*M0+v_v*E_v)-u_v*I_v, Rate Law: (psi_v*I_v/N_v*u_v*M0+v_v*E_v)-u_v*I_v
u_v = 0.05; lambda_v = 0.0; v_v = 0.0714285714285714Reaction: E_v = lambda_v*S_v-(u_v+v_v)*E_v, Rate Law: lambda_v*S_v-(u_v+v_v)*E_v
C0 = 1000.0; lambda_h = 5.143359375E-5; u_h = 4.5662100456621E-4Reaction: S_h = (u_h*C0-lambda_h*S_h)-u_h*S_h, Rate Law: (u_h*C0-lambda_h*S_h)-u_h*S_h
delta_h = 0.1; gamma_h = 0.25; lambda_h = 5.143359375E-5; theta_h = 0.4; u_h = 4.5662100456621E-4Reaction: I_h = (1-theta_h)*lambda_h*S_h-(u_h+gamma_h+delta_h)*I_h, Rate Law: (1-theta_h)*lambda_h*S_h-(u_h+gamma_h+delta_h)*I_h

States:

NameDescription
I v[0000460; 0004757]
S h[C66819; 0003748]
A h[C3833; Infection]
E v[0003748; PATO:0002425]
S v[C66819; 0004757]
R h[0003748; Recovered or Resolved]
I h[0003748; 0000460]

ChowHall2008 Dynamics of Human Weight Change_ODE_1: BIOMD0000000901v0.0.1

This ODE model is a representation of the two compartment macronutrient partition model that Chow and Hall outlined in t…

Details

An imbalance between energy intake and energy expenditure will lead to a change in body weight (mass) and body composition (fat and lean masses). A quantitative understanding of the processes involved, which currently remains lacking, will be useful in determining the etiology and treatment of obesity and other conditions resulting from prolonged energy imbalance. Here, we show that a mathematical model of the macronutrient flux balances can capture the long-term dynamics of human weight change; all previous models are special cases of this model. We show that the generic dynamic behavior of body composition for a clamped diet can be divided into two classes. In the first class, the body composition and mass are determined uniquely. In the second class, the body composition can exist at an infinite number of possible states. Surprisingly, perturbations of dietary energy intake or energy expenditure can give identical responses in both model classes, and existing data are insufficient to distinguish between these two possibilities. Nevertheless, this distinction has important implications for the efficacy of clinical interventions that alter body composition and mass. link: http://identifiers.org/pubmed/18369435

Parameters:

NameDescription
Energy_Expenditure_Rate = 11.05; p___Ratio = 0.038480263286012; Psy = 0.00102051309738594; rho_F = 39.5; Intake_Rate = 9.2Reaction: Fat_Mass = ((1-p___Ratio)*(Intake_Rate-Energy_Expenditure_Rate)-Psy)/rho_F, Rate Law: ((1-p___Ratio)*(Intake_Rate-Energy_Expenditure_Rate)-Psy)/rho_F
Energy_Expenditure_Rate = 11.05; rho_L = 7.6; p___Ratio = 0.038480263286012; Psy = 0.00102051309738594; Intake_Rate = 9.2Reaction: Lean_Mass = (p___Ratio*(Intake_Rate-Energy_Expenditure_Rate)+Psy)/rho_L, Rate Law: (p___Ratio*(Intake_Rate-Energy_Expenditure_Rate)+Psy)/rho_L

States:

NameDescription
Lean Mass[C71258; C81328]
Body Mass[C81328]
Fat Mass[C158256; C81328]

Chrobak2011 - A mathematical model of induced cancer-adaptive immune system competition: BIOMD0000000815v0.0.1

This is a mathematical model describing competition between an artificially induced tumor and the adaptive immune system…

Details

We present a model of competition between an artificially induced tumor and the adaptive immune system based on the use of an autonomous system of ordinary differential equations (ODE). The aim of this work is to reproduce experimental results which find two possible outcomes depending on the initial quantities of the tumor and the adaptive immune cells. The ODE system is positively invariant and its solutions are bounded. The linear stability analysis of the fixed points of the model yields two groups of solutions depending on the initial conditions. In the first one, the immune system wins against the tumor cells, so the cancer disappears (elimination). In the second one, the cancer keeps on growing (escape). These results are coherent with experimental results which show these two possibilities, so the model reproduces the macroscopic behavior of the experiments. From the model some conclusions on the underlying competitive behavior can be derived. link: http://identifiers.org/doi/10.1142/S0218339011004111

Parameters:

NameDescription
a = 0.0625Reaction: => x_Cancer, Rate Law: compartment*a*x_Cancer
d = 0.03125Reaction: => y_Immune_System, Rate Law: compartment*d*y_Immune_System
c = 0.03125Reaction: x_Cancer =>, Rate Law: compartment*c*x_Cancer^2
e = 0.0859375Reaction: y_Immune_System => ; x_Cancer, Rate Law: compartment*e*x_Cancer*y_Immune_System
b = 0.125Reaction: x_Cancer => ; y_Immune_System, Rate Law: compartment*b*x_Cancer*y_Immune_System
f = 0.03125Reaction: y_Immune_System =>, Rate Law: compartment*f*y_Immune_System^2

States:

NameDescription
y Immune System[Immune Cell]
x Cancer[neoplastic cell]

Chung2010 - Genome-scale metabolic network of Pichia pastoris (iPP668): MODEL1507180023v0.0.1

Chung2010 - Genome-scale metabolic network of Pichia pastoris (iPP668)This model is described in the article: [Genome-s…

Details

Pichia pastoris has been recognized as an effective host for recombinant protein production. A number of studies have been reported for improving this expression system. However, its physiology and cellular metabolism still remained largely uncharacterized. Thus, it is highly desirable to establish a systems biotechnological framework, in which a comprehensive in silico model of P. pastoris can be employed together with high throughput experimental data analysis, for better understanding of the methylotrophic yeast's metabolism.A fully compartmentalized metabolic model of P. pastoris (iPP668), composed of 1,361 reactions and 1,177 metabolites, was reconstructed based on its genome annotation and biochemical information. The constraints-based flux analysis was then used to predict achievable growth rate which is consistent with the cellular phenotype of P. pastoris observed during chemostat experiments. Subsequent in silico analysis further explored the effect of various carbon sources on cell growth, revealing sorbitol as a promising candidate for culturing recombinant P. pastoris strains producing heterologous proteins. Interestingly, methanol consumption yields a high regeneration rate of reducing equivalents which is substantial for the synthesis of valuable pharmaceutical precursors. Hence, as a case study, we examined the applicability of P. pastoris system to whole-cell biotransformation and also identified relevant metabolic engineering targets that have been experimentally verified.The genome-scale metabolic model characterizes the cellular physiology of P. pastoris, thus allowing us to gain valuable insights into the metabolism of methylotrophic yeast and devise possible strategies for strain improvement through in silico simulations. This computational approach, combined with synthetic biology techniques, potentially forms a basis for rational analysis and design of P. pastoris metabolic network to enhance humanized glycoprotein production. link: http://identifiers.org/pubmed/20594333

Ciliberto2003 - CyclinE / Cdk2 timer in the cell cycle of Xenopus laevis embryo: BIOMD0000000697v0.0.1

Ciliberto2003 - CyclinE / Cdk2 timer in the cell cycle of Xenopus laevis embryoThis model is described in the article:…

Details

Early cell cycles of Xenopus laevis embryos are characterized by rapid oscillations in the activity of two cyclin-dependent kinases. Cdk1 activity peaks at mitosis, driven by periodic degradation of cyclins A and B. In contrast, Cdk2 activity oscillates twice per cell cycle, despite a constant level of its partner, cyclin E. Cyclin E degrades at a fixed time after fertilization, normally corresponding to the midblastula transition. Based on published data and new experiments, we constructed a mathematical model in which: (1) oscillations in Cdk2 activity depend upon changes in phosphorylation, (2) Cdk2 participates in a negative feedback loop with the inhibitory kinase Wee1; (3) cyclin E is cooperatively removed from the oscillatory system; and (4) removed cyclin E is degraded by a pathway activated by cyclin E/Cdk2 itself. The model's predictions about embryos injected with Xic1, a stoichiometric inhibitor of cyclin E/Cdk2, were experimentally validated. link: http://identifiers.org/pubmed/12914904

Parameters:

NameDescription
kon = 0.02 1/s; phi = 0.0390625 1Reaction: Cdk2_CycE => Cdk2_CycErem, Rate Law: compartment*kon*phi*Cdk2_CycE
kwee = 1.5 1/sReaction: Cdk2_CycE => PCdk2_CycE; Wee1_a, Rate Law: compartment*kwee*Wee1_a*Cdk2_CycE
kwinact = 1.5 1/s; kwact = 0.75 1/s; Jwact = 0.01; Jwinact = 0.01 1Reaction: => Wee1_a; Kin_a, Wee1_total, Rate Law: compartment*(kwact*(Wee1_total-Wee1_a)/((Jwact+Wee1_total)-Wee1_a)-kwinact*Kin_a*Wee1_a/(Jwinact+Wee1_a))
kxdeg = 0.01 1/sReaction: Xic_Cdk2_CycErem => Cdk2_CycErem, Rate Law: compartment*kxdeg*Xic_Cdk2_CycErem
kedeg = 0.017 1/sReaction: Xic_PCdk2_CycErem => Xicrem; Deg_a, Rate Law: compartment*kedeg*Xic_PCdk2_CycErem*Deg_a
Heav = 0.0 1; kdact = 0.023 1/sReaction: => Deg_a, Rate Law: compartment*kdact*Heav
koff = 1.0E-4 1/sReaction: PCdk2_CycErem => PCdk2_CycE, Rate Law: compartment*koff*PCdk2_CycErem
kassoc = 0.1 1/sReaction: Xic + Cdk2_CycErem => Xic_Cdk2_CycErem, Rate Law: compartment*kassoc*Xic*Cdk2_CycErem
kdissoc = 0.001 1/sReaction: Xic_Cdk2_CycErem => Xic + Cdk2_CycErem, Rate Law: compartment*kdissoc*Xic_Cdk2_CycErem
k25A = 0.1 1/sReaction: PCdk2_CycE => Cdk2_CycE, Rate Law: compartment*k25A*PCdk2_CycE
Jiinact = 0.01; kiinact = 0.6 1/s; kiact = 0.15 1/s; Jiact = 0.01Reaction: Cdk2_CycE => Kin_a + Cdk2_CycE; Cdk2_CycE, Rate Law: compartment*(kiact*(1-Kin_a)/((Jiact+1)-Kin_a)-kiinact*Cdk2_CycE*Kin_a/(Jiinact+Kin_a))

States:

NameDescription
PCdk2 CycE[G1/S-specific cyclin-E1; cyclin E1-CDK2 complex; Cyclin-dependent kinase 2]
Xic Cdk2 CycE[G1/S-specific cyclin-E1; protein-containing complex; Cyclin-dependent kinase 2; NP_001108275.1]
PCdk2 CycErem[G1/S-specific cyclin-E1; cyclin E1-CDK2 complex; inactive; Cyclin-dependent kinase 2; phosphorylated]
Cdk2 CycE[G1/S-specific cyclin-E1; cyclin E1-CDK2 complex; Cyclin-dependent kinase 2]
Xicrem[NP_001108275.1; inactive]
Kin a[Kinase]
Xic Cdk2 CycErem[NP_001108275.1; protein-containing complex; inactive; Cyclin-dependent kinase 2; G1/S-specific cyclin-E1]
Xic total[NP_001108275.1]
Xic PCdk2 CycE[G1/S-specific cyclin-E1; protein-containing complex; phosphorylated; Cyclin-dependent kinase 2; NP_001108275.1]
Cyc total[G1/S-specific cyclin-E1]
Deg a[G1/S-specific cyclin-E1; Protein Degradation Process]
Cdk2 CycErem[Cyclin-dependent kinase 2; cyclin E1-CDK2 complex; inactive; G1/S-specific cyclin-E1]
Wee1 a[Wee1-like protein kinase 1-B; urn:miriam:pato:PATO_0002354]
Xic PCdk2 CycErem[Cyclin-dependent kinase 2; protein-containing complex; phosphorylated; G1/S-specific cyclin-E1; inactive; NP_001108275.1]
Xic[NP_001108275.1]

Ciliberto2003_Morphogenesis_Checkpoint: BIOMD0000000297v0.0.1

This a model from the article: Mathematical model of the morphogenesis checkpoint in budding yeast. Ciliberto A, Nov…

Details

The morphogenesis checkpoint in budding yeast delays progression through the cell cycle in response to stimuli that prevent bud formation. Central to the checkpoint mechanism is Swe1 kinase: normally inactive, its activation halts cell cycle progression in G2. We propose a molecular network for Swe1 control, based on published observations of budding yeast and analogous control signals in fission yeast. The proposed Swe1 network is merged with a model of cyclin-dependent kinase regulation, converted into a set of differential equations and studied by numerical simulation. The simulations accurately reproduce the phenotypes of a dozen checkpoint mutants. Among other predictions, the model attributes a new role to Hsl1, a kinase known to play a role in Swe1 degradation: Hsl1 must also be indirectly responsible for potent inhibition of Swe1 activity. The model supports the idea that the morphogenesis checkpoint, like other checkpoints, raises the cell size threshold for progression from one phase of the cell cycle to the next. link: http://identifiers.org/pubmed/14691135

Parameters:

NameDescription
kssic = 0.1Reaction: => Sic, Rate Law: kssic
ksbud = 0.1Reaction: => BE; Cln, Rate Law: ksbud*Cln
kdcln = 0.1Reaction: Cln =>, Rate Law: kdcln*Cln
kdclb_tripleprime = 0.1; kdclb_prime = 0.015; kdclb_doubleprime = 1.0Reaction: Trim => Sic; Cdh1, Cdc20a, Rate Law: Trim*(kdclb_doubleprime*Cdh1+kdclb_tripleprime*Cdc20a+kdclb_prime)
mu = 0.005Reaction: => mass, Rate Law: mu*mass
jicdc20 = 0.001; kicdc20 = 0.25Reaction: Cdc20a => Cdc20, Rate Law: Cdc20a*kicdc20/(jicdc20+Cdc20a)
kscdc20_doubleprime = 0.3; kscdc20_prime = 0.005; jscdc20 = 0.3Reaction: => Cdc20; Clb, Rate Law: kscdc20_prime+kscdc20_doubleprime*Clb^4/(jscdc20^4+Clb^4)
Jawee = 0.05; Vawee = 0.3Reaction: PSwe1 => Swe1, Rate Law: PSwe1*Vawee/(Jawee+PSwe1)
kdswe_prime = 0.007Reaction: Swe1 =>, Rate Law: kdswe_prime*Swe1
jacdc20 = 0.001; kacdc20 = 1.0Reaction: Cdc20 => Cdc20a; IE, Rate Law: kacdc20*Cdc20*IE/(jacdc20+Cdc20)
kdbud = 0.1Reaction: BE =>, Rate Law: kdbud*BE
kdsic = 0.01; kdsic_prime = 1.0; kdsic_doubleprime = 3.0Reaction: Trim => Clb; Cln, Rate Law: Trim*(kdsic_prime*Cln+kdsic_doubleprime*Clb+kdsic)
kdswe_doubleprime = 0.05Reaction: PSwe1M =>, Rate Law: kdswe_doubleprime*PSwe1M
kmih = 0.0Reaction: PTrim => Trim, Rate Law: PTrim*kmih
Vamih = 1.0; Jamih = 0.1; Mih1 = 0.0Reaction: => Mih1a; Clb, Rate Law: Vamih*Mih1*Clb/(Jamih+Mih1)
kimcm = 0.15; jimcm = 0.1Reaction: Mcm =>, Rate Law: Mcm*kimcm/(jimcm+Mcm)
kass = 300.0Reaction: Sic + Clb => Trim, Rate Law: kass*Sic*Clb
jimih = 0.1; Vimih = 0.3Reaction: Mih1a =>, Rate Law: Mih1a*Vimih/(jimih+Mih1a)
BUD = 0.0; khsl1 = 1.0Reaction: Swe1 => Swe1M, Rate Law: khsl1*BUD*Swe1
Kacdh_doubleprime = 10.0; jacdh = 0.01; Cdh1in = 0.0; Kacdh_prime = 1.0Reaction: => Cdh1; Cdc20a, Rate Law: Cdh1in*(Kacdh_prime+Kacdh_doubleprime*Cdc20a)/(jacdh+Cdh1in)
Viwee = 1.0; Jiwee = 0.05Reaction: Swe1M => PSwe1M; Clb, Rate Law: Viwee*Swe1M*Clb/(Jiwee+Swe1M)
ksswe = 0.0025Reaction: => Swe1; SBF, Rate Law: ksswe*SBF
kisbf_doubleprime = 2.0; kisbf_prime = 1.0; jisbf = 0.01Reaction: SBF => ; Clb, Rate Law: SBF*(kisbf_prime+kisbf_doubleprime*Clb)/(jisbf+SBF)
kssweC = 0.0Reaction: => Swe1, Rate Law: kssweC
kscln = 0.1Reaction: => Cln; SBF, Rate Law: kscln*SBF
SBFin = 0.0; kasbf_doubleprime = 0.0; jasbf = 0.01; kasbf_prime = 1.0Reaction: => SBF; mass, Cln, Rate Law: SBFin*(kasbf_prime*mass+kasbf_doubleprime*Cln)/(jasbf+SBFin)
kswe = 0.0Reaction: Clb => PClb, Rate Law: kswe*Clb
Jm = 10.0; ksclb = 0.015; eps = 0.5Reaction: => Clb; mass, Mcm, Rate Law: ksclb*mass*Jm*(eps+Mcm)/(mass+Jm)
kdcdc20 = 0.1Reaction: Cdc20a =>, Rate Law: kdcdc20*Cdc20a
Mcmin = 0.0; jamcm = 0.1; kamcm = 1.0Reaction: => Mcm; Clb, Rate Law: Mcmin*Clb*kamcm/(jamcm+Mcmin)
kdiss = 0.1Reaction: Trim => Sic + Clb, Rate Law: kdiss*Trim
IEin = 0.0; kaie = 0.1; jaie = 0.01Reaction: => IE; Clb, Rate Law: kaie*IEin*Clb/(jaie+IEin)
khsl1r = 0.01Reaction: Swe1M => Swe1, Rate Law: khsl1r*Swe1M
jiie = 0.01; kiie = 0.04Reaction: IE =>, Rate Law: IE*kiie/(jiie+IE)
jicdh = 0.01; kicdh = 35.0; kicdh_prime = 2.0Reaction: Cdh1 => ; Clb, Cln, Rate Law: Cdh1*(kicdh*Clb+kicdh_prime*Cln)/(jicdh+Cdh1)

States:

NameDescription
PSwe1M[Mitosis inhibitor protein kinase SWE1]
Mih1a[M-phase inducer phosphatase]
Trim[Protein SIC1; G2/mitotic-specific cyclin-2; Pre-mRNA-splicing factor ATP-dependent RNA helicase-like protein cdc28]
Cln[G1/S-specific cyclin CLN2; G1/S-specific cyclin CLN1]
Clb[G2/mitotic-specific cyclin-2]
Cdc20a[APC/C activator protein CDC20]
PSwe1[Mitosis inhibitor protein kinase SWE1]
PTrim[G2/mitotic-specific cyclin-2; Protein SIC1; Pre-mRNA-splicing factor ATP-dependent RNA helicase-like protein cdc28]
BEBE
Mcm[Pheromone receptor transcription factor]
IEIntermediary Enzyme
SBF[G1-specific transcription factors activator MSA1]
Swe1M[Mitosis inhibitor protein kinase SWE1]
massmass
Swe1[Mitosis inhibitor protein kinase SWE1]
Cdh1[APC/C activator protein CDH1]
PClb[G2/mitotic-specific cyclin-2]
Cdc20[APC/C activator protein CDC20]
Sic[Protein SIC1]

Ciliberto2003_Swe1Network: MODEL0913285268v0.0.1

This a model from the article: Mathematical model of the morphogenesis checkpoint in budding yeast. Ciliberto A, Nov…

Details

The morphogenesis checkpoint in budding yeast delays progression through the cell cycle in response to stimuli that prevent bud formation. Central to the checkpoint mechanism is Swe1 kinase: normally inactive, its activation halts cell cycle progression in G2. We propose a molecular network for Swe1 control, based on published observations of budding yeast and analogous control signals in fission yeast. The proposed Swe1 network is merged with a model of cyclin-dependent kinase regulation, converted into a set of differential equations and studied by numerical simulation. The simulations accurately reproduce the phenotypes of a dozen checkpoint mutants. Among other predictions, the model attributes a new role to Hsl1, a kinase known to play a role in Swe1 degradation: Hsl1 must also be indirectly responsible for potent inhibition of Swe1 activity. The model supports the idea that the morphogenesis checkpoint, like other checkpoints, raises the cell size threshold for progression from one phase of the cell cycle to the next. link: http://identifiers.org/pubmed/14691135

Ciliberto2005 - Steady states and oscillations in the p53/Mdm2 network: BIOMD0000001006v0.0.1

Its a mathematial model studying steady state and oscialltions in p53-MDM2 network triggered by IR induced DNA Damage.

Details

p53 is activated in response to events compromising the genetic integrity of a cell. Recent data show that p53 activity does not increase steadily with genetic damage but rather fluctuates in an oscillatory fashion. Theoretical studies suggest that oscillations can arise from a combination of positive and negative feedbacks or from a long negative feedback loop alone. Both negative and positive feedbacks are present in the p53/Mdm2 network, but it is not known what roles they play in the oscillatory response to DNA damage. We developed a mathematical model of p53 oscillations based on positive and negative feedbacks in the p53/Mdm2 network. According to the model, the system reacts to DNA damage by moving from a stable steady state into a region of stable limit cycles. Oscillations in the model are born with large amplitude, which guarantees an all-or-none response to damage. As p53 oscillates, damage is repaired and the system moves back to a stable steady state with low p53 activity. The model reproduces experimental data in quantitative detail. We suggest new experiments for dissecting the contributions of negative and positive feedbacks to the generation of oscillations. link: http://identifiers.org/pubmed/15725723

Clancy2001_Kchannel: BIOMD0000000121v0.0.1

This model is according to the paper *Cellular consequences of HEGR mutations in the long QT syndrome: precursors to sud…

Details

A variety of mutations in HERG, the major subunit of the rapidly activating component of the cardiac delayed rectifier I(Kr), have been found to underlie the congenital Long-QT syndrome, LQT2. LQT2 may give rise to severe arrhythmogenic phenotypes leading to sudden cardiac death.We attempt to elucidate the mechanisms by which heterogeneous LQT2 genotypes can lead to prolongation of the action potential duration (APD) and consequently the QT interval on the ECG.We develop Markovian models of wild-type (WT) and mutant I(Kr) channels and incorporate these models into a comprehensive model of the cardiac ventricular cell.Using this virtual transgenic cell model, we describe the effects of HERG mutations on the cardiac ventricular action potential (AP) and provide insight into the mechanism by which each defect results in a net loss of repolarizing current and prolongation of APD.This study demonstrates which mutations can prolong APD sufficiently to generate early afterdepolarizations (EADs), which may trigger life-threatening arrhythmias. The severity of the phenotype is shown to depend on the specific kinetic changes and how they affect I(Kr) during the time course of the action potential. Clarifying how defects in HERG can lead to impaired cellular electrophysiology can improve our understanding of the link between channel structure and cellular function. link: http://identifiers.org/pubmed/11334834

Parameters:

NameDescription
ain = 2.172; bin = 1.077Reaction: c2 => c1, Rate Law: (ain*c2-bin*c1)*cell
ai = NaN; bi = NaNReaction: o => i, Rate Law: (o*bi-ai*i)*cell
b = NaN; a = NaNReaction: c3 => c2, Rate Law: (a*c3-b*c2)*cell
bb = NaN; aa = NaNReaction: c1 => o, Rate Law: (c1*aa-bb*o)*cell
u = NaN; aa = NaNReaction: c1 => i, Rate Law: (aa*c1-u*i)*cell
v = -40.0; vk = NaN; Gk = NaNReaction: ik = Gk*o*(v-vk), Rate Law: missing

States:

NameDescription
c2[IPR003967; voltage-gated potassium channel complex]
c1[IPR003967; voltage-gated potassium channel complex]
c3[IPR003967; voltage-gated potassium channel complex]
o[IPR003967; voltage-gated potassium channel complex]
ikcardiac delayed rectifier current
i[IPR003967; voltage-gated potassium channel complex]

Clancy2002_CardiacSodiumChannel_WT: BIOMD0000000126v0.0.1

The model is according to the paper *Na+ Channel Mutation That Causes Both Brugada and Long-QT Syndrome Phenotypes: A Si…

Details

Complex physiological interactions determine the functional consequences of gene abnormalities and make mechanistic interpretation of phenotypes extremely difficult. A recent example is a single mutation in the C terminus of the cardiac Na(+) channel, 1795insD. The mutation causes two distinct clinical syndromes, long QT (LQT) and Brugada, leading to life-threatening cardiac arrhythmias. Coexistence of these syndromes is seemingly paradoxical; LQT is associated with enhanced Na(+) channel function, and Brugada with reduced function.Using a computational approach, we demonstrate that the 1795insD mutation exerts variable effects depending on the myocardial substrate. We develop Markov models of the wild-type and 1795insD cardiac Na(+) channels. By incorporating the models into a virtual transgenic cell, we elucidate the mechanism by which 1795insD differentially disrupts cellular electrical behavior in epicardial and midmyocardial cell types. We provide a cellular mechanistic basis for the ECG abnormalities observed in patients carrying the 1795insD gene mutation.We demonstrate that the 1795insD mutation can cause both LQT and Brugada syndromes through interaction with the heterogeneous myocardium in a rate-dependent manner. The results highlight the complexity and multiplicity of genotype-phenotype relationships, and the usefulness of computational approaches in establishing a mechanistic link between genetic defects and functional abnormalities. link: http://identifiers.org/pubmed/11889015

Parameters:

NameDescription
a11 = NaN; b11 = NaNReaction: IC3 => IC2, Rate Law: cell*(IC3*a11-IC2*b11)
a4 = NaN; b4 = NaNReaction: IF => IM1, Rate Law: cell*(IF*a4-IM1*b4)
b12 = NaN; a12 = NaNReaction: IC2 => IF, Rate Law: cell*(IC2*a12-IF*b12)
b5 = NaN; a5 = NaNReaction: IM1 => IM2, Rate Law: cell*(IM1*a5-IM2*b5)
a2 = NaN; b2 = NaNReaction: IF => O, Rate Law: cell*(IF*b2-a2*O)
a3 = NaN; b3 = NaNReaction: C2 => IC2, Rate Law: cell*(C2*b3-IC2*a3)
a13 = NaN; b13 = NaNReaction: O => C1, Rate Law: cell*((-C1)*a13+O*b13)

States:

NameDescription
IC3[IPR001696; voltage-gated sodium channel complex]
IC2[IPR001696; voltage-gated sodium channel complex]
IM2[IPR001696; voltage-gated sodium channel complex]
C1[IPR001696; voltage-gated sodium channel complex]
IM1[IPR001696; voltage-gated sodium channel complex]
C2[IPR001696; voltage-gated sodium channel complex]
C3[IPR001696; voltage-gated sodium channel complex]
IF[IPR001696; voltage-gated sodium channel complex]
O[IPR001696; voltage-gated sodium channel complex]

Claret2009 - Predicting phase III overall survival in colorectal cancer: MODEL1708310001v0.0.1

Claret2009 - Predicting phase III overall survival in colorectal cancerThis model is described in the article: [Model-b…

Details

PURPOSE: We developed a drug-disease simulation model to predict antitumor response and overall survival in phase III studies from longitudinal tumor size data in phase II trials. METHODS: We developed a longitudinal exposure-response tumor-growth inhibition (TGI) model of drug effect (and resistance) using phase II data of capecitabine (n = 34) and historical phase III data of fluorouracil (FU; n = 252) in colorectal cancer (CRC); and we developed a parametric survival model that related change in tumor size and patient characteristics to survival time using historical phase III data (n = 245). The models were validated in simulation of antitumor response and survival in an independent phase III study (n = 1,000 replicates) of capecitabine versus FU in CRC. RESULTS: The TGI model provided a good fit of longitudinal tumor size data. A lognormal distribution best described the survival time, and baseline tumor size and change in tumor size from baseline at week 7 were predictors (P < .00001). Predicted change of tumor size and survival time distributions in the phase III study for both capecitabine and FU were consistent with observed values, for example, 431 days (90% prediction interval, 362 to 514 days) versus 401 days observed for survival in the capecitabine arm. A modest survival improvement of 39 days (90% prediction interval, -21 to 110 days) versus 35 days observed was predicted for capecitabine. CONCLUSION: The modeling framework successfully predicted survival in a phase III trial on the basis of capecitabine phase II data in CRC. It is a useful tool to support end-of-phase II decisions and design of phase III studies. link: http://identifiers.org/pubmed/19636014

Clarke2000 - One-hit model of cell death in neuronal degenerations: BIOMD0000000538v0.0.1

Clarke2000 - One-hit model of cell death in neuronal degenerationsThis one-hit model fits different neuronal-death assoc…

Details

In genetic disorders associated with premature neuronal death, symptoms may not appear for years or decades. This delay in clinical onset is often assumed to reflect the occurrence of age-dependent cumulative damage. For example, it has been suggested that oxidative stress disrupts metabolism in neurological degenerative disorders by the cumulative damage of essential macromolecules. A prediction of the cumulative damage hypothesis is that the probability of cell death will increase over time. Here we show in contrast that the kinetics of neuronal death in 12 models of photoreceptor degeneration, hippocampal neurons undergoing excitotoxic cell death, a mouse model of cerebellar degeneration and Parkinson's and Huntington's diseases are all exponential and better explained by mathematical models in which the risk of cell death remains constant or decreases exponentially with age. These kinetics argue against the cumulative damage hypothesis; instead, the time of death of any neuron is random. Our findings are most simply accommodated by a 'one-hit' biochemical model in which mutation imposes a mutant steady state on the neuron and a single event randomly initiates cell death. This model appears to be common to many forms of neurodegeneration and has implications for therapeutic strategies. link: http://identifiers.org/pubmed/10910361

Parameters:

NameDescription
Rrom = 0.103; ONLrom_0 = 40.947; Murom = 0.0708Reaction: ONLrom = ONLrom_0*exp((exp((-Rrom)*time)-1)*Murom/Rrom)*100/ONLrom_0, Rate Law: missing
Mupcd = 0.223Reaction: ONLpcd = (-Mupcd)*ONLpcd, Rate Law: (-Mupcd)*ONLpcd
Munr = 0.278Reaction: ONLnr = (-Munr)*ONLnr, Rate Law: (-Munr)*ONLnr

States:

NameDescription
ONLrom[outer nuclear layer]
ONLpcd[outer nuclear layer]
ONLnr[outer nuclear layer]

Clarke2006_Smad_signalling: BIOMD0000000112v0.0.1

The model reproduces the temporal evolution of four variables depicted in Fig 2a. The solution is generated for median p…

Details

Transforming growth factor-beta (TGFbeta) signalling is an important regulator of cellular growth and differentiation. The principal intracellular mediators of TGFbeta signalling are the Smad proteins, which upon TGFbeta stimulation accumulate in the nucleus and regulate the transcription of target genes. To investigate the mechanisms of Smad nuclear accumulation, we developed a simple mathematical model of canonical Smad signalling. The model was built using both published data and our experimentally determined cellular Smad concentrations (isoforms 2, 3 and 4). We found in mink lung epithelial cells that Smad2 (8.5-12 x 10(4) molecules cell(-1)) was present in similar amounts to Smad4 (9.3-12 x 10(4) molecules cell(-1)), whereas both were in excess of Smad3 (1.1-2.0 x 10(4) molecules cell(-1)). Variation of the model parameters and statistical analysis showed that Smad nuclear accumulation is most sensitive to parameters affecting the rates of R-Smad phosphorylation and dephosphorylation and Smad complex formation/ dissociation in the nucleus. Deleting Smad4 from the model revealed that rate-limiting phospho-R-Smad dephosphorylation could be an important mechanism for Smad nuclear accumulation. Furthermore, we observed that binding factors constitutively localised to the nucleus do not efficiently mediate Smad nuclear accumulation, if dephosphorylation is rapid. We therefore conclude that an imbalance in the rates of R-Smad phosphorylation and dephosphorylation is likely an important mechanism of Smad nuclear accumulation during TGFbeta signalling. link: http://identifiers.org/pubmed/17186703

Parameters:

NameDescription
k6d=0.0492 min_inv; k6a=1.44E-4 per item per minReaction: R_smad_P_smad4_nuc => smad4_nuc + R_smad_P_nuc, Rate Law: k6d*R_smad_P_smad4_nuc-k6a*smad4_nuc*R_smad_P_nuc
k3=16.6 min_invReaction: R_smad_P_smad4_cyt => R_smad_P_smad4_nuc, Rate Law: k3*R_smad_P_smad4_cyt
k5nc=5.63 min_inv; k5cn=0.563 min_invReaction: R_smad_nuc => R_smad_cyt, Rate Law: k5nc*R_smad_nuc-k5cn*R_smad_cyt
k2d=0.0399 min_inv; k2a=6.5E-5 per item per minReaction: R_smad_P_cyt + smad4_cyt => R_smad_P_smad4_cyt, Rate Law: k2a*R_smad_P_cyt*smad4_cyt-k2d*R_smad_P_smad4_cyt
k4cn=0.00497 min_inv; k4nc=0.783 min_invReaction: smad4_nuc => smad4_cyt, Rate Law: k4nc*smad4_nuc-k4cn*smad4_cyt
K7=8950.0 item; Vmax7=17100.0 items per minReaction: R_smad_P_nuc => R_smad_nuc + Pi, Rate Law: Vmax7*R_smad_P_nuc/(K7+R_smad_P_nuc)
KCAT=3.51 min_inv; K1=289000.0 itemReaction: R_smad_cyt => R_smad_P_cyt; receptor, Rate Law: KCAT*receptor*R_smad_cyt/(K1+R_smad_cyt)

States:

NameDescription
receptor[TGF-beta receptor type-1; TGF-beta receptor type-2]
smad4 cyt[Mothers against decapentaplegic homolog 4]
R smad P cyt[Mothers against decapentaplegic homolog 2]
R smad P nuc[Mothers against decapentaplegic homolog 2]
R smad P smad4 nuc[Mothers against decapentaplegic homolog 2; Mothers against decapentaplegic homolog 4]
R smad cyt[Mothers against decapentaplegic homolog 2]
R smad nuc[Mothers against decapentaplegic homolog 2]
smad4 nuc[Mothers against decapentaplegic homolog 4]
Pi[phosphate(3-); Orthophosphate]
R smad P smad4 cyt[Mothers against decapentaplegic homolog 4; Mothers against decapentaplegic homolog 2]

Cloutier2009 - Brain Energy Metabolism: BIOMD0000000554v0.0.1

Cloutier2009 - Brain Energy Metabolism This model was taken from the  [CellMLrepository](http://www.cellml.org/models)…

Details

An integrative, systems approach to the modelling of brain energy metabolism is presented. Mechanisms such as glutamate cycling between neurons and astrocytes and glycogen storage in astrocytes have been implemented. A unique feature of the model is its calibration using in vivo data of brain glucose and lactate from freely moving rats under various stimuli. The model has been used to perform simulated perturbation experiments that show that glycogen breakdown in astrocytes is significantly activated during sensory (tail pinch) stimulation. This mechanism provides an additional input of energy substrate during high consumption phases. By way of validation, data from the perfusion of 50 microM propranolol in the rat brain was compared with the model outputs. Propranolol affects the glucose dynamics during stimulation, and this was accurately reproduced in the model by a reduction in the glycogen breakdown in astrocytes. The model's predictive capacity was verified by using data from a sensory stimulation (restraint) that was not used for model calibration. Finally, a sensitivity analysis was conducted on the model parameters, this showed that the control of energy metabolism and transport processes are critical in the metabolic behaviour of cerebral tissue. link: http://identifiers.org/pubmed/19396534

Parameters:

NameDescription
Vn_ldh = -0.001026864256; Vn_mito = 0.0129174754920542; Vn_pk = 0.0120203036981555Reaction: PYRn = Vn_pk-(Vn_ldh+Vn_mito), Rate Law: Vn_pk-(Vn_ldh+Vn_mito)
Vn_pgi = 0.00600284722882977; Vn_hk = 0.00600093047858717Reaction: G6Pn = Vn_hk-Vn_pgi, Rate Law: Vn_hk-Vn_pgi
Vn_pgk = 0.012002606302138; Vn_pfk = 0.00599809710207478Reaction: GAPn = 2*Vn_pfk-Vn_pgk, Rate Law: 2*Vn_pfk-Vn_pgk
Vg_hk = 0.00455613617326311; Vg_glyp = 3.51571428571429E-5; Vg_pgi = 0.00451935700191414; Vg_glys = 9.08171994158688E-5Reaction: G6Pg = (Vg_hk+Vg_glyp)-(Vg_pgi+Vg_glys), Rate Law: (Vg_hk+Vg_glyp)-(Vg_pgi+Vg_glys)
Vg_pfk = 0.00450657384340637; Vg_pgk = 0.0090457605321121Reaction: GAPg = 2*Vg_pfk-Vg_pgk, Rate Law: 2*Vg_pfk-Vg_pgk
Vg_glyp = 3.51571428571429E-5; Vg_glys = 9.08171994158688E-5Reaction: GLYg = Vg_glys-Vg_glyp, Rate Law: Vg_glys-Vg_glyp
Vg_pgk = 0.0090457605321121; Vg_pk = 0.00906366080685179Reaction: PEPg = Vg_pgk-Vg_pk, Rate Law: Vg_pgk-Vg_pk
Vn_mito = 0.0129174754920542; NAero = 3.0; Vcn_O2 = 0.0390504186958046Reaction: O2n = Vcn_O2-NAero*Vn_mito, Rate Law: Vcn_O2-NAero*Vn_mito
Vn_ldh = -0.001026864256; Vne_LAC = -0.00101735054205471Reaction: LACn = Vn_ldh-Vne_LAC, Rate Law: Vn_ldh-Vne_LAC
NAero = 3.0; Vcg_O2 = 0.0180867710645166; Vg_mito = 0.0060112916441682Reaction: O2g = Vcg_O2-NAero*Vg_mito, Rate Law: Vcg_O2-NAero*Vg_mito
ATPtot = 2.379Reaction: AMPg = ATPtot-(ATPg+ADPg), Rate Law: missing
Vn_ck = 2.93701651940294E-5Reaction: PCrn = -Vn_ck, Rate Law: -Vn_ck
Rcg = 0.022; Vc_GLC = 0.69774545454546; Vce_GLC = 0.0154673938740423; Vcg_GLC = 0.00297412147754264; Rce = 0.0275Reaction: GLCc = Vc_GLC-(Vce_GLC*1/Rce+Vcg_GLC*1/Rcg), Rate Law: Vc_GLC-(Vce_GLC*1/Rce+Vcg_GLC*1/Rcg)
Reg = 0.8; Veg_GLU = 0.0; Ren = 0.444444444444444; Vn_stim_GLU = 0.0Reaction: GLUe = Vn_stim_GLU*1/Ren-Veg_GLU*1/Reg, Rate Law: Vn_stim_GLU*1/Ren-Veg_GLU*1/Reg
Vn_pgi = 0.00600284722882977; Vn_pfk = 0.00599809710207478Reaction: F6Pn = Vn_pgi-Vn_pfk, Rate Law: Vn_pgi-Vn_pfk
Vgc_LAC = 1.46095762940601E-5; Rcg = 0.022; Vec_LAC = 0.0014407850610198; Vc_LAC = -0.0528; Rce = 0.0275Reaction: LACc = Vc_LAC+Vec_LAC*1/Rce+Vgc_LAC*1/Rcg, Rate Law: Vc_LAC+Vec_LAC*1/Rce+Vgc_LAC*1/Rcg
Vn_pgk = 0.012002606302138; Vn_mito = 0.0129174754920542; Vn_pk = 0.0120203036981555; Vn_ATPase = 0.0488683691708698; Vn_pump = 0.158300842198194; Vn_ck = 2.93701651940294E-5; dAMP_dATPn = -0.101010798503538; nOP = 15.0; Vn_hk = 0.00600093047858717; Vn_pfk = 0.00599809710207478Reaction: ATPn = ((Vn_pgk+Vn_pk+nOP*Vn_mito+Vn_ck)-(Vn_hk+Vn_pfk+Vn_ATPase+Vn_pump))*(1-dAMP_dATPn)^(-1), Rate Law: ((Vn_pgk+Vn_pk+nOP*Vn_mito+Vn_ck)-(Vn_hk+Vn_pfk+Vn_ATPase+Vn_pump))*(1-dAMP_dATPn)^(-1)
Vg_pgi = 0.00451935700191414; Vg_pfk = 0.00450657384340637Reaction: F6Pg = Vg_pgi-Vg_pfk, Rate Law: Vg_pgi-Vg_pfk
Vn_pgk = 0.012002606302138; Vn_pk = 0.0120203036981555Reaction: PEPn = Vn_pgk-Vn_pk, Rate Law: Vn_pgk-Vn_pk
Vn_ldh = -0.001026864256; Vn_pgk = 0.012002606302138; Vn_mito = 0.0129174754920542Reaction: NADHn = Vn_pgk-(Vn_ldh+Vn_mito), Rate Law: Vn_pgk-(Vn_ldh+Vn_mito)
Vg_ldh = 0.003039015294; Vg_pgk = 0.0090457605321121; Vg_mito = 0.0060112916441682Reaction: NADHg = Vg_pgk-(Vg_ldh+Vg_mito), Rate Law: Vg_pgk-(Vg_ldh+Vg_mito)
qak = 0.92; ATPtot = 2.379Reaction: ADPg = ATPg/2*((-qak)+(qak^2+4*qak*(ATPtot/ATPg-1))^(1/2)), Rate Law: missing
Vc_O2 = 4.01410909090909; Rcg = 0.022; Vcn_O2 = 0.0390504186958046; Rcn = 0.01222; Vcg_O2 = 0.0180867710645166Reaction: O2c = Vc_O2-(Vcn_O2*1/Rcn+Vcg_O2*1/Rcg), Rate Law: Vc_O2-(Vcn_O2*1/Rcn+Vcg_O2*1/Rcg)
Vg_hk = 0.00455613617326311; Vg_ck = 8.98869880248884E-5; Vg_pgk = 0.0090457605321121; Vg_pfk = 0.00450657384340637; Vg_pump = 0.0634531133946177; Vg_pk = 0.00906366080685179; Vg_gs = 0.0; nOP = 15.0; dAMP_dATPg = -0.115857415908852; Vg_ATPase = 0.035641088799643; Vg_mito = 0.0060112916441682Reaction: ATPg = ((Vg_pgk+Vg_pk+nOP*Vg_mito+Vg_ck)-(Vg_hk+Vg_pfk+Vg_ATPase+Vg_pump+Vg_gs))*(1-dAMP_dATPg)^(-1), Rate Law: ((Vg_pgk+Vg_pk+nOP*Vg_mito+Vg_ck)-(Vg_hk+Vg_pfk+Vg_ATPase+Vg_pump+Vg_gs))*(1-dAMP_dATPg)^(-1)
Vn_leak_Na = 0.474905958264092; Vn_pump = 0.158300842198194; Vn_stim = 0.0Reaction: NAn = (Vn_leak_Na+Vn_stim)-3*Vn_pump, Rate Law: (Vn_leak_Na+Vn_stim)-3*Vn_pump
Rng = 1.8; Vg_gs = 0.0; Vn_stim_GLU = 0.0Reaction: GLUn = Vg_gs*1/Rng-Vn_stim_GLU, Rate Law: Vg_gs*1/Rng-Vn_stim_GLU
Reg = 0.8; Ren = 0.444444444444444; Vec_LAC = 0.0014407850610198; Vge_LAC = 0.00298013264659761; Vne_LAC = -0.00101735054205471Reaction: LACe = (Vne_LAC*1/Ren+Vge_LAC*1/Reg)-Vec_LAC, Rate Law: (Vne_LAC*1/Ren+Vge_LAC*1/Reg)-Vec_LAC
Rcg = 0.022; Vgc_CO2 = 0.0180338749325046; Vc_CO2 = 4.01454545454546; Rcn = 0.01222; Vnc_CO2 = 0.0387524264761627Reaction: CO2c = (Vnc_CO2*1/Rcn+Vgc_CO2*1/Rcg)-Vc_CO2, Rate Law: (Vnc_CO2*1/Rcn+Vgc_CO2*1/Rcg)-Vc_CO2
Vg_leak_Na = 0.190378997692294; Veg_GLU = 0.0; Vg_pump = 0.0634531133946177Reaction: NAg = (Vg_leak_Na+3*Veg_GLU)-3*Vg_pump, Rate Law: (Vg_leak_Na+3*Veg_GLU)-3*Vg_pump
Vg_ldh = 0.003039015294; Vg_pk = 0.00906366080685179; Vg_mito = 0.0060112916441682Reaction: PYRg = Vg_pk-(Vg_ldh+Vg_mito), Rate Law: Vg_pk-(Vg_ldh+Vg_mito)
Veg_GLC = 0.00158470181577655; Vg_hk = 0.00455613617326311; Vcg_GLC = 0.00297412147754264Reaction: GLCg = (Vcg_GLC+Veg_GLC)-Vg_hk, Rate Law: (Vcg_GLC+Veg_GLC)-Vg_hk
Vg_ldh = 0.003039015294; Vgc_LAC = 1.46095762940601E-5; Vge_LAC = 0.00298013264659761Reaction: LACg = Vg_ldh-(Vge_LAC+Vgc_LAC), Rate Law: Vg_ldh-(Vge_LAC+Vgc_LAC)
Veg_GLC = 0.00158470181577655; Reg = 0.8; Ren = 0.444444444444444; Vce_GLC = 0.0154673938740423; V_en_GLC = 0.00599865999248041Reaction: GLCe = Vce_GLC-(Veg_GLC*1/Reg+V_en_GLC*1/Ren), Rate Law: Vce_GLC-(Veg_GLC*1/Reg+V_en_GLC*1/Ren)
V_en_GLC = 0.00599865999248041; Vn_hk = 0.00600093047858717Reaction: GLCn = V_en_GLC-Vn_hk, Rate Law: V_en_GLC-Vn_hk
Veg_GLU = 0.0; Vg_gs = 0.0Reaction: GLUg = Veg_GLU-Vg_gs, Rate Law: Veg_GLU-Vg_gs
Vg_ck = 8.98869880248884E-5Reaction: PCrg = -Vg_ck, Rate Law: -Vg_ck

States:

NameDescription
G6Pg[D-glucopyranose 6-phosphate]
PYRn[pyruvate]
GLCe[glucose]
GLYg[glycogen]
AMPn[AMP]
NADg[NAD(+)]
PCrg[N-phosphocreatine]
NADHn[NADH]
PEPg[phosphoenolpyruvic acid]
PCrn[N-phosphocreatine]
F6Pn[D-fructose 6-phosphate(2-)]
GAPn[glyceraldehyde 3-phosphate]
PEPn[phosphoenolpyruvate]
NAn[sodium(1+)]
LACn[(S)-lactic acid]
O2n[singlet dioxygen]
GLUn[glutamic acid]
AMPg[AMP]
PYRg[pyruvate]
GLUe[glutamic acid]
ADPn[ADP]
F6Pg[D-fructose 6-phosphate(2-)]
O2g[singlet dioxygen]
G6Pn[D-glucopyranose 6-phosphate]
NAg[sodium(1+)]
GLCc[glucose]
CO2c[carbon dioxide]
LACc[(S)-lactic acid]
GLUg[glutamic acid]
GLCn[glucose]
ADPg[ADP]
ATPn[ATP]
GLCg[glucose]
CRn[creatine]
ATPg[ATP]
NADn[NAD(+)]
NADHg[NADH]
O2c[singlet dioxygen]
LACg[(S)-lactic acid]
CRg[creatine]
LACe[(S)-lactic acid]
GAPg[glyceraldehyde 3-phosphate]

Cloutier2009_EnergyMetabolism_ModelA: MODEL1006230010v0.0.1

This a model from the article: The control systems structures of energy metabolism. Cloutier M, Wellstead P. J R Soc…

Details

The biochemical regulation of energy metabolism (EM) allows cells to modulate their energetic output depending on available substrates and requirements. To this end, numerous biomolecular mechanisms exist that allow the sensing of the energetic state and corresponding adjustment of enzymatic reaction rates. This regulation is known to induce dynamic systems properties such as oscillations or perfect adaptation. Although the various mechanisms of energy regulation have been studied in detail from many angles at the experimental and theoretical levels, no framework is available for the systematic analysis of EM from a control systems perspective. In this study, we have used principles well known in control to clarify the basic system features that govern EM. The major result is a subdivision of the biomolecular mechanisms of energy regulation in terms of widely used engineering control mechanisms: proportional, integral, derivative control, and structures: feedback, cascade and feed-forward control. Evidence for each mechanism and structure is demonstrated and the implications for systems properties are shown through simulations. As the equivalence between biological systems and control components presented here is generic, it is also hypothesized that our work could eventually have an applicability that is much wider than the focus of the current study. link: http://identifiers.org/pubmed/19828503

Cloutier2009_EnergyMetabolism_ModelB: MODEL1006230016v0.0.1

This a model from the article: The control systems structures of energy metabolism. Cloutier M, Wellstead P. J R Soc…

Details

The biochemical regulation of energy metabolism (EM) allows cells to modulate their energetic output depending on available substrates and requirements. To this end, numerous biomolecular mechanisms exist that allow the sensing of the energetic state and corresponding adjustment of enzymatic reaction rates. This regulation is known to induce dynamic systems properties such as oscillations or perfect adaptation. Although the various mechanisms of energy regulation have been studied in detail from many angles at the experimental and theoretical levels, no framework is available for the systematic analysis of EM from a control systems perspective. In this study, we have used principles well known in control to clarify the basic system features that govern EM. The major result is a subdivision of the biomolecular mechanisms of energy regulation in terms of widely used engineering control mechanisms: proportional, integral, derivative control, and structures: feedback, cascade and feed-forward control. Evidence for each mechanism and structure is demonstrated and the implications for systems properties are shown through simulations. As the equivalence between biological systems and control components presented here is generic, it is also hypothesized that our work could eventually have an applicability that is much wider than the focus of the current study. link: http://identifiers.org/pubmed/19828503

Cloutier2009_EnergyMetabolism_ModelC: MODEL1006230068v0.0.1

This a model from the article: The control systems structures of energy metabolism. Cloutier M, Wellstead P. J R Soc…

Details

The biochemical regulation of energy metabolism (EM) allows cells to modulate their energetic output depending on available substrates and requirements. To this end, numerous biomolecular mechanisms exist that allow the sensing of the energetic state and corresponding adjustment of enzymatic reaction rates. This regulation is known to induce dynamic systems properties such as oscillations or perfect adaptation. Although the various mechanisms of energy regulation have been studied in detail from many angles at the experimental and theoretical levels, no framework is available for the systematic analysis of EM from a control systems perspective. In this study, we have used principles well known in control to clarify the basic system features that govern EM. The major result is a subdivision of the biomolecular mechanisms of energy regulation in terms of widely used engineering control mechanisms: proportional, integral, derivative control, and structures: feedback, cascade and feed-forward control. Evidence for each mechanism and structure is demonstrated and the implications for systems properties are shown through simulations. As the equivalence between biological systems and control components presented here is generic, it is also hypothesized that our work could eventually have an applicability that is much wider than the focus of the current study. link: http://identifiers.org/pubmed/19828503

Cloutier2009_EnergyMetabolism_ModelD: MODEL1006230095v0.0.1

This a model from the article: The control systems structures of energy metabolism. Cloutier M, Wellstead P. J R Soc…

Details

The biochemical regulation of energy metabolism (EM) allows cells to modulate their energetic output depending on available substrates and requirements. To this end, numerous biomolecular mechanisms exist that allow the sensing of the energetic state and corresponding adjustment of enzymatic reaction rates. This regulation is known to induce dynamic systems properties such as oscillations or perfect adaptation. Although the various mechanisms of energy regulation have been studied in detail from many angles at the experimental and theoretical levels, no framework is available for the systematic analysis of EM from a control systems perspective. In this study, we have used principles well known in control to clarify the basic system features that govern EM. The major result is a subdivision of the biomolecular mechanisms of energy regulation in terms of widely used engineering control mechanisms: proportional, integral, derivative control, and structures: feedback, cascade and feed-forward control. Evidence for each mechanism and structure is demonstrated and the implications for systems properties are shown through simulations. As the equivalence between biological systems and control components presented here is generic, it is also hypothesized that our work could eventually have an applicability that is much wider than the focus of the current study. link: http://identifiers.org/pubmed/19828503

Cloutier2009_EnergyMetabolism_ModelE: MODEL1006230059v0.0.1

This a model from the article: The control systems structures of energy metabolism. Cloutier M, Wellstead P. J R Soc…

Details

The biochemical regulation of energy metabolism (EM) allows cells to modulate their energetic output depending on available substrates and requirements. To this end, numerous biomolecular mechanisms exist that allow the sensing of the energetic state and corresponding adjustment of enzymatic reaction rates. This regulation is known to induce dynamic systems properties such as oscillations or perfect adaptation. Although the various mechanisms of energy regulation have been studied in detail from many angles at the experimental and theoretical levels, no framework is available for the systematic analysis of EM from a control systems perspective. In this study, we have used principles well known in control to clarify the basic system features that govern EM. The major result is a subdivision of the biomolecular mechanisms of energy regulation in terms of widely used engineering control mechanisms: proportional, integral, derivative control, and structures: feedback, cascade and feed-forward control. Evidence for each mechanism and structure is demonstrated and the implications for systems properties are shown through simulations. As the equivalence between biological systems and control components presented here is generic, it is also hypothesized that our work could eventually have an applicability that is much wider than the focus of the current study. link: http://identifiers.org/pubmed/19828503

Cloutier2009_EnergyMetabolism_ModelF: MODEL1006230096v0.0.1

This a model from the article: The control systems structures of energy metabolism. Cloutier M, Wellstead P. J R Soc…

Details

The biochemical regulation of energy metabolism (EM) allows cells to modulate their energetic output depending on available substrates and requirements. To this end, numerous biomolecular mechanisms exist that allow the sensing of the energetic state and corresponding adjustment of enzymatic reaction rates. This regulation is known to induce dynamic systems properties such as oscillations or perfect adaptation. Although the various mechanisms of energy regulation have been studied in detail from many angles at the experimental and theoretical levels, no framework is available for the systematic analysis of EM from a control systems perspective. In this study, we have used principles well known in control to clarify the basic system features that govern EM. The major result is a subdivision of the biomolecular mechanisms of energy regulation in terms of widely used engineering control mechanisms: proportional, integral, derivative control, and structures: feedback, cascade and feed-forward control. Evidence for each mechanism and structure is demonstrated and the implications for systems properties are shown through simulations. As the equivalence between biological systems and control components presented here is generic, it is also hypothesized that our work could eventually have an applicability that is much wider than the focus of the current study. link: http://identifiers.org/pubmed/19828503

Cloutier2012 - Feedback motif for Parkinson's disease: BIOMD0000000558v0.0.1

Cloutier2012 - Feedback motif for Parkinson's diseaseThis model is described in the article: [Feedback motif for the pa…

Details

Previous article on the integrative modelling of Parkinson's disease (PD) described a mathematical model with properties suggesting that PD pathogenesis is associated with a feedback-induced biochemical bistability. In this article, the authors show that the dynamics of the mathematical model can be extracted and distilled into an equivalent two-state feedback motif whose stability properties are controlled by multi-factorial combinations of risk factors and genetic mutations associated with PD. Based on this finding, the authors propose a principle for PD pathogenesis in the form of the switch-like transition of a bistable feedback process from 'healthy' homeostatic levels of reactive oxygen species and the protein α-synuclein, to an alternative 'disease' state in which concentrations of both molecules are stable at the damagingly high-levels associated with PD. The bistability is analysed using the rate curves and steady-state response characteristics of the feedback motif. In particular, the authors show how a bifurcation in the feedback motif marks the pathogenic moment at which the 'healthy' state is lost and the 'disease' state is initiated. Further analysis shows how known risks (such as: age, toxins and genetic predisposition) modify the stability characteristics of the feedback motif in a way that is compatible with known features of PD, and which explain properties such as: multi-factorial causality, variability in susceptibility and severity, multi-timescale progression and the special cases of familial Parkinson's and Parkinsonian symptoms induced purely by toxic stress. link: http://identifiers.org/pubmed/22757587

Parameters:

NameDescription
S1 = 0.0; k1 = 17280.0; kalphasyn = 8.5; dalphasyn = 15.0Reaction: => ROS; alpha_syn, alpha_syn, Rate Law: Neuron*k1*(1+S1+dalphasyn*(alpha_syn/kalphasyn)^4/(1+(alpha_syn/kalphasyn)^4))
k3 = 0.168; S2_4 = 1.0Reaction: => alpha_syn; ROS, ROS, Rate Law: Neuron*k3*ROS*S2_4
S2_4 = 1.0; k4 = 0.168Reaction: alpha_syn => ; alpha_syn, Rate Law: Neuron*k4*alpha_syn*S2_4
S2_4 = 1.0; k2 = 17280.0Reaction: ROS => ; ROS, Rate Law: Neuron*k2*ROS*S2_4

States:

NameDescription
ROS[reactive oxygen species]
alpha syn[Alpha-synuclein]

Collombet2016 - Lymphoid and myeloid cell specification and transdifferentiation: MODEL1610240000v0.0.1

Collombet2016 - Lymphoid and myeloid cell specification and transdifferentiationThis model is described in the article:…

Details

Blood cells are derived from a common set of hematopoietic stem cells, which differentiate into more specific progenitors of the myeloid and lymphoid lineages, ultimately leading to differentiated cells. This developmental process is controlled by a complex regulatory network involving cytokines and their receptors, transcription factors, and chromatin remodelers. Using public data and data from our own molecular genetic experiments (quantitative PCR, Western blot, EMSA) or genome-wide assays (RNA-sequencing, ChIP-sequencing), we have assembled a comprehensive regulatory network encompassing the main transcription factors and signaling components involved in myeloid and lymphoid development. Focusing on B-cell and macrophage development, we defined a qualitative dynamical model recapitulating cytokine-induced differentiation of common progenitors, the effect of various reported gene knockdowns, and the reprogramming of pre-B cells into macrophages induced by the ectopic expression of specific transcription factors. The resulting network model can be used as a template for the integration of new hematopoietic differentiation and transdifferentiation data to foster our understanding of lymphoid/myeloid cell-fate decisions. link: http://identifiers.org/doi/10.1073/pnas.1610622114

Conant2007_glycolysis_2C: BIOMD0000000177v0.0.1

This a model from the article: Increased glycolytic flux as an outcome of whole-genome duplication in yeast. Conant…

Details

After whole-genome duplication (WGD), deletions return most loci to single copy. However, duplicate loci may survive through selection for increased dosage. Here, we show how the WGD increased copy number of some glycolytic genes could have conferred an almost immediate selective advantage to an ancestor of Saccharomyces cerevisiae, providing a rationale for the success of the WGD. We propose that the loss of other redundant genes throughout the genome resulted in incremental dosage increases for the surviving duplicated glycolytic genes. This increase gave post-WGD yeasts a growth advantage through rapid glucose fermentation; one of this lineage's many adaptations to glucose-rich environments. Our hypothesis is supported by data from enzyme kinetics and comparative genomics. Because changes in gene dosage follow directly from post-WGD deletions, dosage selection can confer an almost instantaneous benefit after WGD, unlike neofunctionalization or subfunctionalization, which require specific mutations. We also show theoretically that increased fermentative capacity is of greatest advantage when glucose resources are both large and dense, an observation potentially related to the appearance of angiosperms around the time of WGD. link: http://identifiers.org/pubmed/17667951

Parameters:

NameDescription
Katp_11=1.5 mM; Vmax_11=1000.0 mMpermin; WGD_E = 0.65 dimensionless; Kadp_11=0.53 mM; Kpyr_11=21.0 mM; Kpep_11=0.14 mM; Keq_11=6500.0 dimensionlessReaction: ADP + PEP => ATP + PYR, Rate Law: cyto*Vmax_11*WGD_E*(PEP*ADP/(Kpep_11*Kadp_11)-PYR*ATP/(Kpep_11*Kadp_11*Keq_11))/((1+PEP/Kpep_11+PYR/Kpyr_11)*(1+ADP/Kadp_11+ATP/Katp_11))
Kf26_4=6.82E-4 mM; Ciatp_4=100.0 dimensionless; Kiatp_4=0.65 mM; Vmax_4=110.0 mMpermin; Cf26_4=0.0174 dimensionless; Camp_4=0.0845 dimensionless; Kf6p_4=0.1 mM; Katp_4=0.71 mM; WGD_E = 0.65 dimensionless; Kamp_4=0.0995 mM; gR_4=5.12 dimensionless; Cf16_4=0.397 dimensionless; Kf16_4=0.111 mM; L0_4=0.66 dimensionless; Catp_4=3.0 dimensionlessReaction: ATP + F6P => ADP + F16bP; AMP, F26bP, Rate Law: cyto*Vmax_4*WGD_E*gR_4*F6P/Kf6p_4*ATP/Katp_4*(1+F6P/Kf6p_4+ATP/Katp_4+gR_4*F6P/Kf6p_4*ATP/Katp_4)/((1+F6P/Kf6p_4+ATP/Katp_4+gR_4*F6P/Kf6p_4*ATP/Katp_4)^2+L0_4*((1+Ciatp_4*ATP/Kiatp_4)/(1+ATP/Kiatp_4))^2*((1+Camp_4*AMP/Kamp_4)/(1+AMP/Kamp_4))^2*((1+Cf26_4*F26bP/Kf26_4+Cf16_4*F16bP/Kf16_4)/(1+F26bP/Kf26_4+F16bP/Kf16_4))^2*(1+Catp_4*ATP/Katp_4)^2)
Ktrehalose_18=2.4 mMperminReaction: ATP + G6P => ADP + Trehalose, Rate Law: cyto*Ktrehalose_18
k_19=21.4 perminReaction: NAD + AcAld => NADH + Succinate, Rate Law: cyto*k_19*AcAld
WGD_E = 0.65 dimensionless; nH_12=1.9 dimensionless; Vmax_12=857.8 mMpermin; Kpyr_12=4.33 mMReaction: PYR => AcAld + CO2, Rate Law: cyto*Vmax_12*WGD_E*(PYR/Kpyr_12)^nH_12/(1+(PYR/Kpyr_12)^nH_12)
Kigap_5=10.0 mM; Keq_5=0.069 mM; Kgap_5=2.4 mM; WGD_E = 0.65 dimensionless; Kdhap_5=2.0 mM; Vmax_5=94.69 mMpermin; Kf16bp_5=0.3 mMReaction: F16bP => DHAP + GAP, Rate Law: cyto*Vmax_5*WGD_E*(F16bP/Kf16bp_5-DHAP*GAP/(Kf16bp_5*Keq_5))/(1+F16bP/Kf16bp_5+DHAP/Kdhap_5+GAP/Kgap_5+F16bP*GAP/(Kf16bp_5*Kigap_5)+DHAP*GAP/(Kdhap_5*Kgap_5))
Ki_NADH=50.0 mM; WGD_E = 0.65 dimensionless; Vmax_PDH=379.2 mMpermin; NADX_tot=8.01 mM; K_PYR=70.0 mM; Ki_PYR=20.0 mM; K_NAD=160.0 mMReaction: PYRmito => AcCoA + CO2mito; NAD, NADH, Rate Law: mito*WGD_E*Vmax_PDH*PYRmito*(NADX_tot-NADX_tot/(1+NAD/NADH))/(NADX_tot*Ki_PYR*K_NAD/Ki_NADH/(1+NAD/NADH)+K_PYR*(NADX_tot-NADX_tot/(1+NAD/NADH))+K_NAD*PYRmito+NADX_tot*K_NAD/Ki_NADH*PYRmito/(1+NAD/NADH)+(NADX_tot-NADX_tot/(1+NAD/NADH))*PYRmito)
k2_6=1.0E7 permin; k1_6=450000.0 perminReaction: DHAP => GAP, Rate Law: cyto*(k1_6*DHAP-k2_6*GAP)
Keq_3=0.29 dimensionless; Kf6p_3=0.3 mM; WGD_E = 0.65 dimensionless; Kg6p_3=1.4 mM; Vmax_3=1056.0 mMperminReaction: G6P => F6P, Rate Law: cyto*Vmax_3*WGD_E*(G6P/Kg6p_3-F6P/(Kg6p_3*Keq_3))/(1+G6P/Kg6p_3+F6P/Kf6p_3)
Keq_8=3200.0 dimensionless; Vmax_8=1288.0 mMpermin; Kbpg_8=0.003 mM; WGD_E = 0.65 dimensionless; Kp3g_8=0.53 mM; Kadp_8=0.2 mM; Katp_8=0.3 mMReaction: ADP + BPG => ATP + P3G, Rate Law: cyto*Vmax_8*WGD_E*(Keq_8*BPG*ADP-P3G*ATP)/(Kp3g_8*Katp_8)/((1+BPG/Kbpg_8+P3G/Kp3g_8)*(1+ADP/Kadp_8+ATP/Katp_8))
k2_15=100.0 permMpermin; k1_15=45.0 permMperminReaction: ADP => ATP + AMP, Rate Law: cyto*(k1_15*ADP*ADP-k2_15*ATP*AMP)
Vmax_1=97.24 mmolepermin; WGD_E = 0.65 dimensionless; Kglc_1=1.1918 mM; Ki_1=0.91 dimensionlessReaction: GLCo => GLCi, Rate Law: Vmax_1*WGD_E*(GLCo-GLCi)/Kglc_1/(1+(GLCo+GLCi)/Kglc_1+Ki_1*GLCo*GLCi/Kglc_1^2)
Knadh_7=0.06 mM; Kgap_7=0.21 mM; Vmaxr_7=6719.0 mMpermin; WGD_E = 0.65 dimensionless; C_7=1.0 dimensionless; Kbpg_7=0.0098 mM; Knad_7=0.09 mM; Vmaxf_7=1152.0 mMperminReaction: GAP + NAD => BPG + NADH, Rate Law: cyto*C_7*(Vmaxf_7*WGD_E*GAP*NAD/(Kgap_7*Knad_7)-Vmaxr_7*WGD_E*BPG*NADH/(Kbpg_7*Knadh_7))/((1+GAP/Kgap_7+BPG/Kbpg_7)*(1+NAD/Knad_7+NADH/Knadh_7))
Kp2g_9=0.08 mM; WGD_E = 0.65 dimensionless; Vmax_9=2585.0 mMpermin; Kp3g_9=1.2 mM; Keq_9=0.19 dimensionlessReaction: P3G => P2G, Rate Law: cyto*Vmax_9*WGD_E*(P3G/Kp3g_9-P2G/(Kp3g_9*Keq_9))/(1+P3G/Kp3g_9+P2G/Kp2g_9)
Kglc_2=0.08 mM; Kadp_2=0.23 mM; Vmax_2=236.7 mMpermin; WGD_E = 0.65 dimensionless; Keq_2=2000.0 dimensionless; Kg6p_2=30.0 mM; Katp_2=0.15 mMReaction: GLCi + ATP => G6P + ADP, Rate Law: cyto*WGD_E*Vmax_2*(GLCi*ATP/(Kglc_2*Katp_2)-G6P*ADP/(Kglc_2*Katp_2*Keq_2))/((1+GLCi/Kglc_2+G6P/Kg6p_2)*(1+ATP/Katp_2+ADP/Kadp_2))
Ketoh_13=17.0 mM; Knad_13=0.17 mM; Kietoh_13=90.0 mM; Kacald_13=1.11 mM; Vmax_13=209.5 mMpermin; Knadh_13=0.11 mM; WGD_E = 0.65 dimensionless; Kinad_13=0.92 mM; Keq_13=6.9E-5 dimensionless; Kiacald_13=1.1 mM; Kinadh_13=0.031 mMReaction: NAD + EtOH => NADH + AcAld, Rate Law: cyto*Vmax_13*WGD_E*(EtOH*NAD/(Ketoh_13*Kinad_13)-AcAld*NADH/(Ketoh_13*Kinad_13*Keq_13))/(1+NAD/Kinad_13+EtOH*Knad_13/(Kinad_13*Ketoh_13)+AcAld*Knadh_13/(Kinadh_13*Kacald_13)+NADH/Kinadh_13+EtOH*NAD/(Kinad_13*Ketoh_13)+NAD*AcAld*Knadh_13/(Kinad_13*Kinadh_13*Kacald_13)+EtOH*NADH*Knad_13/(Kinad_13*Kinadh_13*Ketoh_13)+AcAld*NADH/(Kacald_13*Kinadh_13)+EtOH*NAD*AcAld/(Kinad_13*Kiacald_13*Ketoh_13)+EtOH*AcAld*NADH/(Kietoh_13*Kinadh_13*Kacald_13))
Kpep_10=0.5 mM; Kp2g_10=0.04 mM; WGD_E = 0.65 dimensionless; Vmax_10=201.6 mMpermin; Keq_10=6.7 dimensionlessReaction: P2G => PEP, Rate Law: cyto*Vmax_10*WGD_E*(P2G/Kp2g_10-PEP/(Kp2g_10*Keq_10))/(1+P2G/Kp2g_10+PEP/Kpep_10)
KGLYCOGEN_17=6.0 mMperminReaction: ATP + G6P => ADP + Glycogen, Rate Law: cyto*KGLYCOGEN_17
Katpase_14=39.5 perminReaction: ATP => ADP, Rate Law: cyto*Katpase_14*ATP
k1=1.0 permin; k2=1.0 permin; t_m = 1.0 dimensionlessReaction: PYR => PYRmito, Rate Law: t_m*(k1*PYR-k2*PYRmito)
Knadh_16=0.023 mM; Kdhap_16=0.4 mM; Kglycerol_16=1.0 mM; WGD_E = 0.65 dimensionless; Vmax_16=47.11 mMpermin; Keq_16=4300.0 dimensionless; Knad_16=0.93 mMReaction: DHAP + NADH => NAD + Glycerol, Rate Law: cyto*Vmax_16*WGD_E*(DHAP/Kdhap_16*NADH/Knadh_16-Glycerol/Kdhap_16*NAD/Knadh_16*1/Keq_16)/((1+DHAP/Kdhap_16+Glycerol/Kglycerol_16)*(1+NADH/Knadh_16+NAD/Knad_16))

States:

NameDescription
ATP[ATP; ATP]
Trehalose[alpha,alpha-trehalose; alpha,alpha-Trehalose]
F16bP[beta-D-fructofuranose 1,6-bisphosphate; beta-D-Fructose 1,6-bisphosphate]
AMP[AMP; AMP]
CO2mito[carbon dioxide; CO2]
DHAP[dihydroxyacetone phosphate; Glycerone phosphate]
GLCi[D-glucopyranose; D-Glucose]
P2G[2-phospho-D-glyceric acid; 2-Phospho-D-glycerate]
AcCoAFru2,6-P2
Succinate[succinate(2-); Succinate]
P3G[3-phospho-D-glyceric acid; 3-Phospho-D-glycerate]
GLCo[D-glucopyranose; D-Glucose]
NADH[NADH; NADH]
PYR[pyruvate; Pyruvate]
PYRmito[pyruvate; Pyruvate]
AcAld[acetaldehyde; Acetaldehyde]
EtOH[ethanol; Ethanol]
BPG[3-phospho-D-glyceroyl dihydrogen phosphate; 3-Phospho-D-glyceroyl phosphate]
F6P[beta-D-fructofuranose 6-phosphate; beta-D-Fructose 6-phosphate]
CO2[carbon dioxide; CO2]
Glycerol[glycerol; Glycerol]
G6P[alpha-D-glucose 6-phosphate; alpha-D-Glucose 6-phosphate]
GAP[D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]
Glycogen[glycogen; Glycogen]
PEP[phosphoenolpyruvate; Phosphoenolpyruvate]
ADP[ADP; ADP]
NAD[NAD(+); NAD+]

Conant2007_WGD_glycolysis_2A3AB: BIOMD0000000176v0.0.1

This a model from the article: Increased glycolytic flux as an outcome of whole-genome duplication in yeast. Conant…

Details

After whole-genome duplication (WGD), deletions return most loci to single copy. However, duplicate loci may survive through selection for increased dosage. Here, we show how the WGD increased copy number of some glycolytic genes could have conferred an almost immediate selective advantage to an ancestor of Saccharomyces cerevisiae, providing a rationale for the success of the WGD. We propose that the loss of other redundant genes throughout the genome resulted in incremental dosage increases for the surviving duplicated glycolytic genes. This increase gave post-WGD yeasts a growth advantage through rapid glucose fermentation; one of this lineage's many adaptations to glucose-rich environments. Our hypothesis is supported by data from enzyme kinetics and comparative genomics. Because changes in gene dosage follow directly from post-WGD deletions, dosage selection can confer an almost instantaneous benefit after WGD, unlike neofunctionalization or subfunctionalization, which require specific mutations. We also show theoretically that increased fermentative capacity is of greatest advantage when glucose resources are both large and dense, an observation potentially related to the appearance of angiosperms around the time of WGD. link: http://identifiers.org/pubmed/17667951

Parameters:

NameDescription
nH_12=1.9 dimensionless; WGD_E = 1.0 dimensionless; Vmax_12=857.8 mMpermin; Kpyr_12=4.33 mMReaction: PYR => AcAld + CO2, Rate Law: cyto*Vmax_12*WGD_E*(PYR/Kpyr_12)^nH_12/(1+(PYR/Kpyr_12)^nH_12)
Kpep_10=0.5 mM; Kp2g_10=0.04 mM; Vmax_10=201.6 mMpermin; WGD_E = 1.0 dimensionless; Keq_10=6.7 dimensionless; fV_ENO = 1.0 dimensionlessReaction: P2G => PEP, Rate Law: cyto*Vmax_10*fV_ENO*WGD_E*(P2G/Kp2g_10-PEP/(Kp2g_10*Keq_10))/(1+P2G/Kp2g_10+PEP/Kpep_10)
Ktrehalose_18=2.4 mMperminReaction: ATP + G6P => ADP + Trehalose, Rate Law: cyto*Ktrehalose_18
k_19=21.4 perminReaction: NAD + AcAld => NADH + Succinate, Rate Law: cyto*k_19*AcAld
Kf26_4=6.82E-4 mM; Ciatp_4=100.0 dimensionless; Kiatp_4=0.65 mM; Vmax_4=110.0 mMpermin; Cf26_4=0.0174 dimensionless; fV_PFK = 1.0 dimensionless; Camp_4=0.0845 dimensionless; Kf6p_4=0.1 mM; Katp_4=0.71 mM; Kamp_4=0.0995 mM; gR_4=5.12 dimensionless; Cf16_4=0.397 dimensionless; WGD_E = 1.0 dimensionless; Kf16_4=0.111 mM; L0_4=0.66 dimensionless; Catp_4=3.0 dimensionlessReaction: ATP + F6P => ADP + F16bP; AMP, F26bP, Rate Law: cyto*Vmax_4*fV_PFK*WGD_E*gR_4*F6P/Kf6p_4*ATP/Katp_4*(1+F6P/Kf6p_4+ATP/Katp_4+gR_4*F6P/Kf6p_4*ATP/Katp_4)/((1+F6P/Kf6p_4+ATP/Katp_4+gR_4*F6P/Kf6p_4*ATP/Katp_4)^2+L0_4*((1+Ciatp_4*ATP/Kiatp_4)/(1+ATP/Kiatp_4))^2*((1+Camp_4*AMP/Kamp_4)/(1+AMP/Kamp_4))^2*((1+Cf26_4*F26bP/Kf26_4+Cf16_4*F16bP/Kf16_4)/(1+F26bP/Kf26_4+F16bP/Kf16_4))^2*(1+Catp_4*ATP/Katp_4)^2)
fV_HXT = 1.0 dimensionless; Vmax_1=97.24 mmolepermin; Kglc_1=1.1918 mM; Ki_1=0.91 dimensionless; WGD_E = 1.0 dimensionlessReaction: GLCo => GLCi, Rate Law: Vmax_1*fV_HXT*WGD_E*(GLCo-GLCi)/Kglc_1/(1+(GLCo+GLCi)/Kglc_1+Ki_1*GLCo*GLCi/Kglc_1^2)
k2_6=1.0E7 permin; k1_6=450000.0 perminReaction: DHAP => GAP, Rate Law: cyto*(k1_6*DHAP-k2_6*GAP)
Kglc_2=0.08 mM; Kadp_2=0.23 mM; Vmax_2=236.7 mMpermin; Keq_2=2000.0 dimensionless; fV_HXK = 1.0 dimensionless; Kg6p_2=30.0 mM; WGD_E = 1.0 dimensionless; Katp_2=0.15 mMReaction: GLCi + ATP => G6P + ADP, Rate Law: cyto*WGD_E*fV_HXK*Vmax_2*(GLCi*ATP/(Kglc_2*Katp_2)-G6P*ADP/(Kglc_2*Katp_2*Keq_2))/((1+GLCi/Kglc_2+G6P/Kg6p_2)*(1+ATP/Katp_2+ADP/Kadp_2))
Keq_8=3200.0 dimensionless; Vmax_8=1288.0 mMpermin; Kbpg_8=0.003 mM; WGD_E = 1.0 dimensionless; Kp3g_8=0.53 mM; Kadp_8=0.2 mM; fV_PGK = 1.0 dimensionless; Katp_8=0.3 mMReaction: ADP + BPG => ATP + P3G, Rate Law: cyto*fV_PGK*Vmax_8*WGD_E*(Keq_8*BPG*ADP-P3G*ATP)/(Kp3g_8*Katp_8)/((1+BPG/Kbpg_8+P3G/Kp3g_8)*(1+ADP/Kadp_8+ATP/Katp_8))
Knadh_16=0.023 mM; Kdhap_16=0.4 mM; Kglycerol_16=1.0 mM; Vmax_16=47.11 mMpermin; WGD_E = 1.0 dimensionless; Keq_16=4300.0 dimensionless; Knad_16=0.93 mMReaction: DHAP + NADH => NAD + Glycerol, Rate Law: cyto*Vmax_16*WGD_E*(DHAP/Kdhap_16*NADH/Knadh_16-Glycerol/Kdhap_16*NAD/Knadh_16*1/Keq_16)/((1+DHAP/Kdhap_16+Glycerol/Kglycerol_16)*(1+NADH/Knadh_16+NAD/Knad_16))
Katp_11=1.5 mM; Vmax_11=1000.0 mMpermin; fV_PYK = 1.0 dimensionless; Kadp_11=0.53 mM; Kpyr_11=21.0 mM; WGD_E = 1.0 dimensionless; Kpep_11=0.14 mM; Keq_11=6500.0 dimensionlessReaction: ADP + PEP => ATP + PYR, Rate Law: cyto*Vmax_11*fV_PYK*WGD_E*(PEP*ADP/(Kpep_11*Kadp_11)-PYR*ATP/(Kpep_11*Kadp_11*Keq_11))/((1+PEP/Kpep_11+PYR/Kpyr_11)*(1+ADP/Kadp_11+ATP/Katp_11))
Kigap_5=10.0 mM; Keq_5=0.069 mM; Kgap_5=2.4 mM; Kdhap_5=2.0 mM; WGD_E = 1.0 dimensionless; Vmax_5=94.69 mMpermin; fV_FBA = 1.0 dimensionless; Kf16bp_5=0.3 mMReaction: F16bP => DHAP + GAP, Rate Law: cyto*Vmax_5*fV_FBA*WGD_E*(F16bP/Kf16bp_5-DHAP*GAP/(Kf16bp_5*Keq_5))/(1+F16bP/Kf16bp_5+DHAP/Kdhap_5+GAP/Kgap_5+F16bP*GAP/(Kf16bp_5*Kigap_5)+DHAP*GAP/(Kdhap_5*Kgap_5))
k2_15=100.0 permMpermin; k1_15=45.0 permMperminReaction: ADP => ATP + AMP, Rate Law: cyto*(k1_15*ADP*ADP-k2_15*ATP*AMP)
Ketoh_13=17.0 mM; Knad_13=0.17 mM; Kietoh_13=90.0 mM; Kacald_13=1.11 mM; Vmax_13=209.5 mMpermin; Knadh_13=0.11 mM; Kinad_13=0.92 mM; Keq_13=6.9E-5 dimensionless; WGD_E = 1.0 dimensionless; Kiacald_13=1.1 mM; Kinadh_13=0.031 mMReaction: NAD + EtOH => NADH + AcAld, Rate Law: cyto*Vmax_13*WGD_E*(EtOH*NAD/(Ketoh_13*Kinad_13)-AcAld*NADH/(Ketoh_13*Kinad_13*Keq_13))/(1+NAD/Kinad_13+EtOH*Knad_13/(Kinad_13*Ketoh_13)+AcAld*Knadh_13/(Kinadh_13*Kacald_13)+NADH/Kinadh_13+EtOH*NAD/(Kinad_13*Ketoh_13)+NAD*AcAld*Knadh_13/(Kinad_13*Kinadh_13*Kacald_13)+EtOH*NADH*Knad_13/(Kinad_13*Kinadh_13*Ketoh_13)+AcAld*NADH/(Kacald_13*Kinadh_13)+EtOH*NAD*AcAld/(Kinad_13*Kiacald_13*Ketoh_13)+EtOH*AcAld*NADH/(Kietoh_13*Kinadh_13*Kacald_13))
fV_TDH = 1.0 dimensionless; Knadh_7=0.06 mM; Kgap_7=0.21 mM; Vmaxr_7=6719.0 mMpermin; C_7=1.0 dimensionless; Kbpg_7=0.0098 mM; WGD_E = 1.0 dimensionless; Knad_7=0.09 mM; Vmaxf_7=1152.0 mMperminReaction: GAP + NAD => BPG + NADH, Rate Law: cyto*C_7*(Vmaxf_7*fV_TDH*WGD_E*GAP*NAD/(Kgap_7*Knad_7)-Vmaxr_7*fV_TDH*WGD_E*BPG*NADH/(Kbpg_7*Knadh_7))/((1+GAP/Kgap_7+BPG/Kbpg_7)*(1+NAD/Knad_7+NADH/Knadh_7))
KGLYCOGEN_17=6.0 mMperminReaction: ATP + G6P => ADP + Glycogen, Rate Law: cyto*KGLYCOGEN_17
Katpase_14=39.5 perminReaction: ATP => ADP, Rate Law: cyto*Katpase_14*ATP
Keq_3=0.29 dimensionless; Kf6p_3=0.3 mM; Kg6p_3=1.4 mM; WGD_E = 1.0 dimensionless; Vmax_3=1056.0 mMpermin; fV_PGI = 1.0 dimensionlessReaction: G6P => F6P, Rate Law: cyto*Vmax_3*fV_PGI*WGD_E*(G6P/Kg6p_3-F6P/(Kg6p_3*Keq_3))/(1+G6P/Kg6p_3+F6P/Kf6p_3)
Kp2g_9=0.08 mM; fV_GPM = 1.0 dimensionless; Vmax_9=2585.0 mMpermin; WGD_E = 1.0 dimensionless; Kp3g_9=1.2 mM; Keq_9=0.19 dimensionlessReaction: P3G => P2G, Rate Law: cyto*Vmax_9*fV_GPM*WGD_E*(P3G/Kp3g_9-P2G/(Kp3g_9*Keq_9))/(1+P3G/Kp3g_9+P2G/Kp2g_9)

States:

NameDescription
ATP[ATP; ATP]
Trehalose[alpha,alpha-trehalose; alpha,alpha-Trehalose]
F16bP[beta-D-fructofuranose 1,6-bisphosphate; beta-D-Fructose 1,6-bisphosphate]
AMP[AMP; AMP]
DHAP[dihydroxyacetone phosphate; Glycerone phosphate]
GLCi[D-glucopyranose; D-Glucose]
P2G[2-phospho-D-glyceric acid; 2-Phospho-D-glycerate]
P3G[3-phospho-D-glyceric acid; 3-Phospho-D-glycerate]
Succinate[succinate(2-); Succinate]
GLCo[D-glucopyranose; D-Glucose]
NADH[NADH; NADH]
PYR[pyruvate; Pyruvate]
AcAld[acetaldehyde; Acetaldehyde]
EtOH[ethanol; Ethanol]
BPG[3-phospho-D-glyceroyl dihydrogen phosphate; 3-Phospho-D-glyceroyl phosphate]
F6P[beta-D-fructofuranose 6-phosphate; beta-D-Fructose 6-phosphate]
CO2[carbon dioxide; CO2]
Glycerol[glycerol; Glycerol]
G6P[alpha-D-glucose 6-phosphate; alpha-D-Glucose 6-phosphate]
GAP[D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]
PEP[phosphoenolpyruvate; Phosphoenolpyruvate]
Glycogen[glycogen; Glycogen]
ADP[ADP; ADP]
NAD[NAD(+); NAD+]

Condorelli2001_GuanylateCyclase: MODEL4780441670v0.0.1

This model features the observations of <a href = "http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=pubmed&…

Details

Free nitric oxide (NO) activates soluble guanylate cyclase (sGC), an enzyme, within both pulmonary and vascular smooth muscle. sGC catalyzes the cyclization of guanosine 5'-triphosphate to guanosine 3',5'-cyclic monophosphate (cGMP). Binding rates of NO to the ferrous heme(s) of sGC have been measured in vitro. However, a missing link in our understanding of the control mechanism of sGC by NO is a comprehensive in vivo kinetic analysis. Available literature data suggests that NO dissociation from the heme center of sGC is accelerated by its interaction with one or more cofactors in vivo. We present a working model for sGC activation and NO consumption in vivo. Our model predicts that NO influences the cGMP formation rate over a concentration range of approximately 5-100 nM (apparent Michaelis constant approximately 23 nM), with Hill coefficients between 1.1 and 1.5. The apparent reaction order for NO consumption by sGC is dependent on NO concentration, and varies between 0 and 1.5. Finally, the activation of sGC (half-life approximately 1-2 s) is much more rapid than deactivation (approximately 50 s). We conclude that control of sGC in vivo is most likely ultra-sensitive, and that activation in vivo occurs at lower NO concentrations than previously reported. link: http://identifiers.org/pubmed/11325714

Conradie2010_RPControl_CellCycle: BIOMD0000000265v0.0.1

This model is from the article: Restriction point control of the mammalian cell cycle via the cyclin E/Cdk2:p27 comp…

Details

Numerous top-down kinetic models have been constructed to describe the cell cycle. These models have typically been constructed, validated and analyzed using model species (molecular intermediates and proteins) and phenotypic observations, and therefore do not focus on the individual model processes (reaction steps). We have developed a method to: (a) quantify the importance of each of the reaction steps in a kinetic model for the positioning of a switch point [i.e. the restriction point (RP)]; (b) relate this control of reaction steps to their effects on molecular species, using sensitivity and co-control analysis; and thereby (c) go beyond a correlation towards a causal relationship between molecular species and effects. The method is generic and can be applied to responses of any type, but is most useful for the analysis of dynamic and emergent responses such as switch points in the cell cycle. The strength of the analysis is illustrated for an existing mammalian cell cycle model focusing on the RP [Novak B, Tyson J (2004) J Theor Biol230, 563-579]. The reactions in the model with the highest RP control were those involved in: (a) the interplay between retinoblastoma protein and E2F transcription factor; (b) those synthesizing the delayed response genes and cyclin D/Cdk4 in response to growth signals; (c) the E2F-dependent cyclin E/Cdk2 synthesis reaction; as well as (d) p27 formation reactions. Nine of the 23 intermediates were shown to have a good correlation between their concentration control and RP control. Sensitivity and co-control analysis indicated that the strongest control of the RP is mediated via the cyclin E/Cdk2:p27 complex concentration. Any perturbation of the RP could be related to a change in the concentration of this complex; apparent effects of other molecular species were indirect and always worked through cyclin E/Cdk2:p27, indicating a causal relationship between this complex and the positioning of the RP. link: http://identifiers.org/pubmed/20015233

Parameters:

NameDescription
K23a = 0.005 per hour; K23 = 1.0 per hourReaction: var2 => var3; CYCA, CYCB, Rate Law: (K23a+K23*(CYCA+CYCB))*var2
K10 = 5.0 per hourReaction: CD => P27, Rate Law: K10*CD
K25R = 10.0 per hourReaction: CE => CYCE + P27, Rate Law: K25R*CE
LD = 3.3 dimensionless; LB = 5.0 dimensionless; LE = 5.0 dimensionless; LA = 3.0 dimensionless; K20 = 10.0 per hourReaction: var4 => var1; CYCA, CYCB, CD, CYCD, CYCE, Rate Law: K20*(LA*CYCA+LB*CYCB+LD*(CD+CYCD)+LE*CYCE)*var4
PP1A = NaN dimensionless; K19=20.0 per hour; PP1T = 1.0 dimensionless; K19a=0.0 per hourReaction: var1 => var4; CYCB, CYCA, CYCE, Rate Law: (K19*PP1A+K19a*(PP1T-PP1A))*var1
K22 = 1.0 per hourReaction: var3 => var2, Rate Law: K22*var3
J1=0.1 dimensionless; K1a=0.1 per hour; K1=0.6 per hour; eps = 1.0 dimensionlessReaction: => CYCB, Rate Law: eps*(K1a+K1*CYCB^2/(J1^2*(1+CYCB^2/J1^2)))
J14=0.005 dimensionless; K14=2.5 per hourReaction: CDc20 =>, Rate Law: K14*CDc20/(J14+CDc20)
K33=0.05 per hour; eps = 1.0 dimensionlessReaction: => PPX, Rate Law: eps*K33
K3=140.0 per hour; K3a=7.5 per hour; J3=0.01 dimensionlessReaction: => CDh1; CDc20, Rate Law: (K3a+K3*CDc20)*(1-CDh1)/((1+J3)-CDh1)
k15=0.25 per hour; eps = 1.0 dimensionless; J15=0.1 dimensionlessReaction: => ERG; DRG, Rate Law: eps*k15/(1+DRG^2/J15^2)
k17=10.0 per hour; eps = 1.0 dimensionless; J17=0.3 dimensionless; k17a=0.35 per hourReaction: => DRG; ERG, Rate Law: eps*(k17*DRG^2/(J17^2*(1+DRG^2/J17^2))+k17a*ERG)
K30 = 20.0 per hourReaction: CA => P27; CDc20, Rate Law: K30*CA*CDc20
K25 = 1000.0 per hourReaction: CYCE + P27 => CE, Rate Law: K25*CYCE*P27
k18=10.0 per hourReaction: DRG =>, Rate Law: k18*DRG
K26 = 10000.0 per hourReaction: var2 + var4 => var5, Rate Law: K26*var2*var4
V4 = NaN dimensionless; J4=0.01 dimensionlessReaction: CDh1 => ; CYCA, CYCB, CYCE, Rate Law: V4*CDh1/(J4+CDh1)
K27=0.2 per hour; r31switch = 1.0 dimensionlessReaction: => GM; MASS, Rate Law: K27*MASS*r31switch
V2 = NaN dimensionlessReaction: CYCB => ; CDc20, CDh1, Rate Law: V2*CYCB
eps = 1.0 dimensionless; K7a=0.0 per hour; K7=0.6 per hourReaction: => CYCE; var2, Rate Law: eps*(K7a+K7*var2)
k24=1000.0 per hourReaction: CYCD + P27 => CD, Rate Law: k24*CYCD*P27
V6 = NaN dimensionlessReaction: CE => CYCE; CYCA, CYCB, Rate Law: V6*CE
J13=0.005 dimensionless; K13=5.0 per hourReaction: => CDc20; CDc20T, IEP, Rate Law: K13*((-CDc20)+CDc20T)*IEP/((J13-CDc20)+CDc20T)
J31=0.01 dimensionless; K31=0.7 per hourReaction: => IEP; CYCB, Rate Law: K31*CYCB*(1-IEP)/((1+J31)-IEP)
K29=0.05 per hour; eps = 1.0 dimensionlessReaction: => CYCA; MASS, var2, Rate Law: eps*K29*MASS*var2
K11=1.5 per hour; eps = 1.0 dimensionless; K11a=0.0 per hourReaction: => CDc20T; CYCB, Rate Law: eps*(K11a+K11*CYCB)
V8 = NaN dimensionlessReaction: CE => P27; CYCB, CYCA, CYCE, Rate Law: V8*CE
J32=0.01 dimensionless; K32=1.8 per hourReaction: IEP => ; PPX, Rate Law: K32*IEP*PPX/(J32+IEP)
K28=0.2 per hourReaction: GM =>, Rate Law: K28*GM
K26R = 200.0 per hourReaction: var5 => var2 + var4, Rate Law: K26R*var5
eps = 1.0 dimensionless; K5=20.0 per hourReaction: => P27, Rate Law: eps*K5
K34=0.05 per hourReaction: PPX =>, Rate Law: K34*PPX
MU=0.061 per hour; eps = 1.0 dimensionlessReaction: => MASS; GM, Rate Law: eps*MU*GM
K12 = 1.5 per hourReaction: CDc20T =>, Rate Law: K12*CDc20T
k24r=10.0 per hourReaction: CD => CYCD + P27, Rate Law: k24r*CD
k16=0.25 per hourReaction: ERG =>, Rate Law: k16*ERG
K9=2.5 per hour; eps = 1.0 dimensionlessReaction: => CYCD; DRG, Rate Law: eps*K9*DRG

States:

NameDescription
var1[Retinoblastoma-associated protein]
ERGearly-response genes
CYCB[Cyclin-dependent kinase 2; IPR015454]
var2[Transcription factor E2F1]
var4[Retinoblastoma-associated protein]
GMgeneral machinery for protein synthesis
P27[Cyclin-dependent kinase inhibitor 1B]
var3[Transcription factor E2F1]
DRGdelayed-response genes
CDc20[APC/C activator protein CDC20]
var5[Transcription factor E2F1; Retinoblastoma-associated protein]
MASS[cell growth]
IEP[anaphase-promoting complex]
CYCD[Cyclin-dependent kinase 2; IPR015451]
CA[Cyclin-dependent kinase inhibitor 1B; Cyclin-dependent kinase 2; IPR015453]
CE[Cyclin-dependent kinase inhibitor 1B; G1/S-specific cyclin-E1; Cyclin-dependent kinase 2]
CYCE[G1/S-specific cyclin-E1; Cyclin-dependent kinase 2]
CDc20T[APC/C activator protein CDC20]
CDh1[Cadherin-1]
PPX[Exopolyphosphatase]
CYCA[Cyclin-dependent kinase 2; IPR015453]
var6[Transcription factor E2F1; Retinoblastoma-associated protein]
CD[Cyclin-dependent kinase inhibitor 1B; Cyclin-dependent kinase 2; IPR015451]

Cookson2011_EnzymaticQueueingCoupling: BIOMD0000000405v0.0.1

This model is from the article: Queueing up for enzymatic processing: correlated signaling through coupled degradati…

Details

High-throughput technologies have led to the generation of complex wiring diagrams as a post-sequencing paradigm for depicting the interactions between vast and diverse cellular species. While these diagrams are useful for analyzing biological systems on a large scale, a detailed understanding of the molecular mechanisms that underlie the observed network connections is critical for the further development of systems and synthetic biology. Here, we use queueing theory to investigate how 'waiting lines' can lead to correlations between protein 'customers' that are coupled solely through a downstream set of enzymatic 'servers'. Using the E. coli ClpXP degradation machine as a model processing system, we observe significant cross-talk between two networks that are indirectly coupled through a common set of processors. We further illustrate the implications of enzymatic queueing using a synthetic biology application, in which two independent synthetic networks demonstrate synchronized behavior when common ClpXP machinery is overburdened. Our results demonstrate that such post-translational processes can lead to dynamic connections in cellular networks and may provide a mechanistic understanding of existing but currently inexplicable links. link: http://identifiers.org/pubmed/22186735

Parameters:

NameDescription
parameter_3 = 10.0Reaction: species_4 => species_5, Rate Law: compartment_1*parameter_3*species_4
parameter_5 = 0.03465735902799Reaction: species_1 =>, Rate Law: compartment_1*parameter_5*species_1
parameter_4 = 1000.0Reaction: species_1 + species_5 => species_3, Rate Law: compartment_1*parameter_4*species_1*species_5
parameter_1 = 500.0Reaction: => species_1, Rate Law: compartment_1*parameter_1
parameter_2 = 500.0Reaction: => species_2, Rate Law: compartment_1*parameter_2

States:

NameDescription
species 2[protein]
species 3E1
species 1[protein]
species 4E2
species 5E

Cooling2007_IP3transients_CardiacMyocyte: BIOMD0000000400v0.0.1

This a model from the article: Modeling hypertrophic IP3 transients in the cardiac myocyte. Cooling M, Hunter P, Cr…

Details

Cardiac hypertrophy is a known risk factor for heart disease, and at the cellular level is caused by a complex interaction of signal transduction pathways. The IP3-calcineurin pathway plays an important role in stimulating the transcription factor NFAT which binds to DNA cooperatively with other hypertrophic transcription factors. Using available kinetic data, we construct a mathematical model of the IP3 signal production system after stimulation by a hypertrophic alpha-adrenergic agonist (endothelin-1) in the mouse atrial cardiac myocyte. We use a global sensitivity analysis to identify key controlling parameters with respect to the resultant IP3 transient, including the phosphorylation of cell-membrane receptors, the ligand strength and binding kinetics to precoupled (with G(alpha)GDP) receptor, and the kinetics associated with precoupling the receptors. We show that the kinetics associated with the receptor system contribute to the behavior of the system to a great extent, with precoupled receptors driving the response to extracellular ligand. Finally, by reparameterizing for a second hypertrophic alpha-adrenergic agonist, angiotensin-II, we show that differences in key receptor kinetic and membrane density parameters are sufficient to explain different observed IP3 transients in essentially the same pathway. link: http://identifiers.org/pubmed/17693463

Parameters:

NameDescription
J13 = NaN; J9 = NaN; J11 = NaNReaction: Pg = J9-(J11+J13), Rate Law: J9-(J11+J13)
J7 = NaN; J10 = NaN; J9 = NaN; J6 = NaNReaction: Gt = J6-(J7+J9+J10), Rate Law: J6-(J7+J9+J10)
J5 = NaNReaction: Rlgp = J5, Rate Law: J5
J7 = NaN; J13 = NaN; J2 = NaN; J3 = NaN; J12 = NaNReaction: Gd = (J7+J13+J12)-(J2+J3), Rate Law: (J7+J13+J12)-(J2+J3)
J10 = NaN; J11 = NaN; J12 = NaNReaction: Pcg = (J10+J11)-J12, Rate Law: (J10+J11)-J12
J10 = NaN; J8 = NaN; J12 = NaNReaction: Pc = (J8+J12)-J10, Rate Law: (J8+J12)-J10
J13 = NaN; J9 = NaN; J8 = NaNReaction: P = J13-(J9+J8), Rate Law: J13-(J9+J8)
Cpc = NaN; J15 = NaN; J14 = NaN; J16 = NaNReaction: IP3 = Cpc*(J14+J15)-J16, Rate Law: Cpc*(J14+J15)-J16
J4 = NaN; J5 = NaN; J6 = NaN; J3 = NaNReaction: Rlg = ((J3-J5)+J4)-J6, Rate Law: ((J3-J5)+J4)-J6
Cpc = NaN; J11 = NaN; J8 = NaNReaction: Ca = Cpc*(-1)*(J8+J11), Rate Law: Cpc*(-1)*(J8+J11)
J4 = NaN; J2 = NaNReaction: Rg = J2-J4, Rate Law: J2-J4
J1 = NaN; J2 = NaNReaction: R = (-1)*(J1+J2), Rate Law: (-1)*(J1+J2)
J1 = NaN; J6 = NaN; J3 = NaNReaction: Rl = (J1+J6)-J3, Rate Law: (J1+J6)-J3

States:

NameDescription
Rl[G-protein coupled receptor 12; CCO:F0001633]
P[1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase beta-1]
Pcg[calcium(2+); GTP; 1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase beta-1; Guanine nucleotide-binding protein subunit alpha-12]
Gd[GDP; Guanine nucleotide-binding protein subunit alpha-12]
Rlgp[GDP; G-protein coupled receptor 12; Guanine nucleotide-binding protein subunit alpha-12; CCO:F0001633; urn:miriam:mod:MOD%3A00696]
Pc[calcium(2+); 1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase beta-1]
IP3[Inositol-trisphosphate 3-kinase A]
Gt[GTP; Guanine nucleotide-binding protein subunit alpha-12]
Pg[GTP; 1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase beta-1; Guanine nucleotide-binding protein subunit alpha-12]
Ca[calcium(2+)]
R[G-protein coupled receptor 12]
Rlg[GDP; G-protein coupled receptor 12; Guanine nucleotide-binding protein subunit alpha-12; CCO:F0001633]
Rg[GDP; G-protein coupled receptor 12; Guanine nucleotide-binding protein subunit alpha-12]

Cooper2015 - Modeling the effects of systemic mediators on the inflammatory phase of wound healing: BIOMD0000000855v0.0.1

This is an ordinary differential equation-based mathematical model describing the inflammatory phase of the wound healin…

Details

The normal wound healing response is characterized by a progression from clot formation, to an inflammatory phase, to a repair phase, and finally, to remodeling. In many chronic wounds there is an extended inflammatory phase that stops this progression. In order to understand the inflammatory phase in more detail, we developed an ordinary differential equation model that accounts for two systemic mediators that are known to modulate this phase, estrogen (a protective hormone during wound healing) and cortisol (a hormone elevated after trauma that slows healing). This model describes the interactions in the wound between wound debris, pathogens, neutrophils and macrophages and the modulation of these interactions by estrogen and cortisol. A collection of parameter sets, which qualitatively match published data on the dynamics of wound healing, was chosen using Latin Hypercube Sampling. This collection of parameter sets represents normal healing in the population as a whole better than one single parameter set. Including the effects of estrogen and cortisol is a necessary step to creating a patient specific model that accounts for gender and trauma. Utilization of math modeling techniques to better understand the wound healing inflammatory phase could lead to new therapeutic strategies for the treatment of chronic wounds. This inflammatory phase model will later become the inflammatory subsystem of our full wound healing model, which includes fibroblast activity, collagen accumulation and remodeling. link: http://identifiers.org/pubmed/25446708

Parameters:

NameDescription
mupt = 0.37Reaction: Pt =>, Rate Law: compartment*mupt*Pt
mun = 1.02Reaction: N => Pt, Rate Law: compartment*mun*N
fi2 = 0.0; kem = 4.97; E = 0.0; kpm = 34.8Reaction: P =>, Rate Law: compartment*kpm*P*fi2*(1+kem*E)
pinf = 20.0; kpg = 14.4Reaction: => P, Rate Law: compartment*kpg*P*(1-P/pinf)
kpb = 14.4; mub = 0.048; kbp = 0.2; sb = 0.12Reaction: P =>, Rate Law: compartment*kpb*sb*P/(mub+kbp*P)
kpn = 35.03; ken = 5.37; E = 0.0Reaction: P => ; N, Rate Law: compartment*kpn*P*N*(1+ken*E)
ken = 5.37; kptn = 2.03Reaction: Pt => ; N, Rate Law: compartment*kptn*Pt*N*(1+ken*N)
mum = 0.5Reaction: M =>, Rate Law: compartment*mum*M
smr = 0.17; mumr = 0.54; R1 = 85.91Reaction: => M, Rate Law: compartment*smr*R1/(mumr+R1)
kptm = 3.16; fi2 = 0.0; kem = 4.97; E = 0.0Reaction: Pt =>, Rate Law: compartment*kptm*Pt*fi2*(1+kem*E)
fi2 = 0.0; knm = 6.42Reaction: N =>, Rate Law: compartment*knm*N*fi2
fEN = 0.265979268957992Reaction: => N, Rate Law: compartment*fEN

States:

NameDescription
M[macrophage]
P[C80324]
N[CL:0000775]
Pt[C120869]

Corbat2021 - Apoptotic Reaction Model.models: MODEL2105210001v0.0.1

Apoptotic Reaction Model is an ordinary differential equation model describing every species in the apoptotic cascade an…

Details

Apoptosis, a form of programmed cell death central to all multicellular organisms, plays a key role during organism development and is often misregulated in cancer. Devising a single model applicable to distinct stimuli and conditions has been limited by lack of robust observables. Indeed, previous numerical models have been tailored to fit experimental datasets in restricted scenarios, failing to predict response to different stimuli. We quantified the activity of three caspases simultaneously upon intrinsic or extrinsic stimulation to assemble a comprehensive dataset. We measured and modeled the time between maximum activity of intrinsic, extrinsic and effector caspases, a robust observable of network dynamics, to create the first integrated Apoptotic Reaction Model (ARM). Observing how effector caspases reach maximum activity first irrespective of stimuli used, led us to identify and incorporate a missing feedback into a successful model for extrinsic stimulation. By simulating different recently performed experiments, we corroborated that ARM adequately describes them. This integrated model provides further insight into the indispensable feedback from effector caspase to initiator caspases. link: http://identifiers.org/doi/10.1101/2021.05.21.444824

Core Metabolism Arabidopsis thaliana: MODEL1801090001v0.0.1

Arabidopsis thaliana is a well-established model system for the analysis of the basic physiological and metabolic pathwa…

Details

Motivation:Arabidopsis thaliana is a well-established model system for the analysis of the basic physiological and metabolic pathways of plants. Nevertheless, the system is not yet fully understood, although many mechanisms are described, and information for many processes exists. However, the combination and interpretation of the large amount of biological data remain a big challenge, not only because data sets for metabolic paths are still incomplete. Moreover, they are often inconsistent, because they are coming from different experiments of various scales, regarding, for example, accuracy and/or significance. Here, theoretical modeling is powerful to formulate hypotheses for pathways and the dynamics of the metabolism, even if the biological data are incomplete. To develop reliable mathematical models they have to be proven for consistency. This is still a challenging task because many verification techniques fail already for middle-sized models. Consequently, new methods, like decomposition methods or reduction approaches, are developed to circumvent this problem. Methods: We present a new semi-quantitative mathematical model of the metabolism of Arabidopsis thaliana. We used the Petri net formalism to express the complex reaction system in a mathematically unique manner. To verify the model for correctness and consistency we applied concepts of network decomposition and network reduction such as transition invariants, common transition pairs, and invariant transition pairs. Results: We formulated the core metabolism of Arabidopsis thaliana based on recent knowledge from literature, including the Calvin cycle, glycolysis and citric acid cycle, glyoxylate cycle, urea cycle, sucrose synthesis, and the starch metabolism. By applying network decomposition and reduction techniques at steady-state conditions, we suggest a straightforward mathematical modeling process. We demonstrate that potential steady-state pathways exist, which provide the fixed carbon to nearly all parts of the network, especially to the citric acid cycle. There is a close cooperation of important metabolic pathways, e.g., the de novo synthesis of uridine-5-monophosphate, the γ-aminobutyric acid shunt, and the urea cycle. The presented approach extends the established methods for a feasible interpretation of biological network models, in particular of large and complex models. link: http://identifiers.org/pubmed/28713420

Corrias2007_GastricSMCellularActivation: MODEL0913145131v0.0.1

This a model from the article: A quantitative model of gastric smooth muscle cellular activation. Corrias A, Buist M…

Details

A physiologically realistic quantitative description of the electrical behavior of a gastric smooth muscle (SM) cell is presented. The model describes the response of a SM cell when activated by an electrical stimulus coming from the network of interstitial cells of Cajal (ICC) and is mediated by the activation of different ion channels species in the plasma membrane. The conductances (predominantly Ca2+ and K+) that are believed to substantially contribute to the membrane potential fluctuations during slow wave activity have been included in the model. A phenomenological description of intracellular Ca2+ dynamics has also been included because of its primary importance in regulating a number of cellular processes. In terms of shape, duration, and amplitude, the resulting simulated smooth muscle depolarizations (SMDs) are in good agreement with experimentally recordings from mammalian gastric SM in control and altered conditions. This model has also been designed to be suitable for incorporation into large scale multicellular simulations. link: http://identifiers.org/pubmed/17486452

Corrias2008_GastricSlowWaveActivity: MODEL0913095435v0.0.1

This a model from the article: Quantitative cellular description of gastric slow wave activity. Corrias A, Buist ML.…

Details

Interstitial cells of Cajal (ICC) are responsible for the spontaneous and omnipresent electrical activity in the stomach. A quantitative description of the intracellular processes whose coordinated activity is believed to generate electrical slow waves has been developed and is presented here. In line with recent experimental evidence, the model describes how the interplay between the mitochondria and the endoplasmic reticulum in cycling intracellular Ca(2+) provides the primary regulatory signal for the initiation of the slow wave. The major ion channels that have been identified as influencing slow wave activity have been modeled according to data obtained from isolated ICC. The model has been validated by comparing the simulated profile of the slow waves with experimental recordings and shows good correspondence in terms of frequency, amplitude, and shape in both control and pharmacologically altered conditions. link: http://identifiers.org/pubmed/18276830

Cortes2019 - Optimality of the spontaneous prophage induction rate.: BIOMD0000000884v0.0.1

Optimality of the spontaneous prophage induction rate. Cortes MG1, Krog J2, Balázsi G3. 1 Department of Applied Mathema…

Details

Lysogens are bacterial cells that have survived after genomically incorporating the DNA of temperate bacteriophages infecting them. If an infection results in lysogeny, the lysogen continues to grow and divide normally, seemingly unaffected by the integrated viral genome known as a prophage. However, the prophage can still have an impact on the host's phenotype and overall fitness in certain environments. Additionally, the prophage within the lysogen can activate the lytic pathway via spontaneous prophage induction (SPI), killing the lysogen and releasing new progeny phages. These new phages can then lyse or lysogenize other susceptible nonlysogens, thereby impacting the competition between lysogens and nonlysogens. In a scenario with differing growth rates, it is not clear whether SPI would be beneficial or detrimental to the lysogens since it kills the host cell but also attacks nonlysogenic competitors, either lysing or lysogenizing them. Here we study the evolutionary dynamics of a mixture of lysogens and nonlysogens and derive general conditions on SPI rates for lysogens to displace nonlysogens. We show that there exists an optimal SPI rate for bacteriophage λ and explain why it is so low. We also investigate the impact of stochasticity and conclude that even at low cell numbers SPI can still provide an advantage to the lysogens. These results corroborate recent experimental studies showing that lower SPI rates are advantageous for phage-phage competition, and establish theoretical bounds on the SPI rate in terms of ecological and environmental variables associated with lysogens having a competitive advantage over their nonlysogenic counterparts. link: http://identifiers.org/pubmed/31525321

Parameters:

NameDescription
g = 1.0Reaction: => U, Rate Law: compartment*g*U
phi = 0.999899; alpha = 1.0E-7Reaction: U => ; V, Rate Law: compartment*(alpha*U*V+phi*U)
gamma = 0.001; alpha = 1.0E-7Reaction: V => ; L, Rate Law: compartment*(gamma*V+alpha*V*L)
r = 0.99; p = 0.3; alpha = 1.0E-7Reaction: => L; U, V, Rate Law: compartment*(r*L+p*alpha*U*V)
delta = 1.0E-4; b = 150.0; p = 0.3; alpha = 1.0E-7Reaction: => V; U, L, Rate Law: compartment*((1-p)*b*alpha*U*V+b*delta*L)
delta = 1.0E-4; phi = 0.999899Reaction: L =>, Rate Law: compartment*(delta*L+phi*L)

States:

NameDescription
UU
V[C14283]
L[C14283]

Costa2014 - Computational Model of L. lactis Metabolism: BIOMD0000000572v0.0.1

Costa2014 - Computational Model of L. lactis MetabolismThis model is described in the article: [An extended dynamic mod…

Details

Biomedical research and biotechnological production are greatly benefiting from the results provided by the development of dynamic models of microbial metabolism. Although several kinetic models of Lactococcus lactis (a Lactic Acid Bacterium (LAB) commonly used in the dairy industry) have been developed so far, most of them are simplified and focus only on specific metabolic pathways. Therefore, the application of mathematical models in the design of an engineering strategy for the production of industrially important products by L. lactis has been very limited. In this work, we extend the existing kinetic model of L. lactis central metabolism to include industrially relevant production pathways such as mannitol and 2,3-butanediol. In this way, we expect to study the dynamics of metabolite production and make predictive simulations in L. lactis. We used a system of ordinary differential equations (ODEs) with approximate Michaelis-Menten-like kinetics for each reaction, where the parameters were estimated from multivariate time-series metabolite concentrations obtained by our team through in vivo Nuclear Magnetic Resonance (NMR). The results show that the model captures observed transient dynamics when validated under a wide range of experimental conditions. Furthermore, we analyzed the model using global perturbations, which corroborate experimental evidence about metabolic responses upon enzymatic changes. These include that mannitol production is very sensitive to lactate dehydrogenase (LDH) in the wild type (W.T.) strain, and to mannitol phosphoenolpyruvate: a phosphotransferase system (PTS(Mtl)) in a LDH mutant strain. LDH reduction has also a positive control on 2,3-butanediol levels. Furthermore, it was found that overproduction of mannitol-1-phosphate dehydrogenase (MPD) in a LDH/PTS(Mtl) deficient strain can increase the mannitol levels. The results show that this model has prediction capability over new experimental conditions and offers promising possibilities to elucidate the effect of alterations in the main metabolism of L. lactis, with application in strain optimization. link: http://identifiers.org/pubmed/24413179

Parameters:

NameDescription
kiPint_Ptransport=0.561093; kmADP_Ptransport=0.192278; Vmax_Ptransport=3.59588; kmATP_Ptransport=0.523288; kmPint_Ptransport=0.30336; kmPext_Ptransport=0.749898Reaction: Pext + ATP => Pint + ADP; Pint, ADP, ATP, Pext, Pint, Rate Law: kiPint_Ptransport/(Pint+kiPint_Ptransport)*Vmax_Ptransport*ATP/kmATP_Ptransport*Pext/kmPext_Ptransport/(((1+Pext/kmPext_Ptransport)*(1+ATP/kmATP_Ptransport)+(1+Pint/kmPint_Ptransport+(Pint/kmPint_Ptransport)^2)*(1+ADP/kmADP_Ptransport))-1)
kmG6P_PTS_Glc=0.284871; kmPEP_PTS_Glc=0.305604; Vmax_PTS_Glc=3.71082; kmGlucose_PTS_Glc=0.0485045; kmPYR_PTS_Glc=1.95993; kaPint_PTS_Glc=0.070909; kiFBP_PTS_Glc=1.16937Reaction: Glucose + PEP => G6P + PYR; FBP, Pint, FBP, G6P, Glucose, PEP, PYR, Pint, Rate Law: Pint/(Pint+kaPint_PTS_Glc)*kiFBP_PTS_Glc/(FBP+kiFBP_PTS_Glc)*Vmax_PTS_Glc*Glucose/kmGlucose_PTS_Glc*PEP/kmPEP_PTS_Glc/(((1+Glucose/kmGlucose_PTS_Glc)*(1+PEP/kmPEP_PTS_Glc)+(1+G6P/kmG6P_PTS_Glc)*(1+PYR/kmPYR_PTS_Glc))-1)
kmAcetoin_Acetoin_transp=1.89255; kmAcetoin_Ext_Acetoin_transp=7.05248; Vmax_Acetoin_transp=1.60066Reaction: Acetoin => Acetoin_Ext; Acetoin, Acetoin_Ext, Rate Law: Vmax_Acetoin_transp*Acetoin/kmAcetoin_Acetoin_transp/(1+Acetoin/kmAcetoin_Acetoin_transp+Acetoin_Ext/kmAcetoin_Ext_Acetoin_transp)
kmNAD_MPD=0.373149; kmMannitol1Phoshate_MPD=0.0891203; kmF6P_MPD=0.321372; Keq_MPD=200.0; kiF6P_MPD=22.0284; kmNADH_MPD=0.0303446; Vmax_MPD=1.32695Reaction: F6P + NADH => Mannitol1Phosphate + NAD; F6P, F6P, Mannitol1Phosphate, NAD, NADH, Rate Law: compartment_1*kiF6P_MPD/(F6P+kiF6P_MPD)*(Vmax_MPD*F6P/kmF6P_MPD*NADH/kmNADH_MPD-Vmax_MPD/Keq_MPD*Mannitol1Phosphate/kmF6P_MPD*NAD/kmNADH_MPD)/(((1+F6P/kmF6P_MPD)*(1+NADH/kmNADH_MPD)+(1+Mannitol1Phosphate/kmMannitol1Phoshate_MPD)*(1+NAD/kmNAD_MPD))-1)
kmG3P_FBA=10.1058; Keq_FBA=0.056; Vmax_FBA=56.1311; kmFBP_FBA=0.300745Reaction: FBP => G3P; FBP, G3P, Rate Law: compartment_1*(Vmax_FBA*FBP/kmFBP_FBA-Vmax_FBA/Keq_FBA*G3P^2/kmFBP_FBA)/(1+FBP/kmFBP_FBA+G3P/kmG3P_FBA+(G3P/kmG3P_FBA)^2)
kmF6P_PFK=1.01973; kmATP_PFK=0.0616607; kmADP_PFK=10.7357; kmFBP_PFK=86.8048; Vmax_PFK=18.3577Reaction: F6P + ATP => FBP + ADP; ADP, ATP, F6P, FBP, Rate Law: compartment_1*Vmax_PFK*F6P/kmF6P_PFK*ATP/kmATP_PFK/(((1+F6P/kmF6P_PFK)*(1+ATP/kmATP_PFK)+(1+FBP/kmFBP_PFK)*(1+ADP/kmADP_PFK))-1)
Vmax_PYK=2.22404; kmPYR_PYK=96.4227; kiPint_PYK=3.70071; nPYK=3.0; kaFBP_PYK=0.0388651; kmADP_PYK=3.07711; kmATP_PYK=29.6028; kmPEP_PYK=0.330583Reaction: PEP + ADP => PYR + ATP; FBP, Pint, ADP, ATP, FBP, PEP, PYR, Pint, Rate Law: compartment_1*FBP/(FBP+kaFBP_PYK)*kiPint_PYK^nPYK/(Pint^nPYK+kiPint_PYK^nPYK)*Vmax_PYK*ADP/kmADP_PYK*PEP/kmPEP_PYK/(((1+ADP/kmADP_PYK)*(1+PEP/kmPEP_PYK)+(1+ATP/kmATP_PYK)*(1+PYR/kmPYR_PYK))-1)
kmF6P_FBPase=1.90796; kmFBP_FBPase=0.412307; Vmax_FBPase=0.0970486; kmPint_FBPase=0.0109675Reaction: FBP => F6P + Pint; F6P, FBP, Pint, Rate Law: compartment_1*Vmax_FBPase*FBP/kmFBP_FBPase/(FBP/kmFBP_FBPase+(1+F6P/kmF6P_FBPase)*(1+Pint/kmPint_FBPase))
kmATP_ENO=0.748238; Keq_ENO=27.55; kmADP_ENO=0.413195; Vmax_ENO=29.132; kmBPG_ENO=0.0241572; kmPEP_ENO=1.38899Reaction: BPG + ADP => PEP + ATP; ADP, ATP, BPG, PEP, Rate Law: compartment_1*(Vmax_ENO*BPG/kmBPG_ENO*ADP/kmADP_ENO-Vmax_ENO/Keq_ENO*PEP/kmBPG_ENO*ATP/kmADP_ENO)/(((1+BPG/kmBPG_ENO)*(1+ADP/kmADP_ENO)+(1+PEP/kmPEP_ENO)*(1+ATP/kmATP_ENO))-1)
kmPYR_ALS=0.262819; Keq_ALS=900000.0; Vmax_ALS=0.354581; kmAcetoin_ALS=0.0495418Reaction: PYR => Acetoin; Acetoin, PYR, Rate Law: compartment_1*(Vmax_ALS*(PYR/kmPYR_ALS)^2-Vmax_ALS/Keq_ALS*Acetoin/kmPYR_ALS)/((1+PYR/kmPYR_ALS+(PYR/kmPYR_ALS)^2+1+Acetoin/kmAcetoin_ALS)-1)
Vmax_BDH=2.28578; kmNAD_BDH=1.29567; kmButanediol_BDH=1.80684; Keq_BDH=1400.0; kmNADH_BDH=3.54858; kmAcetoin_BDH=5.62373Reaction: Acetoin + NADH => Butanediol + NAD; Acetoin, Butanediol, NAD, NADH, Rate Law: (Vmax_BDH*Acetoin/kmAcetoin_BDH*NADH/kmNADH_BDH-Vmax_BDH/Keq_BDH*Butanediol/kmAcetoin_BDH*NAD/kmNADH_BDH)/(((1+Acetoin/kmAcetoin_BDH)*(1+NADH/kmNADH_BDH)+(1+Butanediol/kmButanediol_BDH)*(1+NAD/kmNAD_BDH))-1)
kmPYR_PTS_Man=0.344134; Vmax_PTS_Man=4.44903; kmMannitol1Phosphate_PTS_Man=0.362571; kmPEP_PTS_Man=2.20816; kmMannitolExt_PTS_Man=0.0127321Reaction: Mannitol_Ext + PEP => Mannitol1Phosphate + PYR; Mannitol1Phosphate, Mannitol_Ext, PEP, PYR, Rate Law: Vmax_PTS_Man*Mannitol_Ext/kmMannitolExt_PTS_Man*PEP/kmPEP_PTS_Man/(((1+Mannitol_Ext/kmMannitolExt_PTS_Man)*(1+PEP/kmPEP_PTS_Man)+(1+Mannitol1Phosphate/kmMannitol1Phosphate_PTS_Man)*(1+PYR/kmPYR_PTS_Man))-1)
Keq_PFL=650.0; kmPYR_PFL=5.77662; kiG3P_PFL=0.511288; kmAcetCoA_PFL=7.34319; KmCoA_PFL=0.124066; kmFormate_PFL=54.2693; Vmax_PFL=0.00230934Reaction: PYR + CoA => AcetCoA + Formate; G3P, AcetCoA, CoA, Formate, G3P, PYR, Rate Law: kiG3P_PFL/(G3P+kiG3P_PFL)*(Vmax_PFL*PYR/kmPYR_PFL*CoA/KmCoA_PFL-Vmax_PFL/Keq_PFL*AcetCoA/kmPYR_PFL*Formate/KmCoA_PFL)/(((1+PYR/kmPYR_PFL)*(1+CoA/KmCoA_PFL)+(1+AcetCoA/kmAcetCoA_PFL)*(1+Formate/kmFormate_PFL))-1)
kaFBP_LDH=0.0184011; Vmax_LDH=566.598; kmNADH_LDH=0.144443; kiPint_LDH=0.0676829; kmPYR_LDH=0.01; kmLactate_LDH=94.1203; kmNAD_LDH=3.08447Reaction: PYR + NADH => Lactate + NAD; FBP, Pint, FBP, Lactate, NAD, NADH, PYR, Pint, Rate Law: FBP/(FBP+kaFBP_LDH)*kiPint_LDH/(Pint+kiPint_LDH)*Vmax_LDH*PYR/kmPYR_LDH*NADH/kmNADH_LDH/(((1+PYR/kmPYR_LDH)*(1+NADH/kmNADH_LDH)+(1+Lactate/kmLactate_LDH)*(1+NAD/kmNAD_LDH))-1)
kmNADH_AE=0.43127; kmAcetCoA_AE=7.38021; kmNAD_AE=1.31442; kiATP_AE=6.28119; kmCoA_AE=0.091813; kmEthanol_AE=2.28106; Vmax_AE=2.11844Reaction: AcetCoA + NADH => Ethanol + NAD + CoA; ATP, ATP, AcetCoA, CoA, Ethanol, NAD, NADH, Rate Law: kiATP_AE/(ATP+kiATP_AE)*Vmax_AE*AcetCoA/kmAcetCoA_AE*(NADH/kmNADH_AE)^2/(((1+NADH/kmNADH_AE+(NADH/kmNADH_AE)^2)*(1+AcetCoA/kmAcetCoA_AE)+(1+Ethanol/kmEthanol_AE)*(1+CoA/kmCoA_AE)*(1+NAD/kmNAD_AE+(NAD/kmNAD_AE)^2))-1)
Vmax_ACK=3.83918; kmADP_ACK=1.17242; kmATP_ACK=14.1556; kmAcetate_ACK=0.552221; kmCoA_ACK=0.173388; kmAcetCoA_ACK=0.55824Reaction: AcetCoA + ADP => Acetate + ATP + CoA; ADP, ATP, AcetCoA, Acetate, CoA, Rate Law: Vmax_ACK*AcetCoA/kmAcetCoA_ACK*ADP/kmADP_ACK/(((1+AcetCoA/kmAcetCoA_ACK)*(1+ADP/kmADP_ACK)+(1+Acetate/kmAcetate_ACK)*(1+ATP/kmATP_ACK)*(1+CoA/kmCoA_ACK))-1)
kmMannitol_Mannitol_transp=0.0223502; kmMannitol_Ext_Mannitol_transp=0.940662; Vmax_Mannitol_transp=1.62459Reaction: Mannitol => Mannitol_Ext; Mannitol, Mannitol_Ext, Rate Law: Vmax_Mannitol_transp*Mannitol/kmMannitol_Mannitol_transp/(1+Mannitol/kmMannitol_Mannitol_transp+Mannitol_Ext/kmMannitol_Ext_Mannitol_transp)
kmMannitol1Phosphate_MP=3.51571; kmMannitol_MP=0.238849; Vmax_MP=3.48563Reaction: Mannitol1Phosphate => Mannitol; Mannitol, Mannitol1Phosphate, Rate Law: compartment_1*Vmax_MP*Mannitol1Phosphate/kmMannitol1Phosphate_MP/((1+Mannitol1Phosphate/kmMannitol1Phosphate_MP+1+Mannitol/kmMannitol_MP)-1)
kmF6P_PGI=3.13894; Keq_PGI=0.43; Vmax_PGI=21.681; kmG6P_PGI=0.199409Reaction: G6P => F6P; F6P, G6P, Rate Law: compartment_1*(Vmax_PGI*G6P/kmG6P_PGI-Vmax_PGI/Keq_PGI*F6P/kmG6P_PGI)/(1+G6P/kmG6P_PGI+F6P/kmF6P_PGI)
kmATP_ATPase=4.34159; Vmax_ATPase=3.2901; nATPase=3.0Reaction: ATP => ADP + Pint; ATP, Rate Law: compartment_1*Vmax_ATPase*(ATP/kmATP_ATPase)^nATPase/((ATP/kmATP_ATPase)^nATPase+1)
kmPint_GAPDH=6.75302; kmNADH_GAPDH=0.643019; kmBPG_GAPDH=0.0481603; Vmax_GAPDH=30.0058; kmG3P_GAPDH=0.181788; Keq_GAPDH=7.0E-4; kmNAD_GAPDH=0.0477445Reaction: G3P + Pint + NAD => BPG + NADH; BPG, G3P, NAD, NADH, Pint, Rate Law: compartment_1*(Vmax_GAPDH*G3P/kmG3P_GAPDH*NAD/kmNAD_GAPDH*Pint/kmPint_GAPDH-Vmax_GAPDH/Keq_GAPDH*BPG/kmG3P_GAPDH*NADH/kmNAD_GAPDH*1/kmPint_GAPDH)/(((1+G3P/kmG3P_GAPDH)*(1+Pint/kmPint_GAPDH)*(1+NAD/kmNAD_GAPDH)+(1+BPG/kmBPG_GAPDH)*(1+NADH/kmNADH_GAPDH))-1)

States:

NameDescription
Glucose[glucose]
Acetoin Ext[acetoin]
ATP[ATP]
FBP[keto-D-fructose 1,6-bisphosphate]
Acetoin[acetoin]
Mannitol Ext[mannitol]
Formate[formate]
AcetCoA[acetyl-CoA]
Mannitol[mannitol]
CoA[coenzyme A]
NADH[NADH]
PYR[pyruvate]
Ethanol[ethanol]
Mannitol1Phosphate[D-mannitol 1-phosphate]
Pext[phosphate(3-)]
BPG[3-phospho-D-glyceroyl dihydrogen phosphate]
F6P[keto-D-fructose 6-phosphate]
Pint[phosphate(3-)]
G6P[alpha-D-glucose 6-phosphate]
Acetate[acetate]
Lactate[(S)-lactic acid]
PEP[phosphoenolpyruvate]
ADP[ADP]
NAD[NAD(+)]
Butanediol[butanediol]
G3P[glyceraldehyde 3-phosphate]

Cottret2010_B_cicadellinicola_Met_Net: MODEL1011080002v0.0.1

This is metabolic network reconstruction of Baumannia cicadellinicola described in the article Graph-Based Analysis o…

Details

Endosymbiotic bacteria from different species can live inside cells of the same eukaryotic organism. Metabolic exchanges occur between host and bacteria but also between different endocytobionts. Since a complete genome annotation is available for both, we built the metabolic network of two endosymbiotic bacteria, Sulcia muelleri and Baumannia cicadellinicola, that live inside specific cells of the sharpshooter Homalodisca coagulata and studied the metabolic exchanges involving transfers of carbon atoms between the three. We automatically determined the set of metabolites potentially exogenously acquired (seeds) for both metabolic networks. We show that the number of seeds needed by both bacteria in the carbon metabolism is extremely reduced. Moreover, only three seeds are common to both metabolic networks, indicating that the complementarity of the two metabolisms is not only manifested in the metabolic capabilities of each bacterium, but also by their different use of the same environment. Furthermore, our results show that the carbon metabolism of S. muelleri may be completely independent of the metabolic network of B. cicadellinicola. On the contrary, the carbon metabolism of the latter appears dependent on the metabolism of S. muelleri, at least for two essential amino acids, threonine and lysine. Next, in order to define which subsets of seeds (precursor sets) are sufficient to produce the metabolites involved in a symbiotic function, we used a graph-based method, PITUFO, that we recently developed. Our results highly refine our knowledge about the complementarity between the metabolisms of the two bacteria and their host. We thus indicate seeds that appear obligatory in the synthesis of metabolites are involved in the symbiotic function. Our results suggest both B. cicadellinicola and S. muelleri may be completely independent of the metabolites provided by the co-resident endocytobiont to produce the carbon backbone of the metabolites provided to the symbiotic system (., thr and lys are only exploited by B. cicadellinicola to produce its proteins). link: http://identifiers.org/pubmed/20838465

Cottret2010_S_muelleri_Met_Net: MODEL1011080000v0.0.1

This is metabolic network reconstruction of Sulcia muelleri described in the article Graph-Based Analysis of the Meta…

Details

Endosymbiotic bacteria from different species can live inside cells of the same eukaryotic organism. Metabolic exchanges occur between host and bacteria but also between different endocytobionts. Since a complete genome annotation is available for both, we built the metabolic network of two endosymbiotic bacteria, Sulcia muelleri and Baumannia cicadellinicola, that live inside specific cells of the sharpshooter Homalodisca coagulata and studied the metabolic exchanges involving transfers of carbon atoms between the three. We automatically determined the set of metabolites potentially exogenously acquired (seeds) for both metabolic networks. We show that the number of seeds needed by both bacteria in the carbon metabolism is extremely reduced. Moreover, only three seeds are common to both metabolic networks, indicating that the complementarity of the two metabolisms is not only manifested in the metabolic capabilities of each bacterium, but also by their different use of the same environment. Furthermore, our results show that the carbon metabolism of S. muelleri may be completely independent of the metabolic network of B. cicadellinicola. On the contrary, the carbon metabolism of the latter appears dependent on the metabolism of S. muelleri, at least for two essential amino acids, threonine and lysine. Next, in order to define which subsets of seeds (precursor sets) are sufficient to produce the metabolites involved in a symbiotic function, we used a graph-based method, PITUFO, that we recently developed. Our results highly refine our knowledge about the complementarity between the metabolisms of the two bacteria and their host. We thus indicate seeds that appear obligatory in the synthesis of metabolites are involved in the symbiotic function. Our results suggest both B. cicadellinicola and S. muelleri may be completely independent of the metabolites provided by the co-resident endocytobiont to produce the carbon backbone of the metabolites provided to the symbiotic system (., thr and lys are only exploited by B. cicadellinicola to produce its proteins). link: http://identifiers.org/pubmed/20838465

Coulibaly2019 - Interleukin-15 Signaling in HIF-1a Regulation in Natural Killer Cells: BIOMD0000000867v0.0.1

This is a mathematical model comprised of non-linear ordinary differential equations describing the dynamic relationship…

Details

Natural killer (NK) cells belong to the first line of host defense against infection and cancer. Cytokines, including interleukin-15 (IL-15), critically regulate NK cell activity, resulting in recognition and direct killing of transformed and infected target cells. NK cells have to adapt and respond in inflamed and often hypoxic areas. Cellular stabilization and accumulation of the transcription factor hypoxia-inducible factor-1α (HIF-1α) is a key mechanism of the cellular hypoxia response. At the same time, HIF-1α plays a critical role in both innate and adaptive immunity. While the HIF-1α hydroxylation and degradation pathway has been recently described with the help of mathematical methods, less is known concerning the mechanistic mathematical description of processes regulating the levels of HIF-1α mRNA and protein. In this work we combine mathematical modeling with experimental laboratory analysis and examine the dynamic relationship between HIF-1α mRNA, HIF-1α protein, and IL-15-mediated upstream signaling events in NK cells from human blood. We propose a system of non-linear ordinary differential equations with positive and negative feedback loops for describing the complex interplay of HIF-1α regulators. The experimental design is optimized with the help of mathematical methods, and numerical optimization techniques yield reliable parameter estimates. The mathematical model allows for the investigation and prediction of HIF-1α stabilization under different inflammatory conditions and provides a better understanding of mechanisms mediating cellular enrichment of HIF-1α. Thanks to the combination of in vitro experimental data and in silico predictions we identified the mammalian target of rapamycin (mTOR), the nuclear factor-κB (NF-κB), and the signal transducer and activator of transcription 3 (STAT3) as central regulators of HIF-1α accumulation. We hypothesize that the regulatory pathway proposed here for NK cells can be extended to other types of immune cells. Understanding the molecular mechanisms involved in the dynamic regulation of the HIF-1α pathway in immune cells is of central importance to the immune cell function and could be a promising strategy in the design of treatments for human inflammatory diseases and cancer. link: http://identifiers.org/pubmed/31681292

Parameters:

NameDescription
k1 = 2.0E-5Reaction: => y2_Akt; y1_IL_15, Rate Law: compartment*k1*y1_IL_15
d8 = 0.577Reaction: y8_STAT3 =>, Rate Law: compartment*d8*y8_STAT3
d2 = 0.848Reaction: y2_Akt =>, Rate Law: compartment*d2*y2_Akt
delta = 200.0; xi10 = 8.127; a11 = 4.17; K_O2 = 0.96; k12 = 0.061Reaction: y10_HIF_1a_aOH => ; y6_HIF_1_Complex, Rate Law: compartment*k12*K_O2*y10_HIF_1a_aOH*(delta*y6_HIF_1_Complex+a11)/(xi10+y10_HIF_1a_aOH)
d1 = 0.062Reaction: y1_IL_15 =>, Rate Law: compartment*d1*y1_IL_15
d7 = 0.914Reaction: y7_NF_KB =>, Rate Law: compartment*d7*y7_NF_KB
d5 = 0.196Reaction: y5_HIF_1b =>, Rate Law: compartment*d5*y5_HIF_1b
k9 = 0.753Reaction: => y9_HIF_1a_mRNA; y7_NF_KB, Rate Law: compartment*k9*y7_NF_KB
kalpha = 1.034Reaction: => y4_HIF_1a; y9_HIF_1a_mRNA, Rate Law: compartment*kalpha*y9_HIF_1a_mRNA
phi = 0.829; K_O2 = 0.96; rho6 = 0.99; xi4 = 15.018; D = 1.0; k10 = 421.353Reaction: y4_HIF_1a => y10_HIF_1a_aOH, Rate Law: compartment*k10*K_O2*phi*y4_HIF_1a*(1-rho6*D)/(xi4+y4_HIF_1a)
a7 = 0.0Reaction: => y7_NF_KB, Rate Law: compartment*a7
k7 = 2.903Reaction: => y7_NF_KB; y1_IL_15, Rate Law: compartment*k7*y1_IL_15
k3 = 0.181Reaction: => y9_HIF_1a_mRNA; y8_STAT3, Rate Law: compartment*k3*y8_STAT3
d6 = 0.301Reaction: y6_HIF_1_Complex =>, Rate Law: compartment*d6*y6_HIF_1_Complex
k15 = 0.088Reaction: => y7_NF_KB; y3_mTOR, Rate Law: compartment*k15*y3_mTOR
a2 = 0.848Reaction: => y2_Akt, Rate Law: compartment*a2
d10 = 0.935Reaction: y10_HIF_1a_aOH =>, Rate Law: compartment*d10*y10_HIF_1a_aOH
d4 = 0.623Reaction: y4_HIF_1a =>, Rate Law: compartment*d4*y4_HIF_1a
a1 = 0.0Reaction: => y1_IL_15, Rate Law: compartment*a1
d9 = 0.934Reaction: y9_HIF_1a_mRNA =>, Rate Law: compartment*d9*y9_HIF_1a_mRNA
xi28 = 38.44; kS = 9.0E-4; n2 = 2.0Reaction: => y2_Akt; y8_STAT3, Rate Law: compartment*kS*y8_STAT3^n2/(xi28^n2+y8_STAT3^n2)
delta = 200.0; a11 = 4.17; K_O2 = 0.96; rho6 = 0.99; D = 1.0; xi44 = 128.022; k13 = 12.152Reaction: y4_HIF_1a => ; y6_HIF_1_Complex, Rate Law: compartment*k13*K_O2*y4_HIF_1a*(delta*y6_HIF_1_Complex+a11)*(1-rho6*D)/(xi44+y4_HIF_1a)
alpha1 = 1.163; R = 0.0; k2 = 0.307; a3 = 0.037; alpha2 = 0.386Reaction: => y3_mTOR; y2_Akt, y6_HIF_1_Complex, Rate Law: compartment*(a3+k2*y2_Akt)*alpha1*(1-R)/(alpha2*y6_HIF_1_Complex)
k4 = 76.196Reaction: y4_HIF_1a + y5_HIF_1b => y6_HIF_1_Complex, Rate Law: compartment*k4*y4_HIF_1a*y5_HIF_1b
k11 = 0.211Reaction: y10_HIF_1a_aOH => y4_HIF_1a, Rate Law: compartment*k11*y10_HIF_1a_aOH
a9 = 0.0Reaction: => y9_HIF_1a_mRNA, Rate Law: compartment*a9
d3 = 0.919Reaction: y3_mTOR =>, Rate Law: compartment*d3*y3_mTOR
a5 = 0.211Reaction: => y5_HIF_1b, Rate Law: compartment*a5
k5 = 75.895Reaction: y6_HIF_1_Complex => y4_HIF_1a + y5_HIF_1b, Rate Law: compartment*k5*y6_HIF_1_Complex
k8 = 0.577; a8 = 0.0; rho4 = 0.863; D = 1.0; k6 = 25.001; S3 = 0.0; rho3 = 1.0Reaction: => y8_STAT3; y3_mTOR, y1_IL_15, Rate Law: compartment*(a8+k8*y3_mTOR+k6*(1-rho4*D)*y1_IL_15)*(1-rho3*S3)
k14 = 16.528Reaction: => y7_NF_KB; y6_HIF_1_Complex, Rate Law: compartment*k14*y6_HIF_1_Complex

States:

NameDescription
y3 mTOR[PR:000003041]
y5 HIF 1b[C28553]
y7 NF KB[NF-kB]
y9 HIF 1a mRNA[C20214; Messenger RNA]
y1 IL 15[Interleukin-15]
y2 Akt[C41625]
y10 HIF 1a aOH[C20214; MOD:00677]
y8 STAT3[C28664]
y4 HIF 1a[C20214]
y6 HIF 1 Complex[C28553; C20214; Complex]

Courtemanche1998_AtrialActionPotential: MODEL0913049417v0.0.1

This a model from the article: Ionic mechanisms underlying human atrial action potential properties: insights from a m…

Details

The mechanisms underlying many important properties of the human atrial action potential (AP) are poorly understood. Using specific formulations of the K+, Na+, and Ca2+ currents based on data recorded from human atrial myocytes, along with representations of pump, exchange, and background currents, we developed a mathematical model of the AP. The model AP resembles APs recorded from human atrial samples and responds to rate changes, L-type Ca2+ current blockade, Na+/Ca2+ exchanger inhibition, and variations in transient outward current amplitude in a fashion similar to experimental recordings. Rate-dependent adaptation of AP duration, an important determinant of susceptibility to atrial fibrillation, was attributable to incomplete L-type Ca2+ current recovery from inactivation and incomplete delayed rectifier current deactivation at rapid rates. Experimental observations of variable AP morphology could be accounted for by changes in transient outward current density, as suggested experimentally. We conclude that this mathematical model of the human atrial AP reproduces a variety of observed AP behaviors and provides insights into the mechanisms of clinically important AP properties. link: http://identifiers.org/pubmed/9688927

Crespo2012 - Kinetics of Amyloid Fibril Formation: BIOMD0000000531v0.0.1

Crespo2012 - Kinetics of Amyloid Fibril FormationThis model is described in the article: [A generic crystallization-lik…

Details

Associated with neurodegenerative disorders such as Alzheimer, Parkinson, or prion diseases, the conversion of soluble proteins into amyloid fibrils remains poorly understood. Extensive "in vitro" measurements of protein aggregation kinetics have been reported, but no consensus mechanism has emerged until now. This contribution aims at overcoming this gap by proposing a theoretically consistent crystallization-like model (CLM) that is able to describe the classic types of amyloid fibrillization kinetics identified in our literature survey. Amyloid conversion represented as a function of time is shown to follow different curve shapes, ranging from sigmoidal to hyperbolic, according to the relative importance of the nucleation and growth steps. Using the CLM, apparently unrelated data are deconvoluted into generic mechanistic information integrating the combined influence of seeding, nucleation, growth, and fibril breakage events. It is notable that this complex assembly of interdependent events is ultimately reduced to a mathematically simple model, whose two parameters can be determined by little more than visual inspection. The good fitting results obtained for all cases confirm the CLM as a good approximation to the generalized underlying principle governing amyloid fibrillization. A perspective is presented on possible applications of the CLM during the development of new targets for amyloid disease therapeutics. link: http://identifiers.org/pubmed/22767606

Parameters:

NameDescription
kb = 1.6E-10; Ka = 1.44Reaction: alpha = 1-1/(kb*(exp(Ka*time)-1)+1), Rate Law: missing

States:

NameDescription
alpha[amyloid fibril]

Croft2013 - GPCR-RGS interaction that compartmentalizes RGS activity: BIOMD0000000479v0.0.1

Croft2013 - GPCR-RGS interaction that compartmentalizes RGS activityThrough modelling studies, the classic quaternary co…

Details

G protein-coupled receptors (GPCRs) can interact with regulator of G protein signaling (RGS) proteins. However, the effects of such interactions on signal transduction and their physiological relevance have been largely undetermined. Ligand-bound GPCRs initiate by promoting exchange of GDP for GTP on the Gα subunit of heterotrimeric G proteins. Signaling is terminated by hydrolysis of GTP to GDP through intrinsic GTPase activity of the Gα subunit, a reaction catalyzed by RGS proteins. Using yeast as a tool to study GPCR signaling in isolation, we define an interaction between the cognate GPCR (Mam2) and RGS (Rgs1), mapping the interaction domains. This reaction tethers Rgs1 at the plasma membrane and is essential for physiological signaling response. In vivo quantitative data inform the development of a kinetic model of the GTPase cycle, which extends previous attempts by including GPCR-RGS interactions. In vivo and in silico data confirm that GPCR-RGS interactions can impose an additional layer of regulation through mediating RGS subcellular localization to compartmentalize RGS activity within a cell, thus highlighting their importance as potential targets to modulate GPCR signaling pathways. link: http://identifiers.org/pubmed/23900842

Parameters:

NameDescription
k13=5.0E-4 1/hrReaction: RGSc => RGSm; RGSc, Rate Law: compartment*RGSc*k13
k36=50.0 1/(nM*hr)Reaction: GaGTPEffectorOFF + LRRGSm => LRRGSmGaGTPEffectorOFF; GaGTPEffectorOFF, LRRGSm, Rate Law: compartment*GaGTPEffectorOFF*LRRGSm*k36
k12=10.0 1/(nM*hr)Reaction: Effector + GaGTP => GaGTPEffector; Effector, GaGTP, Rate Law: compartment*Effector*GaGTP*k12
k32=0.5 1/hrReaction: RRGSmGaGTP => GaGDPP + RRGSm; RRGSmGaGTP, Rate Law: compartment*RRGSmGaGTP*k32
k6=0.005 1/(nM*hr)Reaction: RRGSm + Gabg => RRGSmGabg; RRGSm, Gabg, Rate Law: compartment*RRGSm*Gabg*k6
k18=100.0 1/hrReaction: LRRGSm => LR + RGSm; LRRGSm, Rate Law: compartment*LRRGSm*k18
k7=0.02 1/(nM*hr)Reaction: LRRGSm + Gabg => LRRGSmGabg; LRRGSm, Gabg, Rate Law: compartment*LRRGSm*Gabg*k7
k38=1000.0 1/hrReaction: GaGDPP => GaGDP + P; GaGDPP, Rate Law: compartment*GaGDPP*k38
k29=100.0 1/(nM*hr)Reaction: GaGTP + LRRGSm => LRRGSmGaGTP; GaGTP, LRRGSm, Rate Law: compartment*GaGTP*LRRGSm*k29
k28=2.5 1/hrReaction: RGSmGaGTP => GaGDPP + RGSc; RGSmGaGTP, Rate Law: compartment*RGSmGaGTP*k28
k27=500.0 1/(nM*hr)Reaction: GaGTP + RGSm => RGSmGaGTP; GaGTP, RGSm, Rate Law: compartment*GaGTP*RGSm*k27
k15=0.1 1/(nM*hr)Reaction: R + RGSc => RRGSm; R, RGSc, Rate Law: compartment*R*RGSc*k15
k21=0.1 1/(nM*hr)Reaction: LRGabg + RGSc => LRRGSmGabg; LRGabg, RGSc, Rate Law: compartment*LRGabg*RGSc*k21
k17=0.1 1/(nM*hr)Reaction: LR + RGSc => LRRGSm; LR, RGSc, Rate Law: compartment*LR*RGSc*k17
k34=50.0 1/(nM*hr)Reaction: GaGTPEffectorOFF + RGSm => RGSmGaGTPEffectorOFF; GaGTPEffectorOFF, RGSm, Rate Law: compartment*GaGTPEffectorOFF*RGSm*k34
k2=0.005 1/(nM*hr)Reaction: R + Gabg => RGabg; R, Gabg, Rate Law: compartment*R*Gabg*k2
k37=0.3 1/hrReaction: LRRGSmGaGTPEffectorOFF => GaGDPP + LRRGSm + Effector; LRRGSmGaGTPEffectorOFF, Rate Law: compartment*LRRGSmGaGTPEffectorOFF*k37
k40=10.0 1/hrReaction: P => ; P, Rate Law: compartment*P*k40
k5=0.005 1/(nM*hr)Reaction: L + RRGSm => LRRGSm; L, RRGSm, Rate Law: compartment*L*RRGSm*k5
k24=1.0E-4 1/(nM*hr)Reaction: GaGTPEffectorOFF + RGSc => RGSmGaGTPEffectorOFF; GaGTPEffectorOFF, RGSc, Rate Law: compartment*GaGTPEffectorOFF*RGSc*k24
k1=0.0025 1/(nM*hr)Reaction: L + R => LR; L, R, Rate Law: compartment*L*R*k1
k35=0.3 1/hrReaction: RGSmGaGTPEffectorOFF => GaGDPP + RGSc + Effector; RGSmGaGTPEffectorOFF, Rate Law: compartment*RGSmGaGTPEffectorOFF*k35
ka = 1.5 1/hrReaction: => z1; GaGTPEffector, GaGTPEffector, Rate Law: compartment*GaGTPEffector*ka
k22=60.0 1/(nM*hr)Reaction: GaGTP + RGSc => RGSmGaGTP; GaGTP, RGSc, Rate Law: compartment*GaGTP*RGSc*k22
k26=0.005 1/hrReaction: GaGTP => GaGDPP; GaGTP, Rate Law: compartment*GaGTP*k26
k39=1000.0 1/(nM*hr)Reaction: GaGDP + Gbg => Gabg; GaGDP, Gbg, Rate Law: compartment*GaGDP*Gbg*k39
k8=0.005 1/(nM*hr)Reaction: L + RRGSmGabg => LRRGSmGabg; L, RRGSmGabg, Rate Law: compartment*L*RRGSmGabg*k8
k10=0.2 1/hrReaction: Gabg => GaGTP + Gbg; Gabg, Rate Law: compartment*Gabg*k10
k4=0.005 1/(nM*hr)Reaction: L + RGabg => LRGabg; L, RGabg, Rate Law: compartment*L*RGabg*k4
k23=0.05 1/hrReaction: RGSmGaGTP => GaGTP + RGSc; RGSmGaGTP, Rate Law: compartment*RGSmGaGTP*k23
k19=0.1 1/(nM*hr)Reaction: RGabg + RGSc => RRGSmGabg; RGabg, RGSc, Rate Law: compartment*RGabg*RGSc*k19
k9=50.0 1/hrReaction: LRGabg => LR + GaGTP + Gbg; LRGabg, Rate Law: compartment*LRGabg*k9
k16=100.0 1/hrReaction: RRGSm => R + RGSm; RRGSm, Rate Law: compartment*RRGSm*k16
k11=40.0 1/hrReaction: LRRGSmGabg => GaGTP + Gbg + LRRGSm; LRRGSmGabg, Rate Law: compartment*LRRGSmGabg*k11
k14=0.005 1/hrReaction: RGSm => RGSc; RGSm, Rate Law: compartment*RGSm*k14
k3=0.02 1/(nM*hr)Reaction: LR + Gabg => LRGabg; LR, Gabg, Rate Law: compartment*LR*Gabg*k3
k25=1.0 1/hrReaction: GaGTPEffector => GaGTPEffectorOFF; GaGTPEffector, Rate Law: compartment*GaGTPEffector*k25
k31=0.5 1/(nM*hr)Reaction: GaGTP + RRGSm => RRGSmGaGTP; GaGTP, RRGSm, Rate Law: compartment*GaGTP*RRGSm*k31
k33=0.005 1/hrReaction: GaGTPEffectorOFF => GaGDPP + Effector; GaGTPEffectorOFF, Rate Law: compartment*GaGTPEffectorOFF*k33
k30=2.5 1/hrReaction: LRRGSmGaGTP => GaGDPP + LRRGSm; LRRGSmGaGTP, Rate Law: compartment*LRRGSmGaGTP*k30
k20=0.1 1/hrReaction: RRGSmGabg => RGabg + RGSm; RRGSmGabg, Rate Law: compartment*RRGSmGabg*k20

States:

NameDescription
RGabg[IPR000239; IPR001019; IPR001632; IPR001770]
GaGDPP[GDP; IPR001019; phosphorylated]
z1[delay]
P[phosphate(3-)]
RGSmGaGTP[Plasma membrane; GTP; IPR000342; IPR001019]
LRGabg[IPR000239; IPR001019; IPR001632; IPR001770; SBO:0000280]
L[SBO:0000280]
Gabg[IPR001019; IPR001632; IPR001770]
LRRGSm[Plasma membrane; IPR000239; IPR000342; SBO:0000280]
LRRGSmGabg[Plasma membrane; IPR000239; IPR001019; IPR000342; IPR001632; IPR001770; SBO:0000280]
RGSc[Cytoplasm; IPR000342]
GaGTP[GTP; IPR001019]
LR[IPR000239; SBO:0000280]
RGSmGaGTPEffectorOFF[effector; GTP; IPR000342; IPR001019; inactive]
LRRGSmGaGTP[GTP; IPR000342; IPR000239; IPR001019; SBO:0000280]
RRGSm[IPR000239; IPR000342; Plasma membrane]
LRRGSmGaGTPEffectorOFF[GTP; IPR000239; IPR001019; IPR000342; SBO:0000280; effector; inactive]
RRGSmGaGTP[GTP; IPR000239; IPR000342; IPR001019]
Gbg[IPR001632; IPR001770]
z3[delay]
GaGDP[GDP; IPR001019]
GaGTPEffector[GTP; IPR001019; effector]
RRGSmGabg[IPR000239; IPR000342; IPR001019; IPR001632; IPR001770; Plasma membrane]
RGSm[Plasma membrane; IPR000342]
Effector[effector]
R[IPR000239]
z2[delay]
GaGTPEffectorOFF[GTP; IPR001019; inactive; effector]

Cronwright2002_Glycerol_Synthesis: BIOMD0000000076v0.0.1

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

Details

Glycerol, a major by-product of ethanol fermentation by Saccharomyces cerevisiae, is of significant importance to the wine, beer, and ethanol production industries. To gain a clearer understanding of and to quantify the extent to which parameters of the pathway affect glycerol flux in S. cerevisiae, a kinetic model of the glycerol synthesis pathway has been constructed. Kinetic parameters were collected from published values. Maximal enzyme activities and intracellular effector concentrations were determined experimentally. The model was validated by comparing experimental results on the rate of glycerol production to the rate calculated by the model. Values calculated by the model agreed well with those measured in independent experiments. The model also mimics the changes in the rate of glycerol synthesis at different phases of growth. Metabolic control analysis values calculated by the model indicate that the NAD(+)-dependent glycerol 3-phosphate dehydrogenase-catalyzed reaction has a flux control coefficient (C(J)v1) of approximately 0.85 and exercises the majority of the control of flux through the pathway. Response coefficients of parameter metabolites indicate that flux through the pathway is most responsive to dihydroxyacetone phosphate concentration (R(J)DHAP= 0.48 to 0.69), followed by ATP concentration (R(J)ATP = -0.21 to -0.50). Interestingly, the pathway responds weakly to NADH concentration (R(J)NADH = 0.03 to 0.08). The model indicates that the best strategy to increase flux through the pathway is not to increase enzyme activity, substrate concentration, or coenzyme concentration alone but to increase all of these parameters in conjunction with each other. link: http://identifiers.org/pubmed/12200299

Parameters:

NameDescription
K2phi=1.0 mM; V2=53.0 mM_per_minute; K2g3p=3.5 mM; Phi=1.0 mMReaction: G3P => Gly, Rate Law: compartment*V2*G3P/K2g3p/((1+G3P/K2g3p)*(1+Phi/K2phi))
Keq1=10000.0 dimensionless; K1f16bp=4.8 mM; K1nadh=0.023 mM; NADH=1.87 mM; K1dhap=0.54 mM; ADP=2.17 mM; K1nad=0.93 mM; ATP=2.37 mM; K1atp=0.73 mM; K1adp=2.0 mM; F16BP=6.01 mM; K1g3p=1.2 mM; Vf1=47.0 mM_per_minute; NAD=1.45 mMReaction: DHAP => G3P, Rate Law: compartment*Vf1/(K1nadh*K1dhap)*(NADH*DHAP-NAD*G3P/Keq1)/((1+F16BP/K1f16bp+ATP/K1atp+ADP/K1adp)*(1+NADH/K1nadh+NAD/K1nad)*(1+DHAP/K1dhap+G3P/K1g3p))

States:

NameDescription
Gly[glycerol; Glycerol]
DHAP[dihydroxyacetone phosphate; Glycerone phosphate]
G3P[sn-glycerol 3-phosphate; sn-Glycerol 3-phosphate]

Csikasz-Nagy2006_Cell_Cycle: MODEL3897771820v0.0.1

This model originates from the [Cell Cycle Database](http://www.itb.cnr.it/cellcycle/) . It is described in: **Analys…

Details

We propose a protein interaction network for the regulation of DNA synthesis and mitosis that emphasizes the universality of the regulatory system among eukaryotic cells. The idiosyncrasies of cell cycle regulation in particular organisms can be attributed, we claim, to specific settings of rate constants in the dynamic network of chemical reactions. The values of these rate constants are determined ultimately by the genetic makeup of an organism. To support these claims, we convert the reaction mechanism into a set of governing kinetic equations and provide parameter values (specific to budding yeast, fission yeast, frog eggs, and mammalian cells) that account for many curious features of cell cycle regulation in these organisms. Using one-parameter bifurcation diagrams, we show how overall cell growth drives progression through the cell cycle, how cell-size homeostasis can be achieved by two different strategies, and how mutations remodel bifurcation diagrams and create unusual cell-division phenotypes. The relation between gene dosage and phenotype can be summarized compactly in two-parameter bifurcation diagrams. Our approach provides a theoretical framework in which to understand both the universality and particularity of cell cycle regulation, and to construct, in modular fashion, increasingly complex models of the networks controlling cell growth and division. link: http://identifiers.org/pubmed/16581849

Cuadros2020 - SIHRD spatiotemporal model of COVID-19 transmission in Ohio: BIOMD0000000969v0.0.1

The role of geospatial disparities in the dynamics of the COVID-19 pandemic is poorly understood. We developed a spatial…

Details

The role of geospatial disparities in the dynamics of the COVID-19 pandemic is poorly understood. We developed a spatially-explicit mathematical model to simulate transmission dynamics of COVID-19 disease infection in relation with the uneven distribution of the healthcare capacity in Ohio, U.S. The results showed substantial spatial variation in the spread of the disease, with localized areas showing marked differences in disease attack rates. Higher COVID-19 attack rates experienced in some highly connected and urbanized areas (274 cases per 100,000 people) could substantially impact the critical health care response of these areas regardless of their potentially high healthcare capacity compared to more rural and less connected counterparts (85 cases per 100,000). Accounting for the spatially uneven disease diffusion linked to the geographical distribution of the critical care resources is essential in designing effective prevention and control programmes aimed at reducing the impact of COVID-19 pandemic. link: http://identifiers.org/pubmed/32736312

Cucuianu2010 - A hypothetical-mathematical model of acute myeloid leukaemia pathogenesis: BIOMD0000000799v0.0.1

This is a simple mathematical model describing the growth and removal of normal and leukemic haematopoietic stem cell po…

Details

Acute myeloid leukaemia is defined by the expansion of a mutated haematopoietic stem cell clone, with the inhibition of surrounding normal clones. Haematopoiesis can be seen as an evolutionary tree, starting with one cell that undergoes several divisions during the expansion phase, afterwards losing functional cells during the aging-related contraction phase. During divisions, offspring cells acquire variations, which can be either normal or abnormal. If an abnormal variation is present in more than 25% of the final cells, a monoclonal, leukemic pattern occurs. Such a pattern develops if: (A1) The abnormal variation occurs early, during the first or second divisions; (A2) The variation confers exceptional proliferative capacity; (B) A sizable proportion of the normal clones are destroyed and a previously non-significant abnormal clone gains relative dominance over a depleted environment; (C) The abnormal variation confers relative immortality, rendering it significant during the contraction phase. Combinations of these pathways further enhance the leukemic risk of the system. A simple mathematical model is used in order to characterize normal and leukemic states and to explain the above cellular processes generating monoclonal leukemic patterns. link: http://identifiers.org/doi/10.1080/17486700902973751

Parameters:

NameDescription
c = 0.1Reaction: x_Normal_Hematopoietic_Stem_Cell =>, Rate Law: compartment*c*x_Normal_Hematopoietic_Stem_Cell
C = 0.1Reaction: y_Leukemic_Cell =>, Rate Law: compartment*C*y_Leukemic_Cell
a = 0.3; b = 0.5Reaction: => x_Normal_Hematopoietic_Stem_Cell; y_Leukemic_Cell, Rate Law: compartment*a*x_Normal_Hematopoietic_Stem_Cell/(1+b*(x_Normal_Hematopoietic_Stem_Cell+y_Leukemic_Cell))
B = 0.5; A = 0.3Reaction: => y_Leukemic_Cell; x_Normal_Hematopoietic_Stem_Cell, Rate Law: compartment*A*y_Leukemic_Cell/(1+B*(x_Normal_Hematopoietic_Stem_Cell+y_Leukemic_Cell))

States:

NameDescription
y Leukemic Cell[leukemic stem cell; bone marrow]
x Normal Hematopoietic Stem Cell[hematopoietic stem cell; bone marrow]

Cui2006_CalciumHomeostasis: MODEL0913003363v0.0.1

This a model from the article: Mathematical modeling of calcium homeostasis in yeast cells. Cui J, Kaandorp JA. Cell…

Details

In this study, based on currently available experimental observations on protein level, we constructed a mathematical model to describe calcium homeostasis in normally growing yeast cells (Saccharomyces cerevisiae). Simulation results show that tightly controlled low cytosolic calcium ion level can be a natural result under the general mechanism of gene expression feedback control. The calmodulin (a sensor protein) behavior in our model cell agrees well with relevant observations in real cells. Moreover, our model can qualitatively reproduce the experimentally observed response curve of real yeast cell responding to step-like disturbance in extracellular calcium ion concentration. Further investigations show that the feedback control mechanism in our model is as robust as it is in real cells. link: http://identifiers.org/pubmed/16445978

Cui2008 - in vitro transcriptional response of zinc homeostasis system in Escherichia coli: BIOMD0000000966v0.0.1

BACKGROUND: The zinc homeostasis system in Escherichia coli is one of the most intensively studied prokaryotic zinc home…

Details

BACKGROUND: The zinc homeostasis system in Escherichia coli is one of the most intensively studied prokaryotic zinc homeostasis systems. Its underlying regulatory machine consists of repression on zinc influx through ZnuABC by Zur (Zn2+ uptake regulator) and activation on zinc efflux via ZntA by ZntR (a zinc-responsive regulator). Although these transcriptional regulations seem to be well characterized, and there is an abundance of detailed in vitro experimental data available, as yet there is no mathematical model to help interpret these data. To our knowledge, the work described here is the first attempt to use a mathematical model to simulate these regulatory relations and to help explain the in vitro experimental data. RESULTS: We develop a unified mathematical model consisting of 14 reactions to simulate the in vitro transcriptional response of the zinc homeostasis system in E. coli. Firstly, we simulate the in vitro Zur-DNA interaction by using two of these reactions, which are expressed as 4 ordinary differential equations (ODEs). By imposing the conservation restraints and solving the relevant steady state equations, we find that the simulated sigmoidal curve matches the corresponding experimental data. Secondly, by numerically solving the ODEs for simulating the Zur and ZntR run-off transcription experiments, and depicting the simulated concentrations of zntA and znuC transcripts as a function of free zinc concentration, we find that the simulated curves fit the corresponding in vitro experimental data. Moreover, we also perform simulations, after taking into consideration the competitive effects of ZntR with the zinc buffer, and depict the simulated concentration of zntA transcripts as a function of the total ZntR concentration, both in the presence and absence of Zn(II). The obtained simulation results are in general agreement with the corresponding experimental data. CONCLUSION: Simulation results show that our model can quantitatively reproduce the results of several of the in vitro experiments conducted by Outten CE and her colleagues. Our model provides a detailed insight into the dynamics of the regulatory system and also provides a general framework for simulating in vitro metal-binding and transcription experiments and interpreting the relevant experimental data. link: http://identifiers.org/pubmed/18950480

Cui2008_CardiacMyocytes: MODEL1172425728v0.0.1

This a model from the article: Simulating Complex Calcium-Calcineurin Signaling Network Jiangjun Cui and Jaap A. Kaa…

Details

Understanding of processes in which calcium signaling is involved is of fundamental importance in systems biology and has many applications in medicine. In this paper we have studied the particular case of the complex calcium-calcineurin-MCIP-NFAT signaling network in cardiac myocytes, the understanding of which is critical for treatment of pathologic hypertrophy and heart failure. By including some most recent experimental findings, we constructed a computational model totally based on biochemical principles. The model can correctly predict the mutant (MCIP1−/−) behavior under different stress such as PO (pressure overload) and Caivated calcineurin) overexpression. link: http://identifiers.org/doi/10.1007/978-3-540-69389-5_14

Curien2003_MetThr_synthesis: BIOMD0000000068v0.0.1

This a model from the article: A kinetic model of the branch-point between the methionine and threonine biosynthesis p…

Details

This work proposes a model of the metabolic branch-point between the methionine and threonine biosynthesis pathways in Arabidopsis thaliana which involves kinetic competition for phosphohomoserine between the allosteric enzyme threonine synthase and the two-substrate enzyme cystathionine gamma-synthase. Threonine synthase is activated by S-adenosylmethionine and inhibited by AMP. Cystathionine gamma-synthase condenses phosphohomoserine to cysteine via a ping-pong mechanism. Reactions are irreversible and inhibited by inorganic phosphate. The modelling procedure included an examination of the kinetic links, the determination of the operating conditions in chloroplasts and the establishment of a computer model using the enzyme rate equations. To test the model, the branch-point was reconstituted with purified enzymes. The computer model showed a partial agreement with the in vitro results. The model was subsequently improved and was then found consistent with flux partition in vitro and in vivo. Under near physiological conditions, S-adenosylmethionine, but not AMP, modulates the partition of a steady-state flux of phosphohomoserine. The computer model indicates a high sensitivity of cystathionine flux to enzyme and S-adenosylmethionine concentrations. Cystathionine flux is sensitive to modulation of threonine flux whereas the reverse is not true. The cystathionine gamma-synthase kinetic mechanism favours a low sensitivity of the fluxes to cysteine. Though sensitivity to inorganic phosphate is low, its concentration conditions the dynamics of the system. Threonine synthase and cystathionine gamma-synthase display similar kinetic efficiencies in the metabolic context considered and are first-order for the phosphohomoserine substrate. Under these conditions outflows are coordinated. link: http://identifiers.org/pubmed/14622248

Parameters:

NameDescription
KmPHSER=2500.0 microM; kcat2=30.0 microM; KmCYS=460.0 microM; Ki2=2000.0 microMReaction: Phser + Cys => Cystathionine + Phi; CGS, Rate Law: CGS*kcat2/(1+KmCYS/Cys)*Phser/(Phser+KmPHSER*(1+Phi/Ki2)/(1+KmCYS/Cys))
V0=1.0 microM_per_secondReaction: Hser => Phser, Rate Law: compartment*V0
Ki3=1000.0 microMReaction: Phser => Thr + Phi; AdoMet, TS, Rate Law: TS*(5.9E-4+0.062*AdoMet^2.9/(32^2.9+AdoMet^2.9))*Phser/(1+Phi/Ki3)

States:

NameDescription
Cys[L-cysteine; L-Cysteine]
Hser[L-homoserine; L-Homoserine]
Thr[L-threonine; L-Threonine]
Cystathionine[L-cystathionine; L-Cystathionine]
Phi[phosphate(3-); Orthophosphate]
Phser[O-phospho-L-homoserine; O-Phospho-L-homoserine]

Curien2009_Aspartate_Metabolism: BIOMD0000000212v0.0.1

This a model described in the article: Understanding the regulation of aspartate metabolism using a model based on mea…

Details

The aspartate-derived amino-acid pathway from plants is well suited for analysing the function of the allosteric network of interactions in branched pathways. For this purpose, a detailed kinetic model of the system in the plant model Arabidopsis was constructed on the basis of in vitro kinetic measurements. The data, assembled into a mathematical model, reproduce in vivo measurements and also provide non-intuitive predictions. A crucial result is the identification of allosteric interactions whose function is not to couple demand and supply but to maintain a high independence between fluxes in competing pathways. In addition, the model shows that enzyme isoforms are not functionally redundant, because they contribute unequally to the flux and its regulation. Another result is the identification of the threonine concentration as the most sensitive variable in the system, suggesting a regulatory role for threonine at a higher level of integration. link: http://identifiers.org/pubmed/19455135

Parameters:

NameDescription
AKII_kreverse_app_exp=0.22 l*μmol^(-1)*s^(-1); AKII_Thr_Ki_app_exp=109.0 μmol*l^(-1); AKII_nH_exp=2.0 dimensionless; AKII_kforward_app_exp=1.35 s^(-1)Reaction: Asp => AspP; AKHSDHII, Thr, Rate Law: c1*AKHSDHII*(AKII_kforward_app_exp-AKII_kreverse_app_exp*AspP)/(1+(Thr/AKII_Thr_Ki_app_exp)^AKII_nH_exp)
AKI_kreverse_app_exp=0.15 l*μmol^(-1)*s^(-1); AKI_nH_exp=2.0 dimensionless; AKI_Thr_Ki_app_exp=124.0 μmol*l^(-1); AKI_kforward_app_exp=0.36 s^(-1)Reaction: Asp => AspP; AKHSDHI, Thr, Rate Law: c1*AKHSDHI*(AKI_kforward_app_exp-AKI_kreverse_app_exp*AspP)/(1+(Thr/AKI_Thr_Ki_app_exp)^AKI_nH_exp)
TS1_kcatmin_exp=0.42 dimensionless; TS1_nH_exp=2.0 dimensionless; TS1_AdoMet_Ka2_exp=0.5 dimensionless; TS1_AdoMet_Ka1_exp=73.0 μmol^2*l^(-2); TS1_AdoMEt_Km_no_AdoMet_exp=250.0 dimensionless; TS1_AdoMet_Ka3_exp=1.09 dimensionless; TS1_AdoMet_kcatmax_exp=3.5 dimensionless; TS1_Phosphate_Ki_exp=1000.0 μmol*l^(-1); TS1_AdoMet_Ka4_exp=140.0 μmol^2*l^(-2)Reaction: PHser => Thr; TS1, Phosphate, AdoMet, Rate Law: c1*TS1*PHser*(TS1_kcatmin_exp+TS1_AdoMet_kcatmax_exp*AdoMet^TS1_nH_exp/TS1_AdoMet_Ka1_exp)/(1+AdoMet^TS1_nH_exp/TS1_AdoMet_Ka1_exp)/(TS1_AdoMEt_Km_no_AdoMet_exp*(1+AdoMet/TS1_AdoMet_Ka2_exp)/(1+AdoMet/TS1_AdoMet_Ka3_exp)/(1+AdoMet^TS1_nH_exp/TS1_AdoMet_Ka4_exp)*(1+Phosphate/TS1_Phosphate_Ki_exp)+PHser)
AK2_kforward_app_exp=3.15 s^(-1); AK2_nH_exp=1.1 dimensionless; AK2_kreverse_app_exp=0.86 l*μmol^(-1)*s^(-1); AK2_Lys_Ki_app_exp=22.0 μmol*l^(-1)Reaction: Asp => AspP; AK2, Lys, Rate Law: c1*AK2*(AK2_kforward_app_exp-AK2_kreverse_app_exp*AspP)/(1+(Lys/AK2_Lys_Ki_app_exp)^AK2_nH_exp)
TD_nH_app_exp=3.0 dimensionless; TD_k_app_exp=0.0124 dimensionless; TD_Ile_Ki_no_Val_app_exp=30.0 dimensionless; TD_Val_Ka1_app_exp=73.0 dimensionless; TD_Val_Ka2_app_exp=615.0 μmol*l^(-1)Reaction: Thr => Ile; TD, Val, Ile, Rate Law: c1*TD*Thr*TD_k_app_exp/(1+(Ile/(TD_Ile_Ki_no_Val_app_exp+TD_Val_Ka1_app_exp*Val/(TD_Val_Ka2_app_exp+Val)))^TD_nH_app_exp)
AK1_nH_exp=2.0 dimensionless; AK1_kreverse_app_exp=1.6 l*μmol^(-1)*s^(-1); AK1_kforward_app_exp=5.65 s^(-1); AK1_Lys_Ki_app_exp=550.0 μmol*l^(-1); AK1_AdoMet_Ka_app_exp=3.5 μmol*l^(-1)Reaction: Asp => AspP; AK1, Lys, AdoMet, Rate Law: c1*AK1*(AK1_kforward_app_exp-AK1_kreverse_app_exp*AspP)/(1+(Lys/(AK1_Lys_Ki_app_exp/(1+AdoMet/AK1_AdoMet_Ka_app_exp)))^AK1_nH_exp)
THA_Thr_Km_exp=7100.0 μmol*l^(-1); THA_kcat_exp=1.7 s^(-1)Reaction: Thr => Gly; THA, Rate Law: c1*THA_kcat_exp*THA*Thr/(THA_Thr_Km_exp+Thr)
ASADH_kforward_app_exp=0.9 l*μmol^(-1)*s^(-1); ASADH_kreverse_app_exp=0.23 l*μmol^(-1)*s^(-1)Reaction: AspP => ASA; ASADH, Rate Law: c1*ASADH*(ASADH_kforward_app_exp*AspP-ASADH_kreverse_app_exp*ASA)
HSDHII_Thr_relative_inhibition_app_exp=0.75 dimensionless; HSDHII_kforward_app_exp=0.64 l*μmol^(-1)*s^(-1); HSDHII_Thr_relative_residual_activity_app_exp=0.25 dimensionless; HSDHII_Thr_Ki_app_exp=8500.0 μmol*l^(-1)Reaction: ASA => Hser; AKHSDHII, Thr, Rate Law: c1*HSDHII_kforward_app_exp*AKHSDHII*ASA*(HSDHII_Thr_relative_residual_activity_app_exp+HSDHII_Thr_relative_inhibition_app_exp/(1+Thr/HSDHII_Thr_Ki_app_exp))
DHDPS2_Lys_Ki_app_exp=33.0 μmol*l^(-1); DHDPS2_nH_exp=2.0 dimensionless; DHDPS2_k_app_exp=1.0 μmol*l^(-1)Reaction: ASA => Lys; DHDPS2, Lys, Rate Law: c1*DHDPS2_k_app_exp*DHDPS2*ASA*1/(1+(Lys/DHDPS2_Lys_Ki_app_exp)^DHDPS2_nH_exp)
V_Thr_RS = 0.43 μmol*l^(-1)*s^(-1); Thr_tRNAS_Thr_Km=100.0 μmol*l^(-1)Reaction: Thr => ThrTRNA, Rate Law: c1*V_Thr_RS*Thr/(Thr_tRNAS_Thr_Km+Thr)
DHDPS1_nH_exp=2.0 dimensionless; DHDPS1_Lys_Ki_app_exp=10.0 μmol*l^(-1); DHDPS1_k_app_exp=1.0 μmol*l^(-1)Reaction: ASA => Lys; DHDPS1, Lys, Rate Law: c1*DHDPS1_k_app_exp*DHDPS1*ASA*1/(1+(Lys/DHDPS1_Lys_Ki_app_exp)^DHDPS1_nH_exp)
LKR_Lys_Km_exp=13000.0 μmol*l^(-1); LKR_kcat_exp=3.1 s^(-1)Reaction: Lys => Sacc; LKR, Rate Law: c1*LKR_kcat_exp*LKR*Lys/(LKR_Lys_Km_exp+Lys)
HSK_Hser_app_exp=14.0 μmol*l^(-1); HSK_kcat_app_exp=2.8 s^(-1)Reaction: Hser => PHser; HSK, Rate Law: c1*HSK_kcat_app_exp*HSK*Hser/(HSK_Hser_app_exp+Hser)
HSDHI_Thr_relative_inhibition_app_exp=0.86 dimensionless; HSDHI_kforward_app_exp=0.84 l*μmol^(-1)*s^(-1); HSDHI_Thr_relative_residual_activity_app_exp=0.14 dimensionless; HSDHI_Thr_Ki_app_exp=400.0 μmol*l^(-1)Reaction: ASA => Hser; AKHSDHI, Thr, Rate Law: c1*HSDHI_kforward_app_exp*AKHSDHI*ASA*(HSDHI_Thr_relative_residual_activity_app_exp+HSDHI_Thr_relative_inhibition_app_exp/(1+Thr/HSDHI_Thr_Ki_app_exp))
CGS_kcat_exp=30.0 dimensionless; CGS_Phosphate_Ki_exp=2000.0 dimensionless; CGS_Cys_Km_exp=460.0 dimensionless; CGS_Phser_Km_exp=2500.0 dimensionlessReaction: PHser => Cysta; CGS, Cys, Phosphate, Rate Law: c1*CGS*PHser*CGS_kcat_exp/(1+CGS_Cys_Km_exp/Cys)/(CGS_Phser_Km_exp/(1+CGS_Cys_Km_exp/Cys)*(1+Phosphate/CGS_Phosphate_Ki_exp)+PHser)
Ile_tRNAS_Ile_Km=20.0 μmol*l^(-1); V_Ile_RS = 0.43 μmol*l^(-1)*s^(-1)Reaction: Ile => IleTRNA, Rate Law: c1*V_Ile_RS*Ile/(Ile_tRNAS_Ile_Km+Ile)
Lys_tRNAS_Lys_Km=25.0 μmol*l^(-1); V_Lys_RS = 0.43 μmol*l^(-1)*s^(-1)Reaction: Lys => LysTRNA, Rate Law: c1*V_Lys_RS*Lys/(Lys_tRNAS_Lys_Km+Lys)

States:

NameDescription
IleTRNA[Ile-tRNA(Ile); L-Isoleucyl-tRNA(Ile)]
Gly[glycine; Glycine]
Lys[L-lysine; L-Lysine; 47205736]
Cysta[cystathionine; Cystathionine]
ASA[L-aspartate 4-semialdehyde; L-Aspartate 4-semialdehyde]
Asp[L-aspartic acid; L-Aspartate; 47205730]
Hser[L-homoserine; L-Homoserine]
Thr[L-threonine; L-Threonine]
AspP[4-phospho-L-aspartic acid; 4-Phospho-L-aspartate]
ThrTRNA[Thr-tRNA(Thr); L-Threonyl-tRNA(Thr)]
PHser[O-phospho-L-homoserine; O-Phospho-L-homoserine]
Sacc[L-saccharopine; N6-(L-1,3-Dicarboxypropyl)-L-lysine]
Ile[L-isoleucine; L-Isoleucine]
LysTRNA[Lys-tRNA(Lys); L-Lysyl-tRNA]

Cursons2015 - Regulation of ERK-MAPK signaling in human epidermis: BIOMD0000000659v0.0.1

Cursons2015 - Regulation of ERK-MAPK signaling in human epidermisModel comparing the abundance of phosphorylated MAPK si…

Details

The skin is largely comprised of keratinocytes within the interfollicular epidermis. Over approximately two weeks these cells differentiate and traverse the thickness of the skin. The stage of differentiation is therefore reflected in the positions of cells within the tissue, providing a convenient axis along which to study the signaling events that occur in situ during keratinocyte terminal differentiation, over this extended two-week timescale. The canonical ERK-MAPK signaling cascade (Raf-1, MEK-1/2 and ERK-1/2) has been implicated in controlling diverse cellular behaviors, including proliferation and differentiation. While the molecular interactions involved in signal transduction through this cascade have been well characterized in cell culture experiments, our understanding of how this sequence of events unfolds to determine cell fate within a homeostatic tissue environment has not been fully characterized.We measured the abundance of total and phosphorylated ERK-MAPK signaling proteins within interfollicular keratinocytes in transverse cross-sections of human epidermis using immunofluorescence microscopy. To investigate these data we developed a mathematical model of the signaling cascade using a normalized-Hill differential equation formalism.These data show coordinated variation in the abundance of phosphorylated ERK-MAPK components across the epidermis. Statistical analysis of these data shows that associations between phosphorylated ERK-MAPK components which correspond to canonical molecular interactions are dependent upon spatial position within the epidermis. The model demonstrates that the spatial profile of activation for ERK-MAPK signaling components across the epidermis may be maintained in a cell-autonomous fashion by an underlying spatial gradient in calcium signaling.Our data demonstrate an extended phospho-protein profile of ERK-MAPK signaling cascade components across the epidermis in situ, and statistical associations in these data indicate canonical ERK-MAPK interactions underlie this spatial profile of ERK-MAPK activation. Using mathematical modelling we have demonstrated that spatially varying calcium signaling components across the epidermis may be sufficient to maintain the spatial profile of ERK-MAPK signaling cascade components in a cell-autonomous manner. These findings may have significant implications for the wide range of cancer drugs which therapeutically target ERK-MAPK signaling components. link: http://identifiers.org/pubmed/26209520

Parameters:

NameDescription
funcHillMEKToERKNuc = 0.387623644887571 1; numCytoToNucVolRatio = 2.35714285714286 1; numERKCytoToNucParam = 0.01 1; numERKNucToCytoParam = 0.01 1; numHillMax = 1.0 1; funcHillERKToERKNuc = 0.16212840948525 1; numHillTau = 1.0 1Reaction: pERK_nuc = 1/numHillTau*((((funcHillMEKToERKNuc-funcHillERKToERKNuc)*numHillMax-pERK_nuc)-numERKCytoToNucParam*pERK_nuc)+numCytoToNucVolRatio*numERKNucToCytoParam*pERK_cyto), Rate Law: 1/numHillTau*((((funcHillMEKToERKNuc-funcHillERKToERKNuc)*numHillMax-pERK_nuc)-numERKCytoToNucParam*pERK_nuc)+numCytoToNucVolRatio*numERKNucToCytoParam*pERK_cyto)
numCytoToNucVolRatio = 2.35714285714286 1; numMEKCytoToNucParam = 0.05 1; numMEKNucToCytoParam = 0.5 1; numHillTau = 1.0 1Reaction: pMEK_nuc = 1/numHillTau*(((-pMEK_nuc)-numMEKNucToCytoParam*pMEK_nuc)+numCytoToNucVolRatio*numMEKCytoToNucParam*pMEK_cyto), Rate Law: 1/numHillTau*(((-pMEK_nuc)-numMEKNucToCytoParam*pMEK_nuc)+numCytoToNucVolRatio*numMEKCytoToNucParam*pMEK_cyto)
funcHillMEKToERKCyto = 0.819554213811451 1; numCytoToNucVolRatio = 2.35714285714286 1; numERKCytoToNucParam = 0.01 1; numERKNucToCytoParam = 0.01 1; numHillMax = 1.0 1; numHillTau = 1.0 1Reaction: pERK_cyto = 1/numHillTau*(((funcHillMEKToERKCyto*numHillMax-pERK_cyto)-numERKCytoToNucParam*pERK_cyto)+1/numCytoToNucVolRatio*numERKNucToCytoParam*pERK_nuc), Rate Law: 1/numHillTau*(((funcHillMEKToERKCyto*numHillMax-pERK_cyto)-numERKCytoToNucParam*pERK_cyto)+1/numCytoToNucVolRatio*numERKNucToCytoParam*pERK_nuc)
numHillMax = 1.0 1; numTotalRafInputs = 0.0 1; numHillTau = 1.0 1Reaction: pRaf_cyto = 1/numHillTau*(numTotalRafInputs*numHillMax-pRaf_cyto), Rate Law: 1/numHillTau*(numTotalRafInputs*numHillMax-pRaf_cyto)
numCytoToNucVolRatio = 2.35714285714286 1; numHillMax = 1.0 1; numMEKCytoToNucParam = 0.05 1; numMEKNucToCytoParam = 0.5 1; numHillTau = 1.0 1; funcHillRafToMEK = 0.501898596166978 1Reaction: pMEK_cyto = 1/numHillTau*(((funcHillRafToMEK*numHillMax-pMEK_cyto)-numMEKCytoToNucParam*pMEK_cyto)+1/numCytoToNucVolRatio*numMEKNucToCytoParam*pMEK_nuc), Rate Law: 1/numHillTau*(((funcHillRafToMEK*numHillMax-pMEK_cyto)-numMEKCytoToNucParam*pMEK_cyto)+1/numCytoToNucVolRatio*numMEKNucToCytoParam*pMEK_nuc)

States:

NameDescription
pRaf cyto[RAF proto-oncogene serine/threonine-protein kinase; urn:miriam:pato:PATO_0002220]
pERK cyto[urn:miriam:pato:PATO_0002220; Mitogen-activated protein kinase 1]
pMEK nuc[Dual specificity mitogen-activated protein kinase kinase 1; urn:miriam:pato:PATO_0002220]
pERK nuc[urn:miriam:pato:PATO_0002220; Mitogen-activated protein kinase 1]
pMEK cyto[urn:miriam:pato:PATO_0002220; Dual specificity mitogen-activated protein kinase kinase 1]

Curto1998 - purine metabolism: BIOMD0000000015v0.0.1

Curto1998 - purine metabolismThis is a purine metabolism model that is geared toward studies of gout. The model uses Ge…

Details

Experimental and clinical data on purine metabolism are collated and analyzed with three mathematical models. The first model is the result of an attempt to construct a traditional kinetic model based on Michaelis-Menten rate laws. This attempt is only partially successful, since kinetic information, while extensive, is not complete, and since qualitative information is difficult to incorporate into this type of model. The data gaps necessitate the complementation of the Michaelis-Menten model with other functional forms that can incorporate different types of data. The most convenient and established representations for this purpose are rate laws formulated as power-law functions, and these are used to construct a Complemented Michaelis-Menten (CMM) model. The other two models are pure power-law-representations, one in the form of a Generalized Mass Action (GMA) system, and the other one in the form of an S-system. The first part of the paper contains a compendium of experimental data necessary for any model of purine metabolism. This is followed by the formulation of the three models and a comparative analysis. For physiological and moderately pathological perturbations in metabolites or enzymes, the results of the three models are very similar and consistent with clinical findings. This is an encouraging result since the three models have different structures and data requirements and are based on different mathematical assumptions. Significant enzyme deficiencies are not so well modeled by the S-system model. The CMM model captures the dynamics better, but judging by comparisons with clinical observations, the best model in this case is the GMA model. The model results are discussed in some detail, along with advantages and disadvantages of each modeling strategy. link: http://identifiers.org/pubmed/9664759

Parameters:

NameDescription
ahx=0.003793; fhx13=1.12Reaction: HX =>, Rate Law: ahx*HX^fhx13
aadna=3.2789; fdnap9=0.42; fdnap10=0.33Reaction: dATP => DNA; dGTP, Rate Law: aadna*dATP^fdnap9*dGTP^fdnap10
frnan11=1.0; arnag=0.04615Reaction: RNA => GTP, Rate Law: arnag*RNA^frnan11
aasli=66544.0; fasli3=0.99; fasli4=-0.95Reaction: SAMP => ATP; ATP, Rate Law: aasli*SAMP^fasli3*ATP^fasli4
fxd14=0.55; axd=0.949Reaction: Xa => UA, Rate Law: axd*Xa^fxd14
fgnuc18=-0.34; agnuc=0.2511; fgnuc8=0.9Reaction: GTP => Gua; Pi, Rate Law: agnuc*GTP^fgnuc8*Pi^fgnuc18
apyr=1.2951; fpyr1=1.27Reaction: PRPP =>, Rate Law: apyr*PRPP^fpyr1
fhxd13=0.65; ahxd=0.2754Reaction: HX => Xa, Rate Law: ahxd*HX^fhxd13
agrna=409.6; frnap4=0.05; frnap8=0.13Reaction: GTP => RNA; ATP, Rate Law: agrna*ATP^frnap4*GTP^frnap8
fgprt15=0.42; fgprt8=-1.2; agprt=361.69; fgprt1=1.2Reaction: Gua + PRPP => GTP; GTP, Rate Law: agprt*PRPP^fgprt1*GTP^fgprt8*Gua^fgprt15
fmat5=-0.6; amat=7.2067; fmat4=0.2Reaction: ATP => SAM; SAM, Rate Law: amat*ATP^fmat4*SAM^fmat5
aadrnr=0.0602; fadrnr10=0.87; fadrnr9=-0.3; fadrnr4=0.1Reaction: ATP => dATP; dGTP, dATP, Rate Law: aadrnr*ATP^fadrnr4*dATP^fadrnr9*dGTP^fadrnr10
fimpd2=0.15; aimpd=1.2823; fimpd8=-0.03; fimpd7=-0.09Reaction: IMP => XMP; GTP, XMP, Rate Law: aimpd*IMP^fimpd2*XMP^fimpd7*GTP^fimpd8
fdada9=1.0; adada=0.03333Reaction: dATP => HX, Rate Law: adada*dATP^fdada9
agmpr=0.3005; fgmpr7=-0.76; fgmpr2=-0.15; fgmpr4=-0.07; fgmpr8=0.7Reaction: GTP => IMP; XMP, ATP, IMP, Rate Law: agmpr*IMP^fgmpr2*ATP^fgmpr4*XMP^fgmpr7*GTP^fgmpr8
aden=5.2728; fden8=-0.2; fden1=2.0; fden4=-0.25; fden18=-0.08; fden2=-0.06Reaction: PRPP => IMP; dGTP, IMP, ATP, GTP, Pi, Rate Law: aden*PRPP^fden1*IMP^fden2*ATP^fden4*GTP^fden8*Pi^fden18
fdnap9=0.42; fdnap10=0.33; agdna=2.2296Reaction: dGTP => DNA; dATP, Rate Law: agdna*dATP^fdnap9*dGTP^fdnap10
fdnan12=1.0; adnag=0.001318Reaction: DNA => dGTP, Rate Law: adnag*DNA^fdnan12
fdnan12=1.0; adnaa=0.001938Reaction: DNA => dATP, Rate Law: adnaa*DNA^fdnan12
fhprt2=-0.89; fhprt1=1.1; fhprt13=0.48; ahprt=12.569Reaction: HX + PRPP => IMP; IMP, Rate Law: ahprt*PRPP^fhprt1*IMP^fhprt2*HX^fhprt13
finuc2=0.8; finuc18=-0.36; ainuc=0.9135Reaction: IMP => HX; Pi, Rate Law: ainuc*IMP^finuc2*Pi^finuc18
atrans=8.8539; ftrans5=0.33Reaction: SAM => ATP, Rate Law: atrans*SAM^ftrans5
apolyam=0.29; fpolyam5=0.9Reaction: SAM => Ade, Rate Law: apolyam*SAM^fpolyam5
fgdrnr10=-0.39; agdrnr=0.1199; fgdrnr8=0.4; fgdrnr9=-1.2Reaction: GTP => dGTP; dATP, dGTP, Rate Law: agdrnr*GTP^fgdrnr8*dATP^fgdrnr9*dGTP^fgdrnr10
fgmps7=0.16; fgmps4=0.12; agmps=0.3738Reaction: XMP => GTP; ATP, Rate Law: agmps*ATP^fgmps4*XMP^fgmps7
aada=0.001062; fada4=0.97Reaction: ATP => HX, Rate Law: aada*ATP^fada4
fprpps1=-0.03; fprpps4=-0.45; fprpps17=0.65; fprpps18=0.7; aprpps=0.9; fprpps8=-0.04Reaction: R5P => PRPP; ATP, GTP, Pi, PRPP, Rate Law: aprpps*PRPP^fprpps1*ATP^fprpps4*GTP^fprpps8*R5P^fprpps17*Pi^fprpps18
adgnuc=0.03333; fdgnuc10=1.0Reaction: dGTP => Gua, Rate Law: adgnuc*dGTP^fdgnuc10
arnaa=0.06923; frnan11=1.0Reaction: RNA => ATP, Rate Law: arnaa*RNA^frnan11
fx14=2.0; ax=0.0012Reaction: Xa =>, Rate Law: ax*Xa^fx14
fasuc18=-0.05; fasuc4=-0.24; fasuc2=0.4; fasuc8=0.2; aasuc=3.5932Reaction: IMP => SAMP; ATP, GTP, Pi, Rate Law: aasuc*IMP^fasuc2*ATP^fasuc4*GTP^fasuc8*Pi^fasuc18
aaprt=233.8; faprt4=-0.8; faprt1=0.5; faprt6=0.75Reaction: PRPP + Ade => ATP; ATP, Rate Law: aaprt*PRPP^faprt1*ATP^faprt4*Ade^faprt6
aua=8.744E-5; fua16=2.21Reaction: UA =>, Rate Law: aua*UA^fua16
agua=0.4919; fgua15=0.5Reaction: Gua => Xa, Rate Law: agua*Gua^fgua15
aarna=614.5; frnap4=0.05; frnap8=0.13Reaction: ATP => RNA; GTP, Rate Law: aarna*ATP^frnap4*GTP^frnap8
fampd4=0.8; fampd8=-0.03; aampd=0.02688; fampd18=-0.1Reaction: ATP => IMP; GTP, Pi, Rate Law: aampd*ATP^fampd4*GTP^fampd8*Pi^fampd18
aade=0.01; fade6=0.55Reaction: Ade =>, Rate Law: aade*Ade^fade6

States:

NameDescription
ATP[ATP; adenosine; AMP; ADP; ADP; Adenosine; AMP; ATP; ADP]
PRPP[5-O-phosphono-alpha-D-ribofuranosyl diphosphate; 5-Phospho-alpha-D-ribose 1-diphosphate]
SAMP[N(6)-(1,2-dicarboxyethyl)-AMP; N6-(1,2-Dicarboxyethyl)-AMP]
R5P[aldehydo-D-ribose 5-phosphate; D-Ribose 5-phosphate]
SAM[S-adenosyl-L-methionine; S-Adenosyl-L-methionine]
DNA[DNA; deoxyribonucleic acid]
Xa[9H-xanthine; Xanthine]
UA[7,9-dihydro-1H-purine-2,6,8(3H)-trione; Urate]
GTP[GDP; GMP; GTP; GDP; GMP; GDP; GTP]
HX[Hypoxanthine; Deoxyinosine; Inosine; inosine; hypoxanthine; 2'-deoxyinosine]
dGTP[dGTP; dGMP; dGDP; dGTP; dGDP; dGMP; dGTP]
Ade[adenine; Adenine]
dATP[dATP; dADP; dAMP; Deoxyadenosine; dATP; dATP; dADP; 2'-deoxyadenosine; dAMP]
Gua[Guanine; Deoxyguanosine; Guanosine; 2'-deoxyuridine; guanine; 2'-deoxyguanosine]
RNA[RNA]
IMP[IMP; IMP]
XMP[5'-xanthylic acid; Xanthosine 5'-phosphate]

D


D1 LTP time window: MODEL1603270000v0.0.1

Nair2016 - Integration of calcium and dopamine signals by D1R-expressing medium-sized spiny neuronsThis model is describ…

Details

In reward learning, the integration of NMDA-dependent calcium and dopamine by striatal projection neurons leads to potentiation of corticostriatal synapses through CaMKII/PP1 signaling. In order to elicit the CaMKII/PP1-dependent response, the calcium and dopamine inputs should arrive in temporal proximity and must follow a specific (dopamine after calcium) order. However, little is known about the cellular mechanism which enforces these temporal constraints on the signal integration. In this computational study, we propose that these temporal requirements emerge as a result of the coordinated signaling via two striatal phosphoproteins, DARPP-32 and ARPP-21. Specifically, DARPP-32-mediated signaling could implement an input-interval dependent gating function, via transient PP1 inhibition, thus enforcing the requirement for temporal proximity. Furthermore, ARPP-21 signaling could impose the additional input-order requirement of calcium and dopamine, due to its Ca2+/calmodulin sequestering property when dopamine arrives first. This highlights the possible role of phosphoproteins in the temporal aspects of striatal signal transduction. link: http://identifiers.org/pubmed/27584878