SBMLBioModels: N - P

N


Nutsch2005_phototaxis_noncyc_attractant_dark: MODEL0404023805v0.0.1

A quantitative model of the switch cycle of an archaeal flagellar motor and its sensory control, Nutsch et al, Biophys.…

Details

A recent phototaxis model of Halobacterium salinarum composed of the signalling pathway and the switch complex of the motor explained all considered experimental data on spontaneous switching and response time to repellent or attractant light stimuli. However, the model which considers symmetric processes in the clockwise and counter-clockwise rotations of the motor cannot explain the behaviour of a CheY(D10K,Yl00W) mutant which always moves forward and does not respond to light. We show that the introduction of asymmetry in the motor switch model can explain this behaviour. Sensitivity analysis allowed us to choose parameters for which the model is sensitive and whose values we then change in either direction to obtain an asymmetric model. We also demonstrate numerically that at low concentrations of CheYP, the symmetric and asymmetric models behave similarly, but at high concentrations, differences in the clockwise and counter-clockwise modes become apparent. Thus, those experimental data that could previously be explained only by ad hoc assumptions are now obtained 'naturally' from the revised model. link: http://identifiers.org/pubmed/17708428

Nutsch2005_phototaxis_noncyc_attractant_light: MODEL0403988150v0.0.1

A quantitative model of the switch cycle of an archaeal flagellar motor and its sensory control, Nutsch et al, Biophys.…

Details

A recent phototaxis model of Halobacterium salinarum composed of the signalling pathway and the switch complex of the motor explained all considered experimental data on spontaneous switching and response time to repellent or attractant light stimuli. However, the model which considers symmetric processes in the clockwise and counter-clockwise rotations of the motor cannot explain the behaviour of a CheY(D10K,Yl00W) mutant which always moves forward and does not respond to light. We show that the introduction of asymmetry in the motor switch model can explain this behaviour. Sensitivity analysis allowed us to choose parameters for which the model is sensitive and whose values we then change in either direction to obtain an asymmetric model. We also demonstrate numerically that at low concentrations of CheYP, the symmetric and asymmetric models behave similarly, but at high concentrations, differences in the clockwise and counter-clockwise modes become apparent. Thus, those experimental data that could previously be explained only by ad hoc assumptions are now obtained 'naturally' from the revised model. link: http://identifiers.org/pubmed/17708428

Nutsch2005_phototaxis_noncyc_repellent_dark: MODEL0403954746v0.0.1

A quantitative model of the switch cycle of an archaeal flagellar motor and its sensory control, Nutsch et al, Biophys.…

Details

A recent phototaxis model of Halobacterium salinarum composed of the signalling pathway and the switch complex of the motor explained all considered experimental data on spontaneous switching and response time to repellent or attractant light stimuli. However, the model which considers symmetric processes in the clockwise and counter-clockwise rotations of the motor cannot explain the behaviour of a CheY(D10K,Yl00W) mutant which always moves forward and does not respond to light. We show that the introduction of asymmetry in the motor switch model can explain this behaviour. Sensitivity analysis allowed us to choose parameters for which the model is sensitive and whose values we then change in either direction to obtain an asymmetric model. We also demonstrate numerically that at low concentrations of CheYP, the symmetric and asymmetric models behave similarly, but at high concentrations, differences in the clockwise and counter-clockwise modes become apparent. Thus, those experimental data that could previously be explained only by ad hoc assumptions are now obtained 'naturally' from the revised model. link: http://identifiers.org/pubmed/17708428

Nutsch2005_phototaxis_noncyc_repellent_light: MODEL0403928902v0.0.1

A quantitative model of the switch cycle of an archaeal flagellar motor and its sensory control, Nutsch et al, Biophys.…

Details

A recent phototaxis model of Halobacterium salinarum composed of the signalling pathway and the switch complex of the motor explained all considered experimental data on spontaneous switching and response time to repellent or attractant light stimuli. However, the model which considers symmetric processes in the clockwise and counter-clockwise rotations of the motor cannot explain the behaviour of a CheY(D10K,Yl00W) mutant which always moves forward and does not respond to light. We show that the introduction of asymmetry in the motor switch model can explain this behaviour. Sensitivity analysis allowed us to choose parameters for which the model is sensitive and whose values we then change in either direction to obtain an asymmetric model. We also demonstrate numerically that at low concentrations of CheYP, the symmetric and asymmetric models behave similarly, but at high concentrations, differences in the clockwise and counter-clockwise modes become apparent. Thus, those experimental data that could previously be explained only by ad hoc assumptions are now obtained 'naturally' from the revised model. link: http://identifiers.org/pubmed/17708428

Nutsch2005_phototaxis_noncyc_spontaneous: MODEL0403888565v0.0.1

A quantitative model of the switch cycle of an archaeal flagellar motor and its sensory control, Nutsch et al, Biophys.…

Details

A recent phototaxis model of Halobacterium salinarum composed of the signalling pathway and the switch complex of the motor explained all considered experimental data on spontaneous switching and response time to repellent or attractant light stimuli. However, the model which considers symmetric processes in the clockwise and counter-clockwise rotations of the motor cannot explain the behaviour of a CheY(D10K,Yl00W) mutant which always moves forward and does not respond to light. We show that the introduction of asymmetry in the motor switch model can explain this behaviour. Sensitivity analysis allowed us to choose parameters for which the model is sensitive and whose values we then change in either direction to obtain an asymmetric model. We also demonstrate numerically that at low concentrations of CheYP, the symmetric and asymmetric models behave similarly, but at high concentrations, differences in the clockwise and counter-clockwise modes become apparent. Thus, those experimental data that could previously be explained only by ad hoc assumptions are now obtained 'naturally' from the revised model. link: http://identifiers.org/pubmed/17708428

Nwabugwu2013 - A Tumor-Immune Mathematical Model of CD4+ T Helper Cell: MODEL2007090001v0.0.1

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

Details

Understanding the complex dynamics between the tumor cells and the host immune system will be key to improved therapeutic strategies against cancer. We propose an ODE-based mathematical model of both the tumor and immune system and how they respond to inactivation of the driving oncogene. Our model supports experimental results showing that cellular senescence of tumor cells is dependent on CD4+ T helper cells, leading to relapse of tumors in immunocompromised hosts. link: http://identifiers.org/pubmed/24110741

Nyman2011_M3Hierarachical_InsulinGlucosedynamics: BIOMD0000000356v0.0.1

This a model from the article: A Hierarchical Whole-body Modeling Approach Elucidates the Link between in Vitro Insu…

Details

Type 2 diabetes is a metabolic disease that profoundly affects energy homeostasis. The disease involves failure at several levels and subsystems and is characterized by insulin resistance in target cells and tissues (i.e. by impaired intracellular insulin signaling). We have previously used an iterative experimental-theoretical approach to unravel the early insulin signaling events in primary human adipocytes. That study, like most insulin signaling studies, is based on in vitro experimental examination of cells, and the in vivo relevance of such studies for human beings has not been systematically examined. Herein, we develop a hierarchical model of the adipose tissue, which links intracellular insulin control of glucose transport in human primary adipocytes with whole-body glucose homeostasis. An iterative approach between experiments and minimal modeling allowed us to conclude that it is not possible to scale up the experimentally determined glucose uptake by the isolated adipocytes to match the glucose uptake profile of the adipose tissue in vivo. However, a model that additionally includes insulin effects on blood flow in the adipose tissue and GLUT4 translocation due to cell handling can explain all data, but neither of these additions is sufficient independently. We also extend the minimal model to include hierarchical dynamic links to more detailed models (both to our own models and to those by others), which act as submodules that can be turned on or off. The resulting multilevel hierarchical model can merge detailed results on different subsystems into a coherent understanding of whole-body glucose homeostasis. This hierarchical modeling can potentially create bridges between other experimental model systems and the in vivo human situation and offers a framework for systematic evaluation of the physiological relevance of in vitro obtained molecular/cellular experimental data. link: http://identifiers.org/pubmed/21572040

Parameters:

NameDescription
Kend = 30.6825110077Reaction: r11x2 => rendP + iendIR, Rate Law: Kend*r11x2
kfbasal = 0.49752158Reaction: r0 => rPbasal, Rate Law: kfbasal*r0
krbasal = 128042.884096176Reaction: rPbasal => r0, Rate Law: krbasal*rPbasal
Kcr = 0.0013640432Reaction: r1 => r1x2, Rate Law: Kcr*r1
d1 = 0.7722612342Reaction: r1 => r0, Rate Law: d1*r1
a2 = 4321891.90327031; S1 = 0.0Reaction: r1 => r12, Rate Law: a2*S1*r1
K8 = 0.0Reaction: r1x22d => r1x22, Rate Law: K8*r1x22d
k21 = 0.009645863; k24 = 0.9430860972; k22 = 2374.9773277896; k23 = 0.1199031163Reaction: IRS => IRSiP; PKC_P, mTOR, r11x2, r11x22, r1x2, r1x22, r1x22d, rPbasal, rendP, Rate Law: k21*IRS*(r1x2+r11x2+r1x22+r1x22d+r11x22+rPbasal+k22*rendP)*(1+k23*PKC_P+k24*mTOR)
Kex = 37.0818924842Reaction: rend => r0, Rate Law: Kex*rend
Kcat = 237.5189220434; Km = 3.0181933402; Kdp = 9.500831E-4Reaction: iendIR => iend; X_P, Rate Law: (Kdp+Kcat*X_P/(Km+X_P))*iendIR
S1 = 0.0; a1 = 4.4825146271204E8Reaction: r1 => r11, Rate Law: a1*S1*r1
d2 = 0.0122057759Reaction: r12 => r1, Rate Law: d2*r12
a2 = 4321891.90327031; S2 = 0.0Reaction: r1x2 => r1x22d, Rate Law: a2*S2*r1x2
S1 = 0.0; K4 = 0.0Reaction: r1x22 => r1x22d, Rate Law: K4*S1*r1x22

States:

NameDescription
r12[Insulin receptor]
rend[p-6Y-insulin receptor [endosome membrane]]
r1x2[Insulin receptor]
IRS[Insulin receptor substrate 1; IRS1,IRS2 [cytosol]]
r1x22d[Insulin receptor]
iend[insulin [endosome lumen]]
iendIR[Insulin:p-6Y-insulin receptor [endosome membrane]]
r1[Insulin receptor]
r2[Insulin receptor]
r11[Insulin receptor]
r1x22[Insulin receptor]
r22[Insulin receptor]
rendP[p-6Y-insulin receptor [endosome membrane]]
r0[Insulin receptor]
r11x22[Insulin receptor]
r11x2[Insulin receptor]
rPbasal[Insulin:p-6Y-Insulin receptor [plasma membrane]]

Nyman2012_InsulinSignalling: BIOMD0000000423v0.0.1

This model is from the article: Mechanistic explanations for counter-intuitive phosphorylation dynamics of the insulin…

Details

Insulin signaling through insulin receptor (IR) and insulin receptor substrate-1 (IRS1) is important for insulin control of target cells. We have previously demonstrated a rapid and simultaneous overshoot behavior in the phosphorylation dynamics of IR and IRS1 in human adipocytes. Herein, we demonstrate that in murine adipocytes a similar overshoot behavior is not simultaneous for IR and IRS1. The peak of IRS1 phosphorylation, which is a direct consequence of the phosphorylation and the activation of IR, occurs earlier than the peak of IR phosphorylation. We used a conclusive modeling framework to unravel the mechanisms behind this counter-intuitive order of phosphorylation. Through a number of rejections, we demonstrate that two fundamentally different mechanisms may create the reversed order of peaks: (i) two pools of phosphorylated IR, where a large pool of internalized IR peaks late, but phosphorylation of IRS1 is governed by a small plasma membrane-localized pool of IR with an early peak, or (ii) inhibition of the IR-catalyzed phosphorylation of IRS1 by negative feedback. Although (i) may explain the reversed order, this two-pool hypothesis alone requires extensive internalization of IR, which is not supported by experimental data. However, with the additional assumption of limiting concentrations of IRS1, (i) can explain all data. Also, (ii) can explain all available data. Our findings illustrate how modeling can potentiate reasoning, to help draw nontrivial conclusions regarding competing mechanisms in signaling networks. Our work also reveals new differences between human and murine insulin signaling. link: http://identifiers.org/pubmed/22248283

Parameters:

NameDescription
k1a = 0.153418; ins = 100.0; k1aBasic = 0.0383389Reaction: IR => IRins, Rate Law: k1a*ins*IR+k1aBasic*IR
k3 = 8.62917E-5Reaction: X => Xp; IRSiP, Rate Law: k3*X*IRSiP
k1e = 5.25027E-5; k1f = 119.353Reaction: IRiP => IRi; Xp, Rate Law: IRiP*(k1e+k1f*Xp/(1+Xp))
k1b = 3.4699E-6Reaction: IRins => IR, Rate Law: k1b*IRins
k1g = 4.14899Reaction: IRp => IR, Rate Law: k1g*IRp
k1r = 37954.7Reaction: IRi => IR, Rate Law: k1r*IRi
km3 = 0.132671Reaction: Xp => X, Rate Law: km3*Xp
k21 = 538004.0; km23 = 88.9096; k22 = 1.7252E-6Reaction: IRS => IRSiP; IRiP, IRp, Xp, Rate Law: k21*IRS*(IRp+k22*IRiP)/(1+km23*Xp)
k1c = 0.574266Reaction: IRins => IRp, Rate Law: k1c*IRins
km2 = 262759.0Reaction: IRSiP => IRS, Rate Law: km2*IRSiP
k1d = 4.78844Reaction: IRp => IRiP, Rate Law: k1d*IRp

States:

NameDescription
IRp[Insulin receptor; Phosphoprotein]
IRiP[Insulin receptor; Phosphoprotein]
X[Intermediate]
IR[Insulin receptor]
IRS[Insulin receptor substrate 1]
IRins[Insulin receptor; Insulin-1]
IRi[Insulin receptor]
IRSiP[Insulin receptor substrate 1; Phosphoprotein]
Xp[Phosphoprotein; Intermediate]

O


Oberhardt2008 - Genome-scale metabolic network of Pseudomonas aeruginosa (iMO1056): MODEL1507180020v0.0.1

Oberhardt2008 - Genome-scale metabolic network of Pseudomonas aeruginosa (iMO1056)This model is described in the article…

Details

Pseudomonas aeruginosa is a major life-threatening opportunistic pathogen that commonly infects immunocompromised patients. This bacterium owes its success as a pathogen largely to its metabolic versatility and flexibility. A thorough understanding of P. aeruginosa's metabolism is thus pivotal for the design of effective intervention strategies. Here we aim to provide, through systems analysis, a basis for the characterization of the genome-scale properties of this pathogen's versatile metabolic network. To this end, we reconstructed a genome-scale metabolic network of Pseudomonas aeruginosa PAO1. This reconstruction accounts for 1,056 genes (19% of the genome), 1,030 proteins, and 883 reactions. Flux balance analysis was used to identify key features of P. aeruginosa metabolism, such as growth yield, under defined conditions and with defined knowledge gaps within the network. BIOLOG substrate oxidation data were used in model expansion, and a genome-scale transposon knockout set was compared against in silico knockout predictions to validate the model. Ultimately, this genome-scale model provides a basic modeling framework with which to explore the metabolism of P. aeruginosa in the context of its environmental and genetic constraints, thereby contributing to a more thorough understanding of the genotype-phenotype relationships in this resourceful and dangerous pathogen. link: http://identifiers.org/pubmed/18192387

Obeyesekere1999_CellCycle: BIOMD0000000168v0.0.1

The model reproduces the time profiles of the different species depicted in Fig 3a of the paper. Model successfully repr…

Details

A modified version of a previously developed mathematical model [Obeyesekere et al., Cell Prolif. (1997)] of the G1-phase of the cell cycle is presented. This model describes the regulation of the G1-phase that includes the interactions of the nuclear proteins, RB, cyclin E, cyclin D, cdk2, cdk4 and E2F. The effects of the growth factors on cyclin D synthesis under saturated or unsaturated growth factor conditions are investigated based on this model. The solutions to this model (a system of nonlinear ordinary differential equations) are discussed with respect to existing experiments. Predictions based on mathematical analysis of this model are presented. In particular, results are presented on the existence of two stable solutions, i.e., bistability within the G1-phase. It is shown that this bistability exists under unsaturated growth factor concentration levels. This phenomenon is very noticeable if the efficiency of the signal transduction, initiated by the growth factors leading to cyclin D synthesis, is low. The biological significance of this result as well as possible experimental designs to test these predictions are presented. link: http://identifiers.org/pubmed/17886749

Parameters:

NameDescription
RT_1 = 2.5Reaction: RP_1 = (RT_1-RS_1)-R_1, Rate Law: missing
qD_1 = 0.6; pD_1 = 0.48Reaction: RS_1 => ; RS_1, D_1, Rate Law: pD_1*RS_1*D_1/(qD_1+RS_1+D_1)
aD_1 = 0.4; k_1 = 0.05; GF_1 = 6.3Reaction: => D_1, Rate Law: aD_1*k_1*GF_1/(1+k_1*GF_1)
dX_1 = 1.04Reaction: X_1 => ; X_1, Rate Law: dX_1*X_1
qX_1 = 0.8; pX_1 = 0.48Reaction: => R_1; RP_1, X_1, Rate Law: pX_1*RP_1*X_1/(qX_1+RP_1+X_1)
qE_1 = 0.6; pE_1 = 0.096Reaction: RS_1 => ; RS_1, E_1, Rate Law: pE_1*RS_1*E_1/(qE_1+RS_1+E_1)
dE_1 = 0.2Reaction: E_1 => ; E_1, X_1, Rate Law: dE_1*X_1*E_1
af_1 = 0.9; aE_1 = 0.16Reaction: => E_1; E2F_1, Rate Law: aE_1*(1+af_1*E2F_1)
theta_1 = 1.5Reaction: E2F_1 = theta_1-RS_1, Rate Law: missing
dD_1 = 0.4Reaction: D_1 => ; D_1, E_1, Rate Law: dD_1*E_1*D_1
pS_1 = 0.6Reaction: R_1 => RS_1; RS_1, R_1, E2F_1, Rate Law: pS_1*E2F_1*R_1
f_1 = 0.2; g_1 = 0.528; aX_1 = 0.08Reaction: => X_1; E_1, E2F_1, X_1, Rate Law: aX_1*E_1+f_1*E2F_1+g_1*X_1^2*E_1

States:

NameDescription
E2F 1[Transcription factor E2F1]
RP 1[Retinoblastoma-associated protein]
R 1[Retinoblastoma-associated protein]
X 1X
D 1[G1/S-specific cyclin-D1]
E 1[G1/S-specific cyclin-E1]
RS 1[Transcription factor E2F1; Retinoblastoma-associated protein]

Oda2005_EGFR: MODEL2463576061v0.0.1

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

Details

The epidermal growth factor receptor (EGFR) signaling pathway is one of the most important pathways that regulate growth, survival, proliferation, and differentiation in mammalian cells. Reflecting this importance, it is one of the best-investigated signaling systems, both experimentally and computationally, and several computational models have been developed for dynamic analysis. A map of molecular interactions of the EGFR signaling system is a valuable resource for research in this area. In this paper, we present a comprehensive pathway map of EGFR signaling and other related pathways. The map reveals that the overall architecture of the pathway is a bow-tie (or hourglass) structure with several feedback loops. The map is created using CellDesigner software that enables us to graphically represent interactions using a well-defined and consistent graphical notation, and to store it in Systems Biology Markup Language (SBML). link: http://identifiers.org/pubmed/16729045

Oda2006_TollLikeR: MODEL2463683119v0.0.1

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

Details

Recognition of pathogen-associated molecular signatures is critically important in proper activation of the immune system. The toll-like receptor (TLR) signaling network is responsible for innate immune response. In mammalians, there are 11 TLRs that recognize a variety of ligands from pathogens to trigger immunological responses. In this paper, we present a comprehensive map of TLRs and interleukin 1 receptor signaling networks based on papers published so far. The map illustrates the possible existence of a main network subsystem that has a bow-tie structure in which myeloid differentiation primary response gene 88 (MyD88) is a nonredundant core element, two collateral subsystems with small GTPase and phosphatidylinositol signaling, and MyD88-independent pathway. There is extensive crosstalk between the main bow-tie network and subsystems, as well as feedback and feedforward controls. One obvious feature of this network is the fragility against removal of the nonredundant core element, which is MyD88, and involvement of collateral subsystems for generating different reactions and gene expressions for different stimuli. link: http://identifiers.org/pubmed/16738560

ODea2007 - A homeostatic model of IκB metabolism to control constitutive NF-κB activity: MODEL1908270002v0.0.1

Insert Citation Questions concerning the paper should be addressed to the corresponding author. Alexander Hoffmann (a…

Details

Cellular signal transduction pathways are usually studied following administration of an external stimulus. However, disease-associated aberrant activity of the pathway is often due to misregulation of the equilibrium state. The transcription factor NF-kappaB is typically described as being held inactive in the cytoplasm by binding its inhibitor, IkappaB, until an external stimulus triggers IkappaB degradation through an IkappaB kinase-dependent degradation pathway. Combining genetic, biochemical, and computational tools, we investigate steady-state regulation of the NF-kappaB signaling module and its impact on stimulus responsiveness. We present newly measured in vivo degradation rate constants for NF-kappaB-bound and -unbound IkappaB proteins that are critical for accurate computational predictions of steady-state IkappaB protein levels and basal NF-kappaB activity. Simulations reveal a homeostatic NF-kappaB signaling module in which differential degradation rates of free and bound pools of IkappaB represent a novel cross-regulation mechanism that imparts functional robustness to the signaling module. link: http://identifiers.org/pubmed/17486138

ODea2007_IkappaB: BIOMD0000000147v0.0.1

O'Dea, E.L., Barken, D., Peralta, R.Q., Tran K.T., Werner, S.L., Kearns, J.D., Levchenko, A., Hoffmann, A. A homeostatic…

Details

Cellular signal transduction pathways are usually studied following administration of an external stimulus. However, disease-associated aberrant activity of the pathway is often due to misregulation of the equilibrium state. The transcription factor NF-kappaB is typically described as being held inactive in the cytoplasm by binding its inhibitor, IkappaB, until an external stimulus triggers IkappaB degradation through an IkappaB kinase-dependent degradation pathway. Combining genetic, biochemical, and computational tools, we investigate steady-state regulation of the NF-kappaB signaling module and its impact on stimulus responsiveness. We present newly measured in vivo degradation rate constants for NF-kappaB-bound and -unbound IkappaB proteins that are critical for accurate computational predictions of steady-state IkappaB protein levels and basal NF-kappaB activity. Simulations reveal a homeostatic NF-kappaB signaling module in which differential degradation rates of free and bound pools of IkappaB represent a novel cross-regulation mechanism that imparts functional robustness to the signaling module. link: http://identifiers.org/pubmed/17486138

Parameters:

NameDescription
k2_a=0.828 per_minReaction: IkBaNFkB_nucleus => IkBaNFkB_cytoplasm, Rate Law: nucleus*k2_a*IkBaNFkB_nucleus
deg2_n=0.18 per_minReaction: IkBb_nucleus =>, Rate Law: nucleus*deg2_n*IkBb_nucleus
tr1b=0.2448 per_minReaction: IkBb_mRNA => IkBb_mRNA + IkBb_cytoplasm, Rate Law: nucleus*tr1b*IkBb_mRNA
a9=4.2 per_uM_per_min; d3_2=0.105 per_minReaction: IkBeNFkB_cytoplasm + IKK => IkBeIKKNFkB, Rate Law: cytoplasm*(a9*IkBeNFkB_cytoplasm*IKK-d3_2*IkBeIKKNFkB)
a6_3=30.0 per_uM_per_min; d6_3=6.0E-5 per_minReaction: IkBeIKK + NFkB_cytoplasm => IkBeIKKNFkB, Rate Law: cytoplasm*(a6_3*IkBeIKK*NFkB_cytoplasm-d6_3*IkBeIKKNFkB)
tp2e=0.012 per_min; tp1e=0.018 per_minReaction: IkBe_cytoplasm => IkBe_nucleus, Rate Law: cytoplasm*tp1e*IkBe_cytoplasm-nucleus*tp2e*IkBe_nucleus
r3=0.036 per_minReaction: IkBeIKK => IKK, Rate Law: cytoplasm*r3*IkBeIKK
k2_b=0.414 per_minReaction: IkBbNFkB_nucleus => IkBbNFkB_cytoplasm, Rate Law: nucleus*k2_b*IkBbNFkB_nucleus
deg4_n=6.0E-5 per_minReaction: IkBaNFkB_nucleus => NFkB_nucleus, Rate Law: nucleus*deg4_n*IkBaNFkB_nucleus
tr2b=4.272E-5 uM_per_minReaction: => IkBb_mRNA, Rate Law: nucleus*tr2b
tr2a=1.848E-4 uM_per_minReaction: => IkBa_mRNA, Rate Law: nucleus*tr2a
deg2_c=0.18 per_minReaction: IkBb_cytoplasm =>, Rate Law: cytoplasm*deg2_c*IkBb_cytoplasm
deg1_n=0.12 per_minReaction: IkBa_nucleus =>, Rate Law: nucleus*deg1_n*IkBa_nucleus
k1_1=0.0048 per_min; k1_2=5.4 per_minReaction: NFkB_cytoplasm => NFkB_nucleus, Rate Law: cytoplasm*k1_2*NFkB_cytoplasm-nucleus*k1_1*NFkB_nucleus
deg4_c=6.0E-5 per_minReaction: IkBaNFkB_cytoplasm => NFkB_cytoplasm, Rate Law: cytoplasm*deg4_c*IkBaNFkB_cytoplasm
deg3_c=0.18 per_minReaction: IkBe_cytoplasm =>, Rate Law: cytoplasm*deg3_c*IkBe_cytoplasm
r2=0.024 per_minReaction: IkBbIKK => IKK, Rate Law: cytoplasm*r2*IkBbIKK
k_IKK_deg=0.0 per_minReaction: IKK =>, Rate Law: cytoplasm*k_IKK_deg*IKK
k2_e=0.414 per_minReaction: IkBeNFkB_nucleus => IkBeNFkB_cytoplasm, Rate Law: nucleus*k2_e*IkBeNFkB_nucleus
deg5_c=6.0E-5 per_minReaction: IkBbNFkB_cytoplasm => NFkB_cytoplasm, Rate Law: cytoplasm*deg5_c*IkBbNFkB_cytoplasm
tr1a=0.2448 per_minReaction: IkBa_mRNA => IkBa_mRNA + IkBa_cytoplasm, Rate Law: nucleus*tr1a*IkBa_mRNA
deg1_c=0.12 per_minReaction: IkBa_cytoplasm =>, Rate Law: cytoplasm*deg1_c*IkBa_cytoplasm
a4_2=30.0 per_uM_per_min; d4_2=6.0E-5 per_minReaction: IkBa_nucleus + NFkB_nucleus => IkBaNFkB_nucleus, Rate Law: nucleus*(a4_2*IkBa_nucleus*NFkB_nucleus-d4_2*IkBaNFkB_nucleus)
tr1e=0.2448 per_minReaction: IkBe_mRNA => IkBe_mRNA + IkBe_cytoplasm, Rate Law: nucleus*tr1e*IkBe_mRNA
deg6_c=6.0E-5 per_minReaction: IkBeNFkB_cytoplasm => NFkB_cytoplasm, Rate Law: cytoplasm*deg6_c*IkBeNFkB_cytoplasm
tp1a=0.018 per_min; tp2a=0.012 per_minReaction: IkBa_cytoplasm => IkBa_nucleus, Rate Law: cytoplasm*tp1a*IkBa_cytoplasm-nucleus*tp2a*IkBa_nucleus
deg6_n=6.0E-5 per_minReaction: IkBeNFkB_nucleus => NFkB_nucleus, Rate Law: nucleus*deg6_n*IkBeNFkB_nucleus
r5=0.12 per_minReaction: IkBbIKKNFkB => NFkB_cytoplasm + IKK, Rate Law: cytoplasm*r5*IkBbIKKNFkB
tr3b=0.0168 per_minReaction: IkBb_mRNA =>, Rate Law: nucleus*tr3b*IkBb_mRNA
d2_2=0.105 per_min; a8=2.88 per_uM_per_minReaction: IkBbNFkB_cytoplasm + IKK => IkBbIKKNFkB, Rate Law: cytoplasm*(a8*IkBbNFkB_cytoplasm*IKK-d2_2*IkBbIKKNFkB)
deg3_n=0.18 per_minReaction: IkBe_nucleus =>, Rate Law: nucleus*deg3_n*IkBe_nucleus
a2=0.36 per_uM_per_min; d2_1=0.105 per_minReaction: IkBb_cytoplasm + IKK => IkBbIKK, Rate Law: cytoplasm*(a2*IkBb_cytoplasm*IKK-d2_1*IkBbIKK)
a1=1.35 per_uM_per_min; d1_1=0.075 per_minReaction: IkBa_cytoplasm + IKK => IkBaIKK, Rate Law: cytoplasm*(a1*IkBa_cytoplasm*IKK-d1_1*IkBaIKK)
r4=0.36 per_minReaction: IkBaIKKNFkB => NFkB_cytoplasm + IKK, Rate Law: cytoplasm*r4*IkBaIKKNFkB
deg5_n=6.0E-5 per_minReaction: IkBbNFkB_nucleus => NFkB_nucleus, Rate Law: nucleus*deg5_n*IkBbNFkB_nucleus
d5_3=6.0E-5 per_min; a5_3=30.0 per_uM_per_minReaction: IkBbIKK + NFkB_cytoplasm => IkBbIKKNFkB, Rate Law: cytoplasm*(a5_3*IkBbIKK*NFkB_cytoplasm-d5_3*IkBbIKKNFkB)
tr2a_i=1.98 per_uM_per_minReaction: => IkBa_mRNA; NFkB_nucleus, Rate Law: nucleus*tr2a_i*NFkB_nucleus^2
tr2e=3.048E-5 uM_per_minReaction: => IkBe_mRNA, Rate Law: nucleus*tr2e
d3_1=0.105 per_min; a3=0.54 per_uM_per_minReaction: IkBe_cytoplasm + IKK => IkBeIKK, Rate Law: cytoplasm*(a3*IkBe_cytoplasm*IKK-d3_1*IkBeIKK)
a5_1=30.0 per_uM_per_min; d5_1=6.0E-5 per_minReaction: IkBb_cytoplasm + NFkB_cytoplasm => IkBbNFkB_cytoplasm, Rate Law: cytoplasm*(a5_1*IkBb_cytoplasm*NFkB_cytoplasm-d5_1*IkBbNFkB_cytoplasm)
tp2b=0.012 per_min; tp1b=0.018 per_minReaction: IkBb_cytoplasm => IkBb_nucleus, Rate Law: cytoplasm*tp1b*IkBb_cytoplasm-nucleus*tp2b*IkBb_nucleus
r6=0.18 per_minReaction: IkBeIKKNFkB => NFkB_cytoplasm + IKK, Rate Law: cytoplasm*r6*IkBeIKKNFkB
r1=0.072 per_minReaction: IkBaIKK => IKK, Rate Law: cytoplasm*r1*IkBaIKK
d6_1=6.0E-5 per_min; a6_1=30.0 per_uM_per_minReaction: IkBe_cytoplasm + NFkB_cytoplasm => IkBeNFkB_cytoplasm, Rate Law: cytoplasm*(a6_1*IkBe_cytoplasm*NFkB_cytoplasm-d6_1*IkBeNFkB_cytoplasm)
d4_1=6.0E-5 per_min; a4_1=30.0 per_uM_per_minReaction: IkBa_cytoplasm + NFkB_cytoplasm => IkBaNFkB_cytoplasm, Rate Law: cytoplasm*(a4_1*IkBa_cytoplasm*NFkB_cytoplasm-d4_1*IkBaNFkB_cytoplasm)
d4_3=6.0E-5 per_min; a4_3=30.0 per_uM_per_minReaction: IkBaIKK + NFkB_cytoplasm => IkBaIKKNFkB, Rate Law: cytoplasm*(a4_3*IkBaIKK*NFkB_cytoplasm-d4_3*IkBaIKKNFkB)
a6_2=30.0 per_uM_per_min; d6_2=6.0E-5 per_minReaction: IkBe_nucleus + NFkB_nucleus => IkBeNFkB_nucleus, Rate Law: nucleus*(a6_2*IkBe_nucleus*NFkB_nucleus-d6_2*IkBeNFkB_nucleus)
a5_2=30.0 per_uM_per_min; d5_2=6.0E-5 per_minReaction: IkBb_nucleus + NFkB_nucleus => IkBbNFkB_nucleus, Rate Law: nucleus*(a5_2*IkBb_nucleus*NFkB_nucleus-d5_2*IkBbNFkB_nucleus)
tr3e=0.0168 per_minReaction: IkBe_mRNA =>, Rate Law: nucleus*tr3e*IkBe_mRNA
a7=11.1 per_uM_per_min; d1_2=0.075 per_minReaction: IkBaNFkB_cytoplasm + IKK => IkBaIKKNFkB, Rate Law: cytoplasm*(a7*IkBaNFkB_cytoplasm*IKK-d1_2*IkBaIKKNFkB)

States:

NameDescription
IkBaIKKNFkB[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B inhibitor alpha]
IkBeIKK[NF-kappa-B inhibitor epsilon; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha]
IkBa cytoplasm[NF-kappa-B inhibitor alpha]
IKK[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha]
IkBe cytoplasm[NF-kappa-B inhibitor epsilon]
IkBa mRNAIkBat
IkBeNFkB nucleus[NF-kappa-B inhibitor epsilon; Nuclear factor NF-kappa-B p105 subunit]
IkBb cytoplasm[NF-kappa-B inhibitor beta]
IkBaNFkB nucleus[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha]
IkBe nucleus[NF-kappa-B inhibitor epsilon]
IkBe mRNAIkBet
NFkB nucleus[Nuclear factor NF-kappa-B p105 subunit]
IkBbIKK[NF-kappa-B inhibitor beta; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha]
IkBbNFkB nucleus[NF-kappa-B inhibitor beta; Nuclear factor NF-kappa-B p105 subunit]
IkBaIKK[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor alpha]
IkBeNFkB cytoplasm[NF-kappa-B inhibitor epsilon; Nuclear factor NF-kappa-B p105 subunit]
IkBaNFkB cytoplasm[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha]
IkBa nucleus[NF-kappa-B inhibitor alpha]
NFkB cytoplasm[Nuclear factor NF-kappa-B p105 subunit]
IkBb nucleus[NF-kappa-B inhibitor beta]
IkBbNFkB cytoplasm[NF-kappa-B inhibitor beta; Nuclear factor NF-kappa-B p105 subunit]
IkBeIKKNFkB[NF-kappa-B inhibitor epsilon; Nuclear factor NF-kappa-B p105 subunit; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha]
IkBb mRNAIkBbt
IkBbIKKNFkB[NF-kappa-B inhibitor beta; Nuclear factor NF-kappa-B p105 subunit; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha]

Oh2007 - Genome-scale metabolic network of Bacillus subtilis (iYO844): MODEL1507180013v0.0.1

Oh2007 - Genome-scale metabolic network of Bacillus subtilis (iYO844)This model is described in the article: [Genome-sc…

Details

In this report, a genome-scale reconstruction of Bacillus subtilis metabolism and its iterative development based on the combination of genomic, biochemical, and physiological information and high-throughput phenotyping experiments is presented. The initial reconstruction was converted into an in silico model and expanded in a four-step iterative fashion. First, network gap analysis was used to identify 48 missing reactions that are needed for growth but were not found in the genome annotation. Second, the computed growth rates under aerobic conditions were compared with high-throughput phenotypic screen data, and the initial in silico model could predict the outcomes qualitatively in 140 of 271 cases considered. Detailed analysis of the incorrect predictions resulted in the addition of 75 reactions to the initial reconstruction, and 200 of 271 cases were correctly computed. Third, in silico computations of the growth phenotypes of knock-out strains were found to be consistent with experimental observations in 720 of 766 cases evaluated. Fourth, the integrated analysis of the large-scale substrate utilization and gene essentiality data with the genome-scale metabolic model revealed the requirement of 80 specific enzymes (transport, 53; intracellular reactions, 27) that were not in the genome annotation. Subsequent sequence analysis resulted in the identification of genes that could be putatively assigned to 13 intracellular enzymes. The final reconstruction accounted for 844 open reading frames and consisted of 1020 metabolic reactions and 988 metabolites. Hence, the in silico model can be used to obtain experimentally verifiable hypothesis on the metabolic functions of various genes. link: http://identifiers.org/pubmed/17573341

Okuonghae2020 - SEAIR model of COVID-19 transmission in Lagos, Nigeria: BIOMD0000000991v0.0.1

This work examines the impact of various non-pharmaceutical control measures (government and personal) on the population…

Details

This work examines the impact of various non-pharmaceutical control measures (government and personal) on the population dynamics of the novel coronavirus disease 2019 (COVID-19) in Lagos, Nigeria, using an appropriately formulated mathematical model. Using the available data, since its first reported case on 16 March 2020, we seek to develop a predicative tool for the cumulative number of reported cases and the number of active cases in Lagos; we also estimate the basic reproduction number of the disease outbreak in the aforementioned State in Nigeria. Using numerical simulations, we show the effect of control measures, specifically the common social distancing, use of face mask and case detection (via contact tracing and subsequent testings) on the dynamics of COVID-19. We also provide forecasts for the cumulative number of reported cases and active cases for different levels of the control measures being implemented. Numerical simulations of the model show that if at least 55% of the population comply with the social distancing regulation with about 55% of the population effectively making use of face masks while in public, the disease will eventually die out in the population and that, if we can step up the case detection rate for symptomatic individuals to about 0.8 per day, with about 55% of the population complying with the social distancing regulations, it will lead to a great decrease in the incidence (and prevalence) of COVID-19. link: http://identifiers.org/pubmed/32834593

Oliveira2005 - Genome-scale metabolic network of Lactococcus lactis (iAO358): MODEL1507180014v0.0.1

Oliveira2005 - Genome-scale metabolic network of Lactococcus lactis (iAO358)This model is described in the article: [Mo…

Details

BACKGROUND: Genome-scale flux models are useful tools to represent and analyze microbial metabolism. In this work we reconstructed the metabolic network of the lactic acid bacteria Lactococcus lactis and developed a genome-scale flux model able to simulate and analyze network capabilities and whole-cell function under aerobic and anaerobic continuous cultures. Flux balance analysis (FBA) and minimization of metabolic adjustment (MOMA) were used as modeling frameworks. RESULTS: The metabolic network was reconstructed using the annotated genome sequence from L. lactis ssp. lactis IL1403 together with physiological and biochemical information. The established network comprised a total of 621 reactions and 509 metabolites, representing the overall metabolism of L. lactis. Experimental data reported in the literature was used to fit the model to phenotypic observations. Regulatory constraints had to be included to simulate certain metabolic features, such as the shift from homo to heterolactic fermentation. A minimal medium for in silico growth was identified, indicating the requirement of four amino acids in addition to a sugar. Remarkably, de novo biosynthesis of four other amino acids was observed even when all amino acids were supplied, which is in good agreement with experimental observations. Additionally, enhanced metabolic engineering strategies for improved diacetyl producing strains were designed. CONCLUSION: The L. lactis metabolic network can now be used for a better understanding of lactococcal metabolic capabilities and potential, for the design of enhanced metabolic engineering strategies and for integration with other types of 'omic' data, to assist in finding new information on cellular organization and function. link: http://identifiers.org/pubmed/15982422

Oliveira2020 - Beta-Alanine pathway for 3-hydroxypropionate production from glucose: MODEL2010030002v0.0.1

Beta-Alanine pathway for 3-hydroxypropionate production from glucose.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Beta-Alanine pathway for 3-hydroxypropionate production from glycerol: MODEL2010030003v0.0.1

Beta-Alanine pathway for 3-hydroxypropionate production from glycerol.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Beta-Alanine pathway for Acrylic Acid production from glucose: MODEL2010040001v0.0.1

Beta-Alanine pathway for Acrylic Acid production from glucose.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Beta-Alanine pathway for Acrylic Acid production from glycerol: MODEL2010040002v0.0.1

Beta-Alanine pathway for Acrylic Acid production from glycerol.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - E. coli Extended Central Carbon Metabolism from Glycerol: MODEL2010160002v0.0.1

The kinetic model of E. coli central carbon metabolism developed by Millard et al. 2016 was extended to include the prod…

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Extended E. coli Central Carbon Metabolism from Glucose: MODEL2010030001v0.0.1

The kinetic model of E. coli central carbon metabolism developed by Millard et al. 2016 was extended to include the prod…

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Glycerol pathway for 3-hydroxypropionate production from glucose: MODEL2010030004v0.0.1

Glycerol pathway for 3-hydroxypropionate production from glucose.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Glycerol pathway for 3-hydroxypropionate production from glycerol: MODEL2010030005v0.0.1

Glycerol pathway for 3-hydroxypropionate production from glycerol.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Glycerol pathway for Acrylic Acid production from glucose: MODEL2010040003v0.0.1

Glycerol pathway for Acrylic Acid production from glucose.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Glycerol pathway for Acrylic Acid production from glycerol: MODEL2010040005v0.0.1

Glycerol pathway for Acrylic Acid production from glycerol.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Malonyl-CoA pathway for 3-hydroxypropionate production from glucose: MODEL2010030006v0.0.1

Malonyl-CoA pathway for 3-hydroxypropionate production from glucose.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Malonyl-CoA pathway for 3-hydroxypropionate production from glycerol: MODEL2010030008v0.0.1

Malonyl-CoA pathway for 3-hydroxypropionate production from glycerol.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Malonyl-CoA pathway for Acrylic Acid production from glucose: MODEL2010040006v0.0.1

Malonyl-CoA pathway for Acrylic Acid production from glucose.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Oliveira2020 - Malonyl-CoA pathway for Acrylic Acid production from glycerol: MODEL2010040007v0.0.1

Malonyl-CoA pathway for Acrylic Acid production from glycerol.

Details

Acrylic acid is a value-added chemical used in industry to produce diapers, coatings, paints, and adhesives, among many others. Due to its economic importance, there is currently a need for new and sustainable ways to synthesise it. Recently, the focus has been laid in the use of Escherichia coli to express the full bio-based pathway using 3-hydroxypropionate as an intermediary through three distinct pathways (glycerol, malonyl-CoA, and β-alanine). Hence, the goals of this work were to use COPASI software to assess which of the three pathways has a higher potential for industrial-scale production, from either glucose or glycerol, and identify potential targets to improve the biosynthetic pathways yields. When compared to the available literature, the models developed during this work successfully predict the production of 3-hydroxypropionate, using glycerol as carbon source in the glycerol pathway, and using glucose as a carbon source in the malonyl-CoA and β-alanine pathways. Finally, this work allowed to identify four potential over-expression targets (glycerol-3-phosphate dehydrogenase (G3pD), acetyl-CoA carboxylase (AccC), aspartate aminotransferase (AspAT), and aspartate carboxylase (AspC)) that should, theoretically, result in higher AA yields. link: http://identifiers.org/doi/10.1371/journal.pcbi.1008704

Olsen2003_neutrophil_oscillatory_metabolism: BIOMD0000000143v0.0.1

Olsen2003_neutrophil_oscillatory_metabolismThis model is described in the article: [A model of the oscillatory metaboli…

Details

We present a two-compartment model to explain the oscillatory behavior observed experimentally in activated neutrophils. Our model is based mainly on the peroxidase-oxidase reaction catalyzed by myeloperoxidase with melatonin as a cofactor and NADPH oxidase, a major protein in the phagosome membrane of the leukocyte. The model predicts that after activation of a neutrophil, an increase in the activity of the hexose monophosphate shunt and the delivery of myeloperoxidase into the phagosome results in oscillations in oxygen and NAD(P)H concentration. The period of oscillation changes from >200 s to 10-30 s. The model is consistent with previously reported oscillations in cell metabolism and oxidant production. Key features and predictions of the model were confirmed experimentally. The requirement of the hexose monophosphate pathway for 10 s oscillations was verified using 6-aminonicotinamide and dexamethasone, which are inhibitors of glucose-6-phosphate dehydrogenase. The role of the NADPH oxidase in promoting oscillations was confirmed by dose-response studies of the effect of diphenylene iodonium, an inhibitor of the NADPH oxidase. Moreover, the model predicted an increase in the amplitude of NADPH oscillations in the presence of melatonin, which was confirmed experimentally. Successful computer modeling of complex chemical dynamics within cells and their chemical perturbation will enhance our ability to identify new antiinflammatory compounds. link: http://identifiers.org/pubmed/12524266

Parameters:

NameDescription
k10 = 10.0Reaction: NADPH_c + MLT_c => NADP_c + MLTH_c, Rate Law: cytoplasm*k10*MLT_c*NADPH_c
k11 = 60.0Reaction: NADP_c => NADP2_c, Rate Law: cytoplasm*k11*NADP_c^2
k14 = 30.0Reaction: O2_p => O2_c, Rate Law: phagosome*(k14*O2_p-k14*O2_c)
k5 = 10.0Reaction: O2minus_p + H_p => O2_p + H2O2_p, Rate Law: phagosome*k5*O2minus_p^2
k16 = 10.0Reaction: MLTH_p => MLTH_c, Rate Law: phagosome*(k16*MLTH_p-k16*MLTH_c)
k2 = 10.0Reaction: MLTH_p + coI_p => MLT_p + coII_p, Rate Law: phagosome*k2*coI_p*MLTH_p
k17 = 10.0Reaction: MLT_p => MLT_c, Rate Law: phagosome*(k17*MLT_p-k17*MLT_c)
k18 = 2.0Reaction: O2minus_p => O2minus_c, Rate Law: phagosome*(k18*O2minus_p-k18*O2minus_c)
k8 = 50.0Reaction: O2_c + NADP_c => O2minus_c + NADPplus_c, Rate Law: cytoplasm*k8*NADP_c*O2_c
k9 = 500.0Reaction: O2minus_c + H_c => O2_c + H2O2_c, Rate Law: cytoplasm*k9*O2minus_c^2
k13 = 12.5Reaction: => O2_c, Rate Law: cytoplasm*k13
k4 = 20.0Reaction: O2minus_p + per3_p => coIII_p, Rate Law: phagosome*k4*per3_p*O2minus_p
k1 = 50.0; kminus1 = 58.0Reaction: per3_p + H2O2_p => coI_p, Rate Law: phagosome*(k1*H2O2_p*per3_p-kminus1*coI_p)
k3 = 0.004Reaction: MLTH_p + coII_p => MLT_p + per3_p, Rate Law: phagosome*k3*coII_p*MLTH_p
V = 288.0; Knadph = 60.0; Ko = 1.5; L = 550.0Reaction: O2_p + NADPH_c => O2minus_p + NADPplus_c, Rate Law: phagosome*V*NADPH_c/Knadph*(1+NADPH_c/Knadph)*O2_p/((L+(1+NADPH_c/Knadph)^2)*(Ko+O2_p))
k7 = 1.0E-6Reaction: O2_c + NADPH_c => H2O2_c + NADPplus_c, Rate Law: cytoplasm*k7*NADPH_c*O2_c
k15 = 30.0Reaction: H2O2_p => H2O2_c, Rate Law: phagosome*(k15*H2O2_p-k15*H2O2_c)
k6 = 0.1Reaction: O2minus_p + coIII_p => O2_p + coI_p, Rate Law: phagosome*k6*coIII_p*O2minus_p
k12 = 25.0Reaction: => NADPH_c, Rate Law: cytoplasm*k12
kminus13 = 0.045Reaction: O2_c =>, Rate Law: cytoplasm*kminus13*O2_c

States:

NameDescription
H p[dihydrogen; Hydrogen]
O2minus p[superoxide; O2.-]
H c[dihydrogen; Hydrogen]
MLTH c[melatonin; Melatonin]
O2 c[dioxygen; Oxygen]
O2minus c[superoxide; O2.-]
NADP2 c[NADP(+); NADP+]
H2O2 p[hydrogen peroxide; Hydrogen peroxide]
NADP c[NADP(+); NADP+]
NADPH c[NADPH; NADPH]
per3 p[IPR000823; PIRSF000293]
MLT c[melatonin; Melatonin]
MLTH p[melatonin; Melatonin]
MLT p[melatonin; Melatonin]
coIII p[IPR000823]
NADPplus c[NADP(+); NADP+]
coII p[IPR000823; PIRSF000293]
O2 p[dioxygen; Oxygen]
H2O2 c[hydrogen peroxide; Hydrogen peroxide]
coI p[IPR000823; PIRSF000293]

Olsen2003_peroxidase: BIOMD0000000046v0.0.1

Notes of the BioModels curators: The current model reproduce the figure 7, panel B of the paper. Note that there is a t…

Details

The peroxidase-oxidase reaction is known to involve reactive oxygen species as intermediates. These intermediates inactivate many types of biomolecules, including peroxidase itself. Previously, we have shown that oscillatory dynamics in the peroxidase-oxidase reaction seem to protect the enzyme from inactivation. It was suggested that this is due to a lower average concentration of reactive oxygen species in the oscillatory state compared to the steady state. Here, we studied the peroxidase-oxidase reaction with either 4-hydroxybenzoic acid or melatonin as cofactors. We show that the protective effect of oscillatory dynamics is present in both cases. We also found that the enzyme degradation depends on the concentration of the cofactor and on the pH of the reaction mixture. We simulated the oscillatory behaviour, including the oscillation/steady state bistability observed experimentally, using a detailed reaction scheme. The computational results confirm the hypothesis that protection is due to lower average concentrations of superoxide radical during oscillations. They also show that the shape of the oscillations changes with increasing cofactor concentration resulting in a further decrease in the average concentration of radicals. We therefore hypothesize that the protective effect of oscillatory dynamics is a general effect in this system. link: http://identifiers.org/pubmed/12823550

Parameters:

NameDescription
k8=40.0Reaction: NADrad + coIII => NAD + coI, Rate Law: compartment*k8*coIII*NADrad
k2=18.0Reaction: per3 + H2O2 => coI, Rate Law: compartment*k2*H2O2*per3
k6=17.0Reaction: per3 + super => coIII, Rate Law: compartment*k6*super*per3
k5=20.0Reaction: NADrad + O2 => NAD + super, Rate Law: compartment*k5*NADrad*O2
k9=60.0Reaction: NADrad => NAD2, Rate Law: compartment*k9*NADrad*NADrad
k1=3.0E-6Reaction: NADH + O2 => H2O2 + NAD, Rate Law: compartment*k1*NADH*O2
k7=20.0Reaction: super => H2O2 + O2, Rate Law: compartment*k7*super*super
k13b=0.006Reaction: O2 => O2g, Rate Law: compartment*k13b*O2
k10=1.8Reaction: per3 + NADrad => per2 + NAD, Rate Law: compartment*k10*per3*NADrad
k13f=0.006Reaction: O2g => O2, Rate Law: compartment*k13f*O2g
k3=0.15Reaction: ArH + coI => Ar + coII, Rate Law: compartment*k3*coI*ArH
k12=0.08Reaction: NADHres => NADH, Rate Law: compartment*k12
k11=0.1Reaction: per2 + O2 => coIII, Rate Law: compartment*k11*per2*O2
k14=0.7Reaction: NADH + Ar => NADrad + ArH, Rate Law: compartment*k14*Ar*NADH
k4=0.0052Reaction: coII + ArH => per3 + Ar, Rate Law: compartment*k4*coII*ArH

States:

NameDescription
O2g[dioxygen; Oxygen]
NADH[NADH; NADH]
coI[IPR000823; PIRSF000293]
per2[IPR000823; PIRSF000293]
NAD2[NAD(+); NAD+]
ArH[4-hydroxybenzoic acid; melatonin; 4-Hydroxybenzoate; Melatonin]
per3[IPR000823; PIRSF000293]
NADrad[NAD(+); NAD+]
Ar[4-hydroxybenzoate; melatonin; 4-Hydroxybenzoate; Melatonin]
coIII[IPR000823; PIRSF000293]
coII[IPR000823; PIRSF000293]
NADHres[NADH; NADH]
NAD[NAD(+); NAD+]
H2O2[hydrogen peroxide; Hydrogen peroxide]
super[superoxide; O2.-]
O2[dioxygen; Oxygen]

Ontah2019 - Dynamic analysis of a tumor treatment model using oncolytic virus and chemotherapy with saturated infection rate: BIOMD0000000877v0.0.1

This is a mathematical model describing the treatment of tumors using oncolytic virus and chemotherapy. The model is com…

Details

Virotherapy is one of the most promising therapies in the treatment of tumors which may be further combined with chemotherapy to accelerate the healing rate. In this article, we propose a mathematical model for the treatment of tumors using oncolytic virus and chemotherapy. This model takes the form of nonlinear ordinary differential equations describing the interactions between uninfected tumor cells, infected tumor cells, an oncolytic virus, and chemotherapy. It is assumed that the rate of infection between uninfected tumor cells and infected tumor cells is in a saturated form. The saturation effect takes into account the fact that the number of contacts between them reaches the maximum value when the immune system works to stop the virus. The dynamical analysis, which includes the existence of equilibrium points, and its stability analysis is investigated. The analysis result shows that the system has three equilibrium points: tumor-free equilibrium point, virus-free equilibrium point and endemic equilibrium point. It is proven that these equilibrium points are conditionally stable. The numerical simulations show the successful combination of chemotherapy and virotherapy using an oncolytic virus in eliminating the tumor cells. link: http://identifiers.org/doi/10.1088/1757-899X/546/3/032025

Parameters:

NameDescription
delta_u = 50.0; K_c = 10000.0Reaction: U => ; C, Rate Law: compartment*delta_u*U*C/(K_c+C)
delta = 0.5Reaction: I =>, Rate Law: compartment*delta*I
psi = 4.17Reaction: C =>, Rate Law: compartment*psi*C
mu = 150.0Reaction: => C, Rate Law: compartment*mu
delta = 0.5; b = 0.5Reaction: => V; I, Rate Law: compartment*b*delta*I
K_c = 10000.0; delta_i = 60.0Reaction: I => ; C, Rate Law: compartment*delta_i*I*C/(K_c+C)
K = 2139.0; r = 0.1Reaction: => U; I, Rate Law: compartment*r*U*(1-(U+I)/K)
gamma = 0.1Reaction: V =>, Rate Law: compartment*gamma*V
beta = 0.01; alpha = 0.5Reaction: U + V => I, Rate Law: compartment*beta*U*V/(U+I+alpha)

States:

NameDescription
U[neoplastic cell]
I[infected cell; neoplastic cell]
C[C15681]
V[Oncolytic Virus]

Orfeo2010 - Simulating the effects of fondaparinux and Rivaroxaban on thrombin generation: MODEL1807240001v0.0.1

Reused mathematical model (Hockin et al., 2002) of blood coagulation simulating the effects of coagulation factor inhibi…

Details

Therapeutic agents that regulate blood coagulation are critical to the management of thrombotic disorders, with the selective targeting of factor (F) Xa emerging as a promising approach.To assess anticoagulant strategies targeting FXa.A deterministic computational model of tissue factor (Tf)-initiated thrombin generation and two empirical experimental systems (a synthetic coagulation proteome reconstruction using purified proteins and a whole blood model) were used to evaluate clinically relevant examples of the two available types of FXa-directed anticoagulants [an antithrombin (AT)-dependent agent, fondaparinux, and an AT-independent inhibitor, Rivaroxaban] in experimental regimens relevant to long-term (suppression of new Tf-initiated events) and acute (suppression of ongoing coagulation processes) clinical applications.Computational representations of each anticoagulant's efficacy in suppressing thrombin generation over a range of anticoagulant concentrations in both anticoagulation regimens were validated by results from corresponding empirical reconstructions and were consistent with those recommended for long-term and acute clinical applications, respectively. All three model systems suggested that Rivaroxaban would prove more effective in the suppression of an ongoing coagulation process than fondaparinux, reflecting its much higher reactivity toward the prothrombinase complex.The success of fondaparinux in acute settings in vivo is not explained solely by its properties as an FXa inhibitor. We have reported that FIXa contributes to the long-term capacity of clot-associated catalysts to restart a coagulation process, suggesting that the enhanced anti-FIXa activity of fondaparinux-AT may be critical to its success in acute settings in vivo. link: http://identifiers.org/pubmed/20492473

Ortega2006 - bistability from double phosphorylation in signal transduction: BIOMD0000000258v0.0.1

Ortega2006 - bistability from double phosphorylation in signal transductionThis model is described in the article: [Bis…

Details

Previous studies have suggested that positive feedback loops and ultrasensitivity are prerequisites for bistability in covalent modification cascades. However, it was recently shown that bistability and hysteresis can also arise solely from multisite phosphorylation. Here we analytically demonstrate that double phosphorylation of a protein (or other covalent modification) generates bistability only if: (a) the two phosphorylation (or the two dephosphorylation) reactions are catalyzed by the same enzyme; (b) the kinetics operate at least partly in the zero-order region; and (c) the ratio of the catalytic constants of the phosphorylation and dephosphorylation steps in the first modification cycle is less than this ratio in the second cycle. We also show that multisite phosphorylation enlarges the region of kinetic parameter values in which bistability appears, but does not generate multistability. In addition, we conclude that a cascade of phosphorylation/dephosphorylation cycles generates multiple steady states in the absence of feedback or feedforward loops. Our results show that bistable behavior in covalent modification cascades relies not only on the structure and regulatory pattern of feedback/feedforward loops, but also on the kinetic characteristics of their component proteins. link: http://identifiers.org/pubmed/16934033

Parameters:

NameDescription
Ks4 = 0.01; r24 = 1.0; Chi14 = 1.1; Ks2 = 0.01; Vm1 = 1.0Reaction: beta => alpha; gamma, Rate Law: r24*Vm1/Chi14*beta/Ks2/(1+gamma/Ks4+beta/Ks2)
Ks3 = 0.01; r31 = 1.0; Ks1 = 0.01; Vm1 = 1.0Reaction: beta => gamma; alpha, Rate Law: r31*Vm1*beta/Ks3/(1+alpha/Ks1+beta/Ks3)
Ks4 = 0.01; Chi14 = 1.1; Ks2 = 0.01; Vm1 = 1.0Reaction: gamma => beta, Rate Law: Vm1/Chi14*gamma/Ks4/(1+gamma/Ks4+beta/Ks2)
Ks3 = 0.01; Ks1 = 0.01; Vm1 = 1.0Reaction: alpha => beta, Rate Law: Vm1*alpha/Ks1/(1+alpha/Ks1+beta/Ks3)

States:

NameDescription
alpha[Protein; protein polypeptide chain]
gamma[Phosphoprotein]
beta[Phosphoprotein]

Ortega2013 - Interplay between secretases determines biphasic amyloid-beta level: BIOMD0000000556v0.0.1

Ortega2013 - Interplay between secretases determines biphasic amyloid-beta levelThis model is described in the article:…

Details

Amyloid-β (Aβ) is produced by the consecutive cleavage of amyloid precursor protein (APP) first by β-secretase, generating C99, and then by γ-secretase. APP is also cleaved by α-secretase. It is hypothesized that reducing the production of Aβ in the brain may slow the progression of Alzheimer disease. Therefore, different γ-secretase inhibitors have been developed to reduce Aβ production. Paradoxically, it has been shown that low to moderate inhibitor concentrations cause a rise in Aβ production in different cell lines, in different animal models, and also in humans. A mechanistic understanding of the Aβ rise remains elusive. Here, a minimal mathematical model has been developed that quantitatively describes the Aβ dynamics in cell lines that exhibit the rise as well as in cell lines that do not. The model includes steps of APP processing through both the so-called amyloidogenic pathway and the so-called non-amyloidogenic pathway. It is shown that the cross-talk between these two pathways accounts for the increase in Aβ production in response to inhibitor, i.e. an increase in C99 will inhibit the non-amyloidogenic pathway, redirecting APP to be cleaved by β-secretase, leading to an additional increase in C99 that overcomes the loss in γ-secretase activity. With a minor extension, the model also describes plasma Aβ profiles observed in humans upon dosing with a γ-secretase inhibitor. In conclusion, this mechanistic model rationalizes a series of experimental results that spans from in vitro to in vivo and to humans. This has important implications for the development of drugs targeting Aβ production in Alzheimer disease. link: http://identifiers.org/pubmed/23152503

Parameters:

NameDescription
v0 = 1.0Reaction: => APP, Rate Law: Brain*v0
vm3 = 14.6; km4 = 0.915; km3 = 28.8Reaction: C83 => p3; C99, C83, C99, Rate Law: Brain*vm3*C83/km3/(1+C83/km3+C99/km4)
kic = 0.173; km4 = 0.915; den = 1.0; vm4 = 1.71Reaction: C99 => AB; X, X, C99, Rate Law: Brain*vm4/(1+X/kic)*C99/km4/den
km4 = 0.915; km3 = 28.8; vm4 = 1.71Reaction: C99 => AB; C83, C99, C83, Rate Law: Brain*vm4*C99/km4/(1+C99/km4+C83/km3)
km1 = 0.186; km5 = 0.0672; vm1 = 1.1Reaction: APP => C83; C99, APP, C99, Rate Law: Brain*vm1*APP/km1/(1+APP/km1+C99/km5)
km5 = 0.0672; km1 = 0.186; vm5 = 0.0223Reaction: C99 => C83; APP, C99, APP, Rate Law: Brain*vm5*C99/km5/(1+C99/km5+APP/km1)
vm3 = 14.6; kic = 0.173; den = 1.0; km3 = 28.8Reaction: C83 => p3; X, X, C83, Rate Law: Brain*vm3/(1+X/kic)*C83/km3/den
km2 = 1.64; vm2 = 0.153Reaction: APP => C99; APP, Rate Law: Brain*vm2*APP/km2/(1+APP/km2)

States:

NameDescription
C83[Amyloid beta A4 protein]
C99[Amyloid beta A4 protein]
p3[Amyloid beta A4 protein]
APP[Amyloid beta A4 protein]
AB[Amyloid beta A4 protein]

Orth2011_E.coli_MetabolicNetwork: MODEL1108160000v0.0.1

This model is from the article: A comprehensive genome-scale reconstruction of Escherichia coli metabolism-2011. Ort…

Details

The initial genome-scale reconstruction of the metabolic network of Escherichia coli K-12 MG1655 was assembled in 2000. It has been updated and periodically released since then based on new and curated genomic and biochemical knowledge. An update has now been built, named iJO1366, which accounts for 1366 genes, 2251 metabolic reactions, and 1136 unique metabolites. iJO1366 was (1) updated in part using a new experimental screen of 1075 gene knockout strains, illuminating cases where alternative pathways and isozymes are yet to be discovered, (2) compared with its predecessor and to experimental data sets to confirm that it continues to make accurate phenotypic predictions of growth on different substrates and for gene knockout strains, and (3) mapped to the genomes of all available sequenced E. coli strains, including pathogens, leading to the identification of hundreds of unannotated genes in these organisms. Like its predecessors, the iJO1366 reconstruction is expected to be widely deployed for studying the systems biology of E. coli and for metabolic engineering applications. link: http://identifiers.org/pubmed/21988831

Orton2009 - Modelling cancerous mutations in the EGFR/ERK pathway - EGF Model: BIOMD0000000623v0.0.1

Orton2009 - Modelling cancerous mutations in the EGFR/ERK pathway - EGF ModelThis model studies the aberrations in ERK s…

Details

The Epidermal Growth Factor Receptor (EGFR) activated Extracellular-signal Regulated Kinase (ERK) pathway is a critical cell signalling pathway that relays the signal for a cell to proliferate from the plasma membrane to the nucleus. Deregulation of the EGFR/ERK pathway due to alterations affecting the expression or function of a number of pathway components has long been associated with numerous forms of cancer. Under normal conditions, Epidermal Growth Factor (EGF) stimulates a rapid but transient activation of ERK as the signal is rapidly shutdown. Whereas, under cancerous mutation conditions the ERK signal cannot be shutdown and is sustained resulting in the constitutive activation of ERK and continual cell proliferation. In this study, we have used computational modelling techniques to investigate what effects various cancerous alterations have on the signalling flow through the ERK pathway.We have generated a new model of the EGFR activated ERK pathway, which was verified by our own experimental data. We then altered our model to represent various cancerous situations such as Ras, B-Raf and EGFR mutations, as well as EGFR overexpression. Analysis of the models showed that different cancerous situations resulted in different signalling patterns through the ERK pathway, especially when compared to the normal EGF signal pattern. Our model predicts that cancerous EGFR mutation and overexpression signals almost exclusively via the Rap1 pathway, predicting that this pathway is the best target for drugs. Furthermore, our model also highlights the importance of receptor degradation in normal and cancerous EGFR signalling, and suggests that receptor degradation is a key difference between the signalling from the EGF and Nerve Growth Factor (NGF) receptors.Our results suggest that different routes to ERK activation are being utilised in different cancerous situations which therefore has interesting implications for drug selection strategies. We also conducted a comparison of the critical differences between signalling from different growth factor receptors (namely EGFR, mutated EGFR, NGF, and Insulin) with our results suggesting the difference between the systems are large scale and can be attributed to the presence/absence of entire pathways rather than subtle difference in individual rate constants between the systems. link: http://identifiers.org/pubmed/19804630

Parameters:

NameDescription
kcat=185.759; km=4768350.0Reaction: species_9 => species_8; species_23, Rate Law: compartment_0*kcat*species_23*species_9/(km+species_9)
Kcat=8.8912; km=3496490.0Reaction: species_10 => species_11; species_26, Rate Law: compartment_0*Kcat*species_26*species_10/(km+species_10)
km=62464.6; Kcat=0.884096Reaction: species_7 => species_6; species_4, Rate Law: compartment_0*Kcat*species_4*species_7/(km+species_7)
Kcat=10.6737; km=184912.0Reaction: species_15 => species_14; species_0, Rate Law: compartment_0*Kcat*species_0*species_15/(km+species_15)
Kcat=0.126329; km=1061.71Reaction: species_6 => species_7; species_27, Rate Law: compartment_0*Kcat*species_27*species_6/(km+species_6)
Kcat=2.83243; km=518753.0Reaction: species_8 => species_9; species_26, Rate Law: compartment_0*Kcat*species_26*species_8/(km+species_8)
Kcat=0.0213697; km=763523.0Reaction: species_13 => species_12; species_10, Rate Law: compartment_0*Kcat*species_10*species_13/(km+species_13)
v=100.0Reaction: species_30 => species_1, Rate Law: compartment_0*v
km=896896.0; Kcat=1611.97Reaction: species_2 => species_3; species_12, Rate Law: compartment_0*Kcat*species_12*species_2/(km+species_2)
kcat=0.884096; km=62464.6Reaction: species_24 => species_23; species_4, Rate Law: compartment_0*kcat*species_4*species_24/(km+species_24)
Kcat=185.759; km=4768350.0Reaction: species_9 => species_8; species_6, Rate Law: compartment_0*Kcat*species_6*species_9/(km+species_9)
k1=2.18503E-5; k2=0.121008Reaction: species_25 + species_1 => species_0, Rate Law: compartment_0*(k1*species_25*species_1-k2*species_0)
km=1432410.0; Kcat=1509.36Reaction: species_4 => species_5; species_28, Rate Law: compartment_0*Kcat*species_28*species_4/(km+species_4)
km=35954.3; kcat=32.344Reaction: species_22 => species_21; species_19, Rate Law: compartment_0*kcat*species_19*species_22/(km+species_22)
Kcat=32.344; km=35954.3Reaction: species_5 => species_4; species_2, Rate Law: compartment_0*Kcat*species_2*species_5/(km+species_5)
Kcat=9.85367; km=1007340.0Reaction: species_11 => species_10; species_8, Rate Law: compartment_0*Kcat*species_8*species_11/(km+species_11)
k1=2.5Reaction: species_2 => species_3, Rate Law: compartment_0*k1*species_2
k1=0.2Reaction: species_0 => species_18, Rate Law: compartment_0*k1*species_0
kcat=1509.36; km=1432410.0Reaction: species_21 => species_22; species_29, Rate Law: compartment_0*kcat*species_29*species_21/(km+species_21)
km=1061.71; kcat=0.126329Reaction: species_23 => species_24; species_27, Rate Law: compartment_0*kcat*species_27*species_23/(km+species_23)
k1=0.005Reaction: species_12 => species_13, Rate Law: compartment_0*k1*species_12
km=6086070.0; Kcat=694.731Reaction: species_3 => species_2; species_0, Rate Law: compartment_0*Kcat*species_0*species_3/(km+species_3)
Kcat=15.1212; km=119355.0Reaction: species_6 => species_7; species_16, Rate Law: compartment_0*Kcat*species_16*species_6/(km+species_6)
Kcat=0.0566279; km=653951.0Reaction: species_17 => species_16; species_14, Rate Law: compartment_0*Kcat*species_14*species_17/(km+species_17)
k1=0.00125Reaction: species_1 => species_18, Rate Law: compartment_0*k1*species_1
Kcat=0.0771067; km=272056.0Reaction: species_15 => species_14; species_4, Rate Law: compartment_0*Kcat*species_4*species_15/(km+species_15)
kcat=694.731; km=6086070.0Reaction: species_20 => species_19; species_0, Rate Law: compartment_0*kcat*species_0*species_20/(km+species_20)

States:

NameDescription
species 9[Dual specificity mitogen-activated protein kinase kinase 1; Dual specificity mitogen-activated protein kinase kinase 2]
species 1[Receptor protein-tyrosine kinase]
species 18[Receptor protein-tyrosine kinase]
species 4[IPR020849]
species 16[RAC-alpha serine/threonine-protein kinase; RAC-beta serine/threonine-protein kinase; RAC-gamma serine/threonine-protein kinase]
species 20[C3G protein]
species 0[Receptor protein-tyrosine kinase]
species 21[Ras-related protein Rap-1b]
species 8[Dual specificity mitogen-activated protein kinase kinase 2; Dual specificity mitogen-activated protein kinase kinase 1]
species 17[RAC-gamma serine/threonine-protein kinase; RAC-beta serine/threonine-protein kinase; RAC-alpha serine/threonine-protein kinase]
species 12[p90 S6 kinase; Ribosomal protein S6 kinase alpha-1]
species 25[Pro-epidermal growth factor]
species 5[IPR020849]
species 15[Phosphatidylinositol 3-kinase regulatory subunit alpha]
species 30[Receptor protein-tyrosine kinase]
species 2[Son of sevenless 1]
species 6[RAF proto-oncogene serine/threonine-protein kinase]
species 19[C3G protein]
species 10[Mitogen-activated protein kinase 1; Mitogen-activated protein kinase 3]
species 11[Mitogen-activated protein kinase 3; Mitogen-activated protein kinase 1]
species 24[B-Raf proto-oncogene, serine/threonine kinase]
species 14[Phosphatidylinositol 3-kinase regulatory subunit alpha]
species 22[Ras-related protein Rap-1b]
species 3[Son of sevenless 1]
species 23[B-Raf proto-oncogene, serine/threonine kinase]
species 7[RAF proto-oncogene serine/threonine-protein kinase]
species 13[p90 S6 kinase; Ribosomal protein S6 kinase alpha-1]

Ota2015 - Positive regulation of Rho GTPase activity by RhoGDIs as a result of their direct interaction with GAPs (GDI integrated): BIOMD0000000899v0.0.1

This is a ordinary differential equation mathematical model describing the Rho GTPase cycle in which Rho GDP-dissociatio…

Details

Rho GTPases function as molecular switches in many different signaling pathways and control a wide range of cellular processes. Rho GDP-dissociation inhibitors (RhoGDIs) regulate Rho GTPase signaling and can function as both negative and positive regulators. The role of RhoGDIs as negative regulators of Rho GTPase signaling has been extensively investigated; however, little is known about how RhoGDIs act as positive regulators. Furthermore, it is unclear how this opposing role of GDIs influences the Rho GTPase cycle. We constructed ordinary differential equation models of the Rho GTPase cycle in which RhoGDIs inhibit the regulatory activities of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) by interacting with them directly as well as by sequestering the Rho GTPases. Using this model, we analyzed the role of RhoGDIs in Rho GTPase signaling.The model constructed in this study showed that the functions of GEFs and GAPs are integrated into Rho GTPase signaling through the interactions of these regulators with GDIs, and that the negative role of GDIs is to suppress the overall Rho activity by inhibiting GEFs. Furthermore, the positive role of GDIs is to sustain Rho activation by inhibiting GAPs under certain conditions. The interconversion between transient and sustained Rho activation occurs mainly through changes in the affinities of GDIs to GAPs and the concentrations of GAPs.RhoGDIs positively regulate Rho GTPase signaling primarily by interacting with GAPs and may participate in the switching between transient and sustained signals of the Rho GTPases. These findings enhance our understanding of the physiological roles of RhoGDIs and Rho GTPase signaling. link: http://identifiers.org/pubmed/25628036

Parameters:

NameDescription
kcatGAP=95.9; KmGAPGDI=0.1; KmGAPRho=4.48Reaction: s6 => s5; s8, s7, Rate Law: default*Function_for_re5(KmGAPGDI, KmGAPRho, default, kcatGAP, s6, s7, s8)
k1=1.0Reaction: s3 => s4; s1, Rate Law: default*Function_for_re1(default, k1, s1, s3)
k2=0.1Reaction: s4 => s3, Rate Law: default*Function_for_re2(default, k2, s4)
kcatGEF=5.64; KmGEFRho=24.5; KmGEFGDI=1.0Reaction: s5 => s6; s4, s7, Rate Law: default*Function_for_re4(KmGEFGDI, KmGEFRho, default, kcatGEF, s4, s5, s7)
k5=0.05; k4=0.5Reaction: s5 + s7 => s10, Rate Law: default*Function_for_re6(default, k4, k5, s10, s5, s7)
k7=0.05; k6=0.5Reaction: s7 + s6 => s13, Rate Law: default*Function_for_re7(default, k6, k7, s13, s6, s7)
k3=0.5Reaction: s1 => s2, Rate Law: default*Function_for_re3(default, k3, s1)
k9=0.18; k8=28.2Reaction: s6 + s9 => s16, Rate Law: default*Function_for_re8(default, k8, k9, s16, s6, s9)

States:

NameDescription
s1[C154897]
s5[PR:000000122]
s7GDI
s13[PR:000004242; PR:000000122]
s2[C154897; C154407]
s4[Active]
s9[effector]
s16[effector; PR:000000122]
s10[PR:000000122; PR:000004242]
s6[PR:000000122]
s3[C17494]

Ouyang2014 - photomorphogenic UV-B signalling network: BIOMD0000000545v0.0.1

Ouyang2014 - photomorphogenic UV-B signalling networkThis model is described in the article: [Coordinated photomorphoge…

Details

Long-wavelength and low-fluence UV-B light is an informational signal known to induce photomorphogenic development in plants. Using the model plant Arabidopsis thaliana, a variety of factors involved in UV-B-specific signaling have been experimentally characterized over the past decade, including the UV-B light receptor UV resistance locus 8; the positive regulators constitutive photomorphogenesis 1 and elongated hypocotyl 5; and the negative regulators cullin4, repressor of UV-B photomorphogenesis 1 (RUP1), and RUP2. Individual genetic and molecular studies have revealed that these proteins function in either positive or negative regulatory capacities for the sufficient and balanced transduction of photomorphogenic UV-B signal. Less is known, however, regarding how these signaling events are systematically linked. In our study, we use a systems biology approach to investigate the dynamic behaviors and correlations of multiple signaling components involved in Arabidopsis UV-B-induced photomorphogenesis. We define a mathematical representation of photomorphogenic UV-B signaling at a temporal scale. Supplemented with experimental validation, our computational modeling demonstrates the functional interaction that occurs among different protein complexes in early and prolonged response to photomorphogenic UV-B. link: http://identifiers.org/pubmed/25049395

Parameters:

NameDescription
kd3 = 0.5508Reaction: UR => UVR8D + RUP; UR, Rate Law: kd3*UR^2
ks2 = 4.0526; UV = 1.0; kdr2 = 0.2118Reaction: => RUP; UCS, UCS, RUP, Rate Law: ks2*(1+UV*UCS)-kdr2*RUP
k2 = 161.62Reaction: UVR8D => UVR8M; UVR8D, Rate Law: k2*UVR8D
UV = 1.0; ksr = 0.7537; kdr3a = 0.9735; ks3 = 0.4397; n2 = 2.0; kdr3b = 0.406; kdr3 = 1.246Reaction: => HY5; CDCS, CDW, UCS, CDCS, CDW, UCS, HY5, Rate Law: ks3*(1+n2*UV)-kdr3*((CDCS/(kdr3a+CDCS)+CDW/(kdr3b+CDW))-UCS/(ksr+UCS))*HY5
kd1 = 94.3524; ka1 = 0.0372Reaction: CS + UVR8M => UCS; CS, UVR8M, UCS, Rate Law: ka1*CS^2*UVR8M^2-kd1*UCS
ka3 = 4.7207Reaction: UVR8M + RUP => UR; UVR8M, RUP, Rate Law: ka3*UVR8M*RUP
kd2 = 50.6973; ka2 = 0.0611Reaction: CS + CD => CDCS; CS, CD, CDCS, Rate Law: ka2*CS^2*CD-kd2*CDCS
UV = 1.0; kdr1 = 0.1; n3 = 3.5; ks1 = 0.23; n1 = 3.0Reaction: => CS; HY5, FHY3, HY5, FHY3, CS, Rate Law: ks1*(1+UV*n3*(HY5+FHY3))-kdr1*(1+n1*UV)*CS
k1 = 0.0043Reaction: UVR8M => UVR8D; UVR8M, Rate Law: k1*UVR8M^2
kd4 = 1.1999; ka4 = 10.1285Reaction: CD + DWD => CDW; CD, DWD, CDW, Rate Law: ka4*CD*DWD-kd4*CDW

States:

NameDescription
RUP[WD repeat-containing protein RUP1]
UVR8M[Ultraviolet-B receptor UVR8]
HY5[Transcription factor HY5]
DWD[IPR001680]
CDW[Cullin-4; DDB1- and CUL4-associated factor homolog 1; IPR001680]
UVR8D[Ultraviolet-B receptor UVR8]
CDCS[E3 ubiquitin-protein ligase COP1; Protein SUPPRESSOR OF PHYA-105 1; DDB1- and CUL4-associated factor homolog 1; Cullin-4]
UCS[Protein SUPPRESSOR OF PHYA-105 1; E3 ubiquitin-protein ligase COP1; Ultraviolet-B receptor UVR8]
CS[Protein SUPPRESSOR OF PHYA-105 1; E3 ubiquitin-protein ligase COP1]
UR[WD repeat-containing protein RUP1; WD repeat-containing protein RUP2]
CD[Cullin-4; DDB1- and CUL4-associated factor homolog 1]

Ouzounoglou2014 - Modeling of alpha-synuclein effects on neuronal homeostasis: BIOMD0000000559v0.0.1

Ouzounoglou2014 - Modeling of alpha-synuclein effects on neuronal homeostasisThis model is described in the article: [I…

Details

BACKGROUND: Alpha-synuclein (ASYN) is central in Parkinson's disease (PD) pathogenesis. Converging pieces of evidence suggest that the levels of ASYN expression play a critical role in both familial and sporadic Parkinson's disease. ASYN fibrils are the main component of inclusions called Lewy Bodies (LBs) which are found mainly in the surviving neurons of the substantia nigra. Despite the accumulated knowledge regarding the involvement of ASYN in molecular mechanisms underlying the development of PD, there is much information missing which prevents understanding the causes of the disease and how to stop its progression. RESULTS: Using a Systems Biology approach, we develop a biomolecular reactions model that describes the intracellular ASYN dynamics in relation to overexpression, post-translational modification, oligomerization and degradation of the protein. Especially for the proteolysis of ASYN, the model takes into account the biological knowledge regarding the contribution of Chaperone Mediated Autophagy (CMA), macro-autophagic and proteasome pathways in the protein's degradation. Importantly, inhibitory phenomena, caused by ASYN, concerning CMA (more specifically the lysosomal-associated membrane protein 2a, abbreviated as Lamp2a receptor, which is the rate limiting step of CMA) and the proteasome are carefully modeled. The model is validated by simulation studies of known experimental overexpression data from SH-SY5Y cells and the unknown model parameters are estimated either computationally or by experimental fitting. The calibrated model is then tested under three hypothetical intervention scenarios and in all cases predicts increased cell viability that agrees with experimental evidence. The biomodel has been annotated and is made available in SBML format. CONCLUSIONS: The mathematical model presented here successfully simulates the dynamic phenomena of ASYN overexpression and oligomerization and predicts the biological system's behavior in a number of scenarios not used for model calibration. It allows, for the first time, to qualitatively estimate the protein levels that are capable of deregulating proteolytic homeostasis. In addition, it can help form new hypotheses for intervention that could be tested experimentally. link: http://identifiers.org/pubmed/24885905

Parameters:

NameDescription
k_DisRate = 4.999533748E-7Reaction: s23 => s24 + s17; s23, Rate Law: c1*k_DisRate*s23
k_ProtOligDegr = 3.70096E-4Reaction: s385 => s35; s385, Rate Law: c1*k_ProtOligDegr*s385
k_M_autophagyDegr = 0.1Reaction: s529 => s448; s529, Rate Law: c2*k_M_autophagyDegr*s529
k_ProteasomeBind = 3.424693672E-9Reaction: s25 + s35 => s477; s25, s35, Rate Law: c1*k_ProteasomeBind*s25*s35
k_OligAutophagUptake = 2.39034347E-8Reaction: s24 => s517; s24, Rate Law: k_OligAutophagUptake*s24
k1=0.0294219Reaction: s3 => s17; s3, Rate Law: c1*k1*s3
k1=4.90556E-7Reaction: s33 + s17 => s33; s33, s17, Rate Law: c1*k1*s33*s17
k_2merForm = 1.462941015E-9Reaction: s17 => s18; s17, Rate Law: c1*k_2merForm*s17^2
k_OligomerForm = 3.350497192E-8Reaction: s24 + s17 => s23; s24, s17, Rate Law: c1*k_OligomerForm*s24*s17
k_WTOligBindOnLamp = 4.0E-6Reaction: s23 + s51 => s496; s23, s51, Rate Law: k_WTOligBindOnLamp*s23*s51
k_LampFreeWTasyn = 3.044571674E-4Reaction: s496 => s23 + s51; s496, Rate Law: k_LampFreeWTasyn*s496

States:

NameDescription
s23[Alpha-synuclein]
s7[Alpha-synuclein]
s24[Alpha-synuclein]
s500[Alpha-synuclein; Lysosome-associated membrane glycoprotein 2]
s35[proteasome complex]
s533[Alpha-synuclein]
s31[Alpha-synuclein]
s32[Alpha-synuclein]
s1[Alpha-synuclein; dopamine]
s501[Alpha-synuclein; Lysosome-associated membrane glycoprotein 2]
s535[Alpha-synuclein; dopamine]
s17[Alpha-synuclein]
s25[Alpha-synuclein; dopamine]
s529[Alpha-synuclein; dopamine]
s33[Alpha-synuclein]
s484[dopamine; Alpha-synuclein; Lysosome-associated membrane glycoprotein 2]
s30[Alpha-synuclein]
s26[Alpha-synuclein; dopamine]
s531[dopamine; Alpha-synuclein]
s490[Alpha-synuclein; dopamine; Lysosome-associated membrane glycoprotein 2]
s29[Alpha-synuclein]
s27[dopamine; Alpha-synuclein]

Overgaard2007_PDmodel_IL21: BIOMD0000000238v0.0.1

This a model from the article: PKPD model of interleukin-21 effects on thermoregulation in monkeys--application and…

Details

PURPOSE: To describe the pharmacodynamic effects of recombinant human interleukin-21 (IL-21) on core body temperature in cynomolgus monkeys using basic mechanisms of heat regulation. A major effort was devoted to compare the use of ordinary differential equations (ODEs) with stochastic differential equations (SDEs) in pharmacokinetic pharmacodynamic (PKPD) modelling. METHODS: A temperature model was formulated including circadian rhythm, metabolism, heat loss, and a thermoregulatory set-point. This model was formulated as a mixed-effects model based on SDEs using NONMEM. RESULTS: The effects of IL-21 were on the set-point and the circadian rhythm of metabolism. The model was able to describe a complex set of IL-21 induced phenomena, including 1) disappearance of the circadian rhythm, 2) no effect after first dose, and 3) high variability after second dose. SDEs provided a more realistic description with improved simulation properties, and further changed the model into one that could not be falsified by the autocorrelation function. CONCLUSIONS: The IL-21 induced effects on thermoregulation in cynomolgus monkeys are explained by a biologically plausible model. The quality of the model was improved by the use of SDEs. link: http://identifiers.org/pubmed/17009101

Owen1998 - Tumour treatment model: BIOMD0000000650v0.0.1

Owen1998 - tumour treatment modelThis model is described in the article: [Modelling the macrophage invasion of tumours:…

Details

Even in the early stages of their development, tumours are not simply a homogeneous grouping of mutant cells; rather, they develop in tandem with normal tissue cells, and also recruit other cell types including lymphatic cells and the endothelial cells required for the development of a blood supply. It has been repeatedly seen that macrophages form a significant proportion of the tumour mass, and that they can have a variety of effects upon the tumour, leading to a delicate balance between growth promotion and inhibition. This paper develops a model for the early, avascular growth of a tumour, concentrating on the inhibitory effect of macrophages due to their cytolytic activity. It is shown that such an immune response is not sufficient to prevent growth, due to it being a second-order process with respect to the density of the tumour cells present. However, the presence of macrophages does have important effects on the tumour composition, and the authors perform a detailed bifurcation analysis of their model to clarify this. An extended model is also considered which incorporates addition of exogenous chemical regulators. In this case, the model admits the possibility of tumour regression, and the therapeutic implications of this are discussed. link: http://identifiers.org/pubmed/9661282

Parameters:

NameDescription
K_l = 17.857; N = 1.0; A = 0.025; I = 0.01; delta_l = 0.1; F = 0.0; S = 62.5Reaction: l = ((A*l*(m+F)*(N+1)/(N+l+m+n)+I*(1+S*(m+F)))-K_l*l*m*(m+F))-delta_l*l, Rate Law: ((A*l*(m+F)*(N+1)/(N+l+m+n)+I*(1+S*(m+F)))-K_l*l*m*(m+F))-delta_l*l
N = 1.0; xi = 2.0; K_m = 25.0; F = 0.0Reaction: m = (xi*m*(N+1)/(N+l+m+n)-m)-K_m*l*m*(m+F), Rate Law: (xi*m*(N+1)/(N+l+m+n)-m)-K_m*l*m*(m+F)
N = 1.0Reaction: n = n*(N+1)/(N+l+m+n)-n, Rate Law: n*(N+1)/(N+l+m+n)-n

States:

NameDescription
m[neoplastic cell]
l[macrophage]
n[urn:miriam:go:G0%3A0005623]

Owen1998 - tumour growth model: BIOMD0000000670v0.0.1

Owen1998 - tumour growth modelDeterministic model for the early, avascular growth of a tumour, concentrating on the inhi…

Details

Even in the early stages of their development, tumours are not simply a homogeneous grouping of mutant cells; rather, they develop in tandem with normal tissue cells, and also recruit other cell types including lymphatic cells and the endothelial cells required for the development of a blood supply. It has been repeatedly seen that macrophages form a significant proportion of the tumour mass, and that they can have a variety of effects upon the tumour, leading to a delicate balance between growth promotion and inhibition. This paper develops a model for the early, avascular growth of a tumour, concentrating on the inhibitory effect of macrophages due to their cytolytic activity. It is shown that such an immune response is not sufficient to prevent growth, due to it being a second-order process with respect to the density of the tumour cells present. However, the presence of macrophages does have important effects on the tumour composition, and the authors perform a detailed bifurcation analysis of their model to clarify this. An extended model is also considered which incorporates addition of exogenous chemical regulators. In this case, the model admits the possibility of tumour regression, and the therapeutic implications of this are discussed. link: http://identifiers.org/pubmed/9661282

Parameters:

NameDescription
K_l = 17.857; N = 1.0; A = 0.025; I = 0.01; delta_l = 0.1; S = 62.5Reaction: l = ((A*l*m*(N+1)/(N+l+m+n)+I*(1+S*m))-K_l*l*m*m)-delta_l*l, Rate Law: ((A*l*m*(N+1)/(N+l+m+n)+I*(1+S*m))-K_l*l*m*m)-delta_l*l
N = 1.0Reaction: n = n*(N+1)/(N+l+m+n)-n, Rate Law: n*(N+1)/(N+l+m+n)-n
N = 1.0; xi = 2.0; K_m = 25.0Reaction: m = (xi*m*(N+1)/(N+l+m+n)-m)-K_m*l*m*m, Rate Law: (xi*m*(N+1)/(N+l+m+n)-m)-K_m*l*m*m

States:

NameDescription
m[neoplastic cell]
l[macrophage]
n[cell]

Oxhamre2005_Ca_oscillation: BIOMD0000000047v0.0.1

The model should reproduce the figure 1C of the article (successfully reproduced in MathSBML). If your software does not…

Details

The toxin alpha-hemolysin expressed by uropathogenic Escherichia coli bacteria was recently shown as the first pathophysiologically relevant protein to induce oscillations of the intracellular Ca(2+) concentration in target cells. Here, we propose a generic three-variable kinetic model describing the Ca(2+) oscillations induced in single rat renal epithelial cells by this toxin. Specifically, we take into account the interplay between 1), the cytosolic Ca(2+) concentration; 2), IP(3)-sensitive Ca(2+) channels located in the membrane separating the cytosol and endoplasmic reticulum; and 3), toxin-related activation of production of IP(3) by phospholipase C. With these ingredients, the predicted response of cells exposed to the toxin is in good agreement with the results of experiments. link: http://identifiers.org/pubmed/15596518

Parameters:

NameDescription
Kpump=0.1; Fpump_0=2.0Reaction: Ca_Cyt => CaER, Rate Law: Fpump_0*Ca_Cyt/(Kpump+Ca_Cyt)
Fleak=0.5Reaction: CaER => Ca_Cyt, Rate Law: Fleak
p2 = 0.0; Fch_0=8.0; p1 = 0.0; p3 = 0.95Reaction: CaER => Ca_Cyt, Rate Law: Fch_0*p1*p2*p3

States:

NameDescription
CaER[calcium(2+)]
Ca Cyt[calcium(2+)]

P


Padala2017- ERK, PI3K/Akt and Wnt signalling network (bRaf mutated): BIOMD0000000653v0.0.1

Padala2017- ERK, PI3K/Akt and Wnt signalling network (bRaf mutated)Crosstalk model of the ERK, Wnt and Akt signalling pa…

Details

Perturbations in molecular signaling pathways are a result of genetic or epigenetic alterations, which may lead to malignant transformation of cells. Despite cellular robustness, specific genetic or epigenetic changes of any gene can trigger a cascade of failures, which result in the malfunctioning of cell signaling pathways and lead to cancer phenotypes. The extent of cellular robustness has a link with the architecture of the network such as feedback and feedforward loops. Perturbation in components within feedback loops causes a transition from a regulated to a persistently activated state and results in uncontrolled cell growth. This work represents the mathematical and quantitative modeling of ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk to show the dynamics of signaling responses during genetic and epigenetic changes in cancer. ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk networks include both intra and inter-pathway feedback loops which function in a controlled fashion in a healthy cell. Our results show that cancerous perturbations of components such as EGFR, Ras, B-Raf, PTEN, and components of the destruction complex cause extreme fragility in the network and constitutively activate inter-pathway positive feedback loops. We observed that the aberrant signaling response due to the failure of specific network components is transmitted throughout the network via crosstalk, generating an additive effect on cancer growth and proliferation. link: http://identifiers.org/pubmed/28367561

Parameters:

NameDescription
k27c = 1.5E-4Reaction: RKIP => RKIP + GSK3B, Rate Law: Cell*k27c*RKIP/Cell
k33a2 = 0.8333; k33a1 = 0.01667Reaction: APC + Axin => APCAxin, Rate Law: Cell*(k33a1*Axin*APC-k33a2*APCAxin)/Cell
W = 0.0; k28 = 0.003Reaction: Dshi => Dsha, Rate Law: Cell*k28*Dshi*W/Cell
Kcat24 = 32.344; Km24 = 35954.3Reaction: pC3G + Rap1 => pC3G + pRap1, Rate Law: Cell*Kcat24*pC3G*Rap1/(Rap1+Km24)/Cell
Km16b = 62464.6; Kcat16b = 0.8841Reaction: pRap1 + BRaf => pRap1 + pBRaf, Rate Law: Cell*Kcat16b*pRap1*BRaf/(BRaf+Km16b)/Cell
k6a = 2.5Reaction: pSOS => SOS, Rate Law: Cell*k6a*pSOS/Cell
k3 = 0.00125Reaction: fEGFR => null, Rate Law: Cell*k3*fEGFR/Cell
Km22b = 100.0; Kcat22b = 48.667Reaction: pAkt => Akt, Rate Law: Cell*Kcat22b*pAkt/(Km22b+pAkt)/Cell
Kcat23a = 694.73; Km23a = 6086100.0Reaction: bEGFR + C3G => bEGFR + pC3G, Rate Law: Cell*Kcat23a*bEGFR*C3G/(C3G+Km23a)/Cell
Kcat8b = 1509.36; Km8b = 1432410.0Reaction: RasGap + pRas => RasGap + Ras, Rate Law: Cell*Kcat8b*RasGap*pRas/(pRas+Km8b)/Cell
Km22a = 100.0; Kcat22a = 0.33Reaction: PIP3 + Akt => PIP3 + pAkt, Rate Law: Cell*Kcat22a*PIP3*Akt/(Akt+Km22a)/Cell
Km20 = 4.0; Kcat20 = 4.0Reaction: pPI3K + PIP2 => pPI3K + PIP3, Rate Law: Cell*Kcat20*pPI3K*PIP2/(PIP2+Km20)/Cell
Km7 = 35954.3; Kcat7 = 32.644Reaction: pSOS + Ras => pSOS + pRas, Rate Law: Cell*Kcat7*pSOS*Ras/(Ras+Km7)/Cell
Kcat13 = 9.8537; Km13 = 1007300.0Reaction: pMEK + ERK => pMEK + pERK, Rate Law: Cell*Kcat13*pMEK*ERK/(ERK+Km13)/Cell
k35 = 3.433Reaction: pAPCpAxinGSK3BBCatenin => pAPCpAxinGSK3BpBCatenin, Rate Law: Cell*k35*pAPCpAxinGSK3BBCatenin/Cell
k19c = 0.005Reaction: pPI3K => PI3K, Rate Law: Cell*k19c*pPI3K/Cell
Kcat27a = 0.002Reaction: pERK + GSK3B => pERK + pGSK3B, Rate Law: Cell*Kcat27a*GSK3B*pERK/Cell
k36 = 3.433Reaction: pAPCpAxinGSK3BpBCatenin => pBCatenin + pAPCpAxinGSK3B, Rate Law: Cell*k36*pAPCpAxinGSK3BpBCatenin/Cell
V8a = 0.0717Reaction: null => Ras, Rate Law: Cell*V8a/Cell
Km9a = 62464.6; Kcat9a = 0.884096Reaction: pRas + Raf1 => pRas + pRaf1, Rate Law: Cell*Kcat9a*pRas*Raf1/(Raf1+Km9a)/Cell
Kcat12 = 2.8324; Km12 = 518750.0Reaction: pMEK + PP2A => MEK + PP2A, Rate Law: Cell*Kcat12*PP2A*pMEK/(pMEK+Km12)/Cell
k53 = 2.8833E-4; k52 = 3.85E-5; k54 = 1.5; k51 = 0.003465Reaction: PKCD + pERK + bEGFR + SOS => PKCD + pERK + bEGFR + pSOS, Rate Law: Cell*(k51*bEGFR+k52+k53*PKCD)/(1+pERK/k54)/Cell
k37a2 = 20.0; k37a1 = 0.01667Reaction: BCatenin + APC => APCBCatenin, Rate Law: Cell*(k37a1*APC*BCatenin-k37a2*APCBCatenin)/Cell
Kcat14 = 8.8912; Km14 = 3496500.0Reaction: pERK + PP2A => ERK + PP2A, Rate Law: Cell*Kcat14*PP2A*pERK/(pERK+Km14)/Cell
k312 = 0.01515; k311 = 0.001515Reaction: APCAxin + GSK3B => APCAxinGSK3B, Rate Law: Cell*(k311*GSK3B*APCAxin-k312*APCAxinGSK3B)/Cell
Kcat19b = 0.07711; Km19b = 272056.0Reaction: PI3K => pPI3K; pRas, Rate Law: Cell*Kcat19b*pRas*PI3K/(PI3K+Km19b)/Cell
k23b = 2.5Reaction: pC3G => C3G, Rate Law: Cell*k23b*pC3G/Cell
k15c = 0.00193Reaction: RKIP => null, Rate Law: Cell*k15c*RKIP/Cell
V37b = 0.00705Reaction: null => BCatenin, Rate Law: Cell*V37b/Cell
Km9b = 15.0; W = 0.0; k9b = 0.025Reaction: X + Raf1 => X + pRaf1, Rate Law: Cell*k9b*W*X*Raf1/(Km9b+Raf1)/Cell
Kcat6b = 1611.97; Km6b = 896896.0Reaction: pP90Rsk + pSOS => pP90Rsk + SOS, Rate Law: Cell*Kcat6b*pP90Rsk*pSOS/(pSOS+Km6b)/Cell
Kcat18b = 0.02137; Km18b = 763523.0Reaction: pERK + P90Rsk => pERK + pP90Rsk, Rate Law: Cell*Kcat18b*pERK*P90Rsk/(P90Rsk+Km18b)/Cell
k32b = 0.002217Reaction: pAPCpAxinGSK3B => APCAxinGSK3B, Rate Law: Cell*k32b*pAPCpAxinGSK3B/Cell
k22 = 0.121008; k21 = 2.18503E-5Reaction: fEGFR + EGF => bEGFR, Rate Law: Cell*(k21*EGF*fEGFR-k22*bEGFR)/Cell
k40 = 2.5E-4Reaction: X => null, Rate Law: Cell*k40*X/Cell
V15b = 4.0Reaction: pRKIP => RKIP, Rate Law: Cell*V15b*pRKIP/Cell
k32a = 0.00445Reaction: APCAxinGSK3B => pAPCpAxinGSK3B, Rate Law: Cell*k32a*APCAxinGSK3B/Cell
Kcat21 = 5.5; Km21 = 0.08Reaction: PTEN + PIP3 => PTEN + PIP2, Rate Law: Cell*Kcat21*PTEN*PIP3/(PIP3+Km21)/Cell
Kcat10b = 15.1212; Km10b = 119355.0Reaction: pAkt + pRaf1 => pAkt + Raf1, Rate Law: Cell*Kcat10b*pAkt*pRaf1/(pRaf1+Km10b)/Cell
k11b2 = 120.0; k11b1 = 1.1167E-5Reaction: pRKIP + pRaf1 + MEK => pRKIP + pRaf1 + pMEK; RKIP, Rate Law: Cell*k11b1*pRaf1*MEK/(1+((RKIP-pRKIP)/k11b2)^2)/Cell
Km10a = 1061.7; Kcat10a = 0.12633Reaction: RafPPtase + pRaf1 => RafPPtase + Raf1, Rate Law: Cell*Kcat10a*RafPPtase*pRaf1/(pRaf1+Km10a)/Cell
k342 = 2.0; k341 = 0.01667Reaction: BCatenin + pAPCpAxinGSK3B => pAPCpAxinGSK3BBCatenin, Rate Law: Cell*(k341*pAPCpAxinGSK3B*BCatenin-k342*pAPCpAxinGSK3BBCatenin)/Cell
Kcat11a = 185.76; Km11a = 4768400.0Reaction: pBRaf + MEK => pBRaf + pMEK, Rate Law: Cell*Kcat11a*pBRaf*MEK/(MEK+Km11a)/Cell
Km19a = 184912.0; Kcat19a = 10.6737Reaction: bEGFR + PI3K => bEGFR + pPI3K, Rate Law: Cell*Kcat19a*bEGFR*PI3K/(PI3K+Km19a)/Cell
k4 = 0.2Reaction: bEGFR => null, Rate Law: Cell*k4*bEGFR/Cell
k381 = 0.01667; k382 = 0.5Reaction: TCF + BCatenin => TCFBCatenin, Rate Law: Cell*(k381*BCatenin*TCF-k382*TCFBCatenin)/Cell
Kcat16a = 0.8841; Km16a = 62645.0Reaction: pRas + BRaf => pRas + pBRaf, Rate Law: Cell*Kcat16a*pRas*BRaf/(BRaf+Km16a)/Cell
V1 = 100.0Reaction: pEGFR => fEGFR, Rate Law: Cell*V1/Cell
Km25 = 1432400.0; Kcat25 = 1509.4Reaction: Rap1Gap + pRap1 => Rap1Gap + Rap1, Rate Law: Cell*Kcat25*Rap1Gap*pRap1/(pRap1+Km25)/Cell
Kcat27d = 0.01541Reaction: pGSK3B => GSK3B, Rate Law: Cell*Kcat27d*pGSK3B/Cell
k41 = 0.00695Reaction: pBCatenin => null, Rate Law: Cell*k41*pBCatenin/Cell
Km39 = 15.0; k39 = 0.01Reaction: TCFBCatenin => X + TCFBCatenin, Rate Law: Cell*k39*TCFBCatenin^2/(Km39^2+TCFBCatenin^2)/Cell
Kcat27b = 0.04596Reaction: pAkt + GSK3B => pAkt + pGSK3B, Rate Law: Cell*Kcat27b*GSK3B*pAkt/Cell
k33b = 0.002783Reaction: Axin => null, Rate Law: Cell*k33b*Axin/Cell
k15a = 1.3Reaction: pERK + RKIP => pERK + pRKIP, Rate Law: Cell*k15a*pERK*(RKIP-pRKIP)/Cell
k33c1 = 1.37E-6; k33c2 = 1.667E-8Reaction: BCatenin + TCFBCatenin => BCatenin + TCFBCatenin + Axin, Rate Law: Cell*(k33c1+k33c2*(TCFBCatenin+BCatenin))/Cell

States:

NameDescription
pC3G[urn:miriam:bao:0002007; Complement C3]
pBCatenin[urn:miriam:bao:0002007; Catenin beta-1]
PIP2[urn:miriam:chebi:0018348]
APCAxin[Adenomatous polyposis coli protein; Axin-1]
pPI3K[urn:miriam:bao:0002007; urn:miriam:omit:0027264]
Akt[RAC-alpha serine/threonine-protein kinase]
bEGFR[Pro-epidermal growth factor; Epidermal growth factor receptor]
pAPCpAxinGSK3BBCatenin[urn:miriam:bao:0002007; Adenomatous polyposis coli protein; Catenin beta-1; Glycogen synthase kinase-3 beta; Axin-1]
pSOS[urn:miriam:bao:0002007; Son of sevenless homolog 1]
RafPPtase[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
pP90Rsk[urn:miriam:bao:0002007; Ribosomal protein S6 kinase alpha-1]
pMEK[urn:miriam:bao:0002007; Dual specificity mitogen-activated protein kinase kinase 1]
pAkt[urn:miriam:bao:0002007; RAC-alpha serine/threonine-protein kinase]
pRKIP[urn:miriam:bao:0002007; Phosphatidylethanolamine-binding protein 1]
BCatenin[Catenin beta-1]
RKIP[Phosphatidylethanolamine-binding protein 1]
PIP3[urn:miriam:chebi:0016618]
Ras[GTPase HRas]
pEGFR[urn:miriam:bao:0002007; Epidermal growth factor receptor]
SOS[Son of sevenless homolog 1]
pRaf1[urn:miriam:bao:0002007; RAF proto-oncogene serine/threonine-protein kinase]
TCFBCatenin[Catenin beta-1; Lymphoid enhancer-binding factor 1]
Rap1[Ras-related protein Rap-1A]
XX
C3G[Rap guanine nucleotide exchange factor 1]
pAPCpAxinGSK3B[urn:miriam:bao:0002007; Glycogen synthase kinase-3 beta; Axin-1; Adenomatous polyposis coli protein]
Dsha[urn:miriam:bao:0002007; Segment polarity protein dishevelled homolog DVL-1]
TCF[Lymphoid enhancer-binding factor 1]
Raf1[RAF proto-oncogene serine/threonine-protein kinase]
pGSK3B[urn:miriam:bao:0002007; Glycogen synthase kinase-3 beta]
Rap1Gap[Rap1 GTPase-activating protein 1]
APCBCatenin[Adenomatous polyposis coli protein; Catenin beta-1]
fEGFR[Epidermal growth factor receptor]
pERK[urn:miriam:bao:0002007; Mitogen-activated protein kinase 1]
APC[Adenomatous polyposis coli protein]
pBRaf[urn:miriam:bao:0002007; urn:miriam:omit:0010192; Serine/threonine-protein kinase B-raf]
pRas[urn:miriam:bao:0002007; Ras-related protein R-Ras2]
RasGap[Ras GTPase-activating protein 1]
APCAxinGSK3B[Glycogen synthase kinase-3 beta; Adenomatous polyposis coli protein; Axin-1]
Axin[Axin-1]
pAPCpAxinGSK3BpBCatenin[urn:miriam:bao:0002007; Adenomatous polyposis coli protein; Axin-1; Glycogen synthase kinase-3 beta; Catenin beta-1]

Padala2017- ERK, PI3K/Akt and Wnt signalling network (EGFR overexpression): BIOMD0000000656v0.0.1

Padala2017- ERK, PI3K/Akt and Wnt signalling network (EGFR overexpression)Crosstalk model of the ERK, Wnt and Akt signal…

Details

Perturbations in molecular signaling pathways are a result of genetic or epigenetic alterations, which may lead to malignant transformation of cells. Despite cellular robustness, specific genetic or epigenetic changes of any gene can trigger a cascade of failures, which result in the malfunctioning of cell signaling pathways and lead to cancer phenotypes. The extent of cellular robustness has a link with the architecture of the network such as feedback and feedforward loops. Perturbation in components within feedback loops causes a transition from a regulated to a persistently activated state and results in uncontrolled cell growth. This work represents the mathematical and quantitative modeling of ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk to show the dynamics of signaling responses during genetic and epigenetic changes in cancer. ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk networks include both intra and inter-pathway feedback loops which function in a controlled fashion in a healthy cell. Our results show that cancerous perturbations of components such as EGFR, Ras, B-Raf, PTEN, and components of the destruction complex cause extreme fragility in the network and constitutively activate inter-pathway positive feedback loops. We observed that the aberrant signaling response due to the failure of specific network components is transmitted throughout the network via crosstalk, generating an additive effect on cancer growth and proliferation. link: http://identifiers.org/pubmed/28367561

Parameters:

NameDescription
k27c = 1.5E-4Reaction: RKIP => RKIP + GSK3B, Rate Law: Cell*k27c*RKIP/Cell
k33a2 = 0.8333; k33a1 = 0.01667Reaction: APC + Axin => APCAxin, Rate Law: Cell*(k33a1*Axin*APC-k33a2*APCAxin)/Cell
W = 0.0; k28 = 0.003Reaction: Dshi => Dsha, Rate Law: Cell*k28*Dshi*W/Cell
Kcat24 = 32.344; Km24 = 35954.3Reaction: pC3G + Rap1 => pC3G + pRap1, Rate Law: Cell*Kcat24*pC3G*Rap1/(Rap1+Km24)/Cell
Km16b = 62464.6; Kcat16b = 0.8841Reaction: pRap1 + BRaf => pRap1 + pBRaf, Rate Law: Cell*Kcat16b*pRap1*BRaf/(BRaf+Km16b)/Cell
k6a = 2.5Reaction: pSOS => SOS, Rate Law: Cell*k6a*pSOS/Cell
Kcat23a = 694.73; Km23a = 6086100.0Reaction: bEGFR + C3G => bEGFR + pC3G, Rate Law: Cell*Kcat23a*bEGFR*C3G/(C3G+Km23a)/Cell
Km22b = 100.0; Kcat22b = 48.667Reaction: pAkt => Akt, Rate Law: Cell*Kcat22b*pAkt/(Km22b+pAkt)/Cell
Kcat8b = 1509.36; Km8b = 1432410.0Reaction: RasGap + pRas => RasGap + Ras, Rate Law: Cell*Kcat8b*RasGap*pRas/(pRas+Km8b)/Cell
k18a = 0.005Reaction: pP90Rsk => P90Rsk, Rate Law: Cell*k18a*pP90Rsk/Cell
Km20 = 4.0; Kcat20 = 4.0Reaction: pPI3K + PIP2 => pPI3K + PIP3, Rate Law: Cell*Kcat20*pPI3K*PIP2/(PIP2+Km20)/Cell
Km22a = 100.0; Kcat22a = 0.33Reaction: PIP3 + Akt => PIP3 + pAkt, Rate Law: Cell*Kcat22a*PIP3*Akt/(Akt+Km22a)/Cell
Km7 = 35954.3; Kcat7 = 32.644Reaction: pSOS + Ras => pSOS + pRas, Rate Law: Cell*Kcat7*pSOS*Ras/(Ras+Km7)/Cell
Kcat13 = 9.8537; Km13 = 1007300.0Reaction: pMEK + ERK => pMEK + pERK, Rate Law: Cell*Kcat13*pMEK*ERK/(ERK+Km13)/Cell
k19c = 0.005Reaction: pPI3K => PI3K, Rate Law: Cell*k19c*pPI3K/Cell
k26b = 3.85E-4Reaction: PKCD => null, Rate Law: Cell*k26b*PKCD/Cell
Kcat27a = 0.002Reaction: pERK + GSK3B => pERK + pGSK3B, Rate Law: Cell*Kcat27a*GSK3B*pERK/Cell
V8a = 0.0717Reaction: null => Ras, Rate Law: Cell*V8a/Cell
Km9a = 62464.6; Kcat9a = 0.884096Reaction: pRas + Raf1 => pRas + pRaf1, Rate Law: Cell*Kcat9a*pRas*Raf1/(Raf1+Km9a)/Cell
Kcat12 = 2.8324; Km12 = 518750.0Reaction: pMEK + PP2A => MEK + PP2A, Rate Law: Cell*Kcat12*PP2A*pMEK/(pMEK+Km12)/Cell
k53 = 2.8833E-4; k52 = 3.85E-5; k54 = 1.5; k51 = 0.003465Reaction: PKCD + pERK + bEGFR + SOS => PKCD + pERK + bEGFR + pSOS, Rate Law: Cell*(k51*bEGFR+k52+k53*PKCD)/(1+pERK/k54)/Cell
k37a2 = 20.0; k37a1 = 0.01667Reaction: BCatenin + APC => APCBCatenin, Rate Law: Cell*(k37a1*APC*BCatenin-k37a2*APCBCatenin)/Cell
Kcat14 = 8.8912; Km14 = 3496500.0Reaction: pERK + PP2A => ERK + PP2A, Rate Law: Cell*Kcat14*PP2A*pERK/(pERK+Km14)/Cell
k312 = 0.01515; k311 = 0.001515Reaction: APCAxin + GSK3B => APCAxinGSK3B, Rate Law: Cell*(k311*GSK3B*APCAxin-k312*APCAxinGSK3B)/Cell
Kcat19b = 0.07711; Km19b = 272056.0Reaction: PI3K => pPI3K; pRas, Rate Law: Cell*Kcat19b*pRas*PI3K/(PI3K+Km19b)/Cell
k23b = 2.5Reaction: pC3G => C3G, Rate Law: Cell*k23b*pC3G/Cell
k15c = 0.00193Reaction: RKIP => null, Rate Law: Cell*k15c*RKIP/Cell
V37b = 0.00705Reaction: null => BCatenin, Rate Law: Cell*V37b/Cell
Km9b = 15.0; W = 0.0; k9b = 0.025Reaction: X + Raf1 => X + pRaf1, Rate Law: Cell*k9b*W*X*Raf1/(Km9b+Raf1)/Cell
Kcat6b = 1611.97; Km6b = 896896.0Reaction: pP90Rsk + pSOS => pP90Rsk + SOS, Rate Law: Cell*Kcat6b*pP90Rsk*pSOS/(pSOS+Km6b)/Cell
Kcat18b = 0.02137; Km18b = 763523.0Reaction: pERK + P90Rsk => pERK + pP90Rsk, Rate Law: Cell*Kcat18b*pERK*P90Rsk/(P90Rsk+Km18b)/Cell
k32b = 0.002217Reaction: pAPCpAxinGSK3B => APCAxinGSK3B, Rate Law: Cell*k32b*pAPCpAxinGSK3B/Cell
k22 = 0.121008; k21 = 2.18503E-5Reaction: fEGFR + EGF => bEGFR, Rate Law: Cell*(k21*EGF*fEGFR-k22*bEGFR)/Cell
V15b = 4.0Reaction: pRKIP => RKIP, Rate Law: Cell*V15b*pRKIP/Cell
k32a = 0.00445Reaction: APCAxinGSK3B => pAPCpAxinGSK3B, Rate Law: Cell*k32a*APCAxinGSK3B/Cell
Kcat21 = 5.5; Km21 = 0.08Reaction: PTEN + PIP3 => PTEN + PIP2, Rate Law: Cell*Kcat21*PTEN*PIP3/(PIP3+Km21)/Cell
Kcat10b = 15.1212; Km10b = 119355.0Reaction: pAkt + pRaf1 => pAkt + Raf1, Rate Law: Cell*Kcat10b*pAkt*pRaf1/(pRaf1+Km10b)/Cell
k11b2 = 120.0; k11b1 = 1.1167E-5Reaction: pRKIP + pRaf1 + MEK => pRKIP + pRaf1 + pMEK; RKIP, Rate Law: Cell*k11b1*pRaf1*MEK/(1+((RKIP-pRKIP)/k11b2)^2)/Cell
Km10a = 1061.7; Kcat10a = 0.12633Reaction: RafPPtase + pRaf1 => RafPPtase + Raf1, Rate Law: Cell*Kcat10a*RafPPtase*pRaf1/(pRaf1+Km10a)/Cell
Kcat11a = 185.76; Km11a = 4768400.0Reaction: pBRaf + MEK => pBRaf + pMEK, Rate Law: Cell*Kcat11a*pBRaf*MEK/(MEK+Km11a)/Cell
Km19a = 184912.0; Kcat19a = 10.6737Reaction: bEGFR + PI3K => bEGFR + pPI3K, Rate Law: Cell*Kcat19a*bEGFR*PI3K/(PI3K+Km19a)/Cell
k381 = 0.01667; k382 = 0.5Reaction: TCF + BCatenin => TCFBCatenin, Rate Law: Cell*(k381*BCatenin*TCF-k382*TCFBCatenin)/Cell
Kcat16a = 0.8841; Km16a = 62645.0Reaction: pRas + BRaf => pRas + pBRaf, Rate Law: Cell*Kcat16a*pRas*BRaf/(BRaf+Km16a)/Cell
Kcat17a = 0.12633; Km17a = 1061.71Reaction: RafPPtase + pBRaf => RafPPtase + BRaf, Rate Law: Cell*Kcat17a*RafPPtase*pBRaf/(Km17a+RafPPtase)/Cell
Km25 = 1432400.0; Kcat25 = 1509.4Reaction: Rap1Gap + pRap1 => Rap1Gap + Rap1, Rate Law: Cell*Kcat25*Rap1Gap*pRap1/(pRap1+Km25)/Cell
V1 = 100.0Reaction: pEGFR => fEGFR, Rate Law: Cell*V1/Cell
V26a = 0.00154; k26a = 20.0Reaction: GSK3B => GSK3B + PKCD, Rate Law: Cell*V26a/(1+(GSK3B/k26a)^2.5)/Cell
Kcat27d = 0.01541Reaction: pGSK3B => GSK3B, Rate Law: Cell*Kcat27d*pGSK3B/Cell
Kcat27b = 0.04596Reaction: pAkt + GSK3B => pAkt + pGSK3B, Rate Law: Cell*Kcat27b*GSK3B*pAkt/Cell
k33b = 0.002783Reaction: Axin => null, Rate Law: Cell*k33b*Axin/Cell
k30 = 8.33E-4Reaction: Dsha + APCAxinGSK3B => GSK3B + APCAxin + Dsha, Rate Law: Cell*k30*Dsha*APCAxinGSK3B/Cell
k15a = 1.3Reaction: pERK + RKIP => pERK + pRKIP, Rate Law: Cell*k15a*pERK*(RKIP-pRKIP)/Cell

States:

NameDescription
pC3G[Complement C3; urn:miriam:bao:0002007]
bEGFR[Epidermal growth factor receptor; Pro-epidermal growth factor]
PTEN[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN]
APCAxin[Axin-1; Adenomatous polyposis coli protein]
pSOS[Son of sevenless homolog 1; urn:miriam:bao:0002007]
EGF[Pro-epidermal growth factor]
BRaf[Serine/threonine-protein kinase B-raf]
BCatenin[Catenin beta-1]
PIP3[urn:miriam:chebi:0016618]
Ras[GTPase HRas]
pEGFR[Epidermal growth factor receptor; urn:miriam:bao:0002007]
SOS[Son of sevenless homolog 1]
pRaf1[RAF proto-oncogene serine/threonine-protein kinase; urn:miriam:bao:0002007]
MEK[Dual specificity mitogen-activated protein kinase kinase 1]
Rap1[Ras-related protein Rap-1A]
TCF[Lymphoid enhancer-binding factor 1]
Raf1[RAF proto-oncogene serine/threonine-protein kinase]
PP2A[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
pRas[Ras-related protein R-Ras2; urn:miriam:bao:0002007]
Dshi[Segment polarity protein dishevelled homolog DVL-1]
fEGFR[Epidermal growth factor receptor]
pERK[Mitogen-activated protein kinase 1; urn:miriam:bao:0002007]
GSK3B[Glycogen synthase kinase-3 beta]
APCAxinGSK3B[Axin-1; Adenomatous polyposis coli protein; Glycogen synthase kinase-3 beta]
pPI3K[urn:miriam:omit:0027264; urn:miriam:bao:0002007]
Akt[RAC-alpha serine/threonine-protein kinase]
RafPPtase[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
pP90Rsk[Ribosomal protein S6 kinase alpha-1; urn:miriam:bao:0002007]
pMEK[Dual specificity mitogen-activated protein kinase kinase 1; urn:miriam:bao:0002007]
pRKIP[Phosphatidylethanolamine-binding protein 1; urn:miriam:bao:0002007]
RKIP[Phosphatidylethanolamine-binding protein 1]
PKCD[Protein kinase C delta type]
C3G[Rap guanine nucleotide exchange factor 1]
Dsha[Segment polarity protein dishevelled homolog DVL-1; urn:miriam:bao:0002007]
pAPCpAxinGSK3B[Adenomatous polyposis coli protein; Axin-1; Glycogen synthase kinase-3 beta; urn:miriam:bao:0002007]
pGSK3B[Glycogen synthase kinase-3 beta; urn:miriam:bao:0002007]
Rap1Gap[Rap1 GTPase-activating protein 1]
APC[Adenomatous polyposis coli protein]
pBRaf[Serine/threonine-protein kinase B-raf; urn:miriam:bao:0002007]
ERK[Mitogen-activated protein kinase 3]
RasGap[Ras GTPase-activating protein 1]
Axin[Axin-1]
pRap1[Ras-related protein Rap-1A; urn:miriam:bao:0002007]

Padala2017- ERK, PI3K/Akt and Wnt signalling network (normal): BIOMD0000000648v0.0.1

Padala2017- ERK, PI3K/Akt and Wnt signalling network (normal)Crosstalk model of the ERK, Wnt and Akt signalling pathways…

Details

Perturbations in molecular signaling pathways are a result of genetic or epigenetic alterations, which may lead to malignant transformation of cells. Despite cellular robustness, specific genetic or epigenetic changes of any gene can trigger a cascade of failures, which result in the malfunctioning of cell signaling pathways and lead to cancer phenotypes. The extent of cellular robustness has a link with the architecture of the network such as feedback and feedforward loops. Perturbation in components within feedback loops causes a transition from a regulated to a persistently activated state and results in uncontrolled cell growth. This work represents the mathematical and quantitative modeling of ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk to show the dynamics of signaling responses during genetic and epigenetic changes in cancer. ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk networks include both intra and inter-pathway feedback loops which function in a controlled fashion in a healthy cell. Our results show that cancerous perturbations of components such as EGFR, Ras, B-Raf, PTEN, and components of the destruction complex cause extreme fragility in the network and constitutively activate inter-pathway positive feedback loops. We observed that the aberrant signaling response due to the failure of specific network components is transmitted throughout the network via crosstalk, generating an additive effect on cancer growth and proliferation. link: http://identifiers.org/pubmed/28367561

Parameters:

NameDescription
k27c = 1.5E-4Reaction: RKIP => RKIP + GSK3B, Rate Law: Cell*k27c*RKIP/Cell
k33a2 = 0.8333; k33a1 = 0.01667Reaction: APC + Axin => APCAxin, Rate Law: Cell*(k33a1*Axin*APC-k33a2*APCAxin)/Cell
W = 0.0; k28 = 0.003Reaction: Dshi => Dsha, Rate Law: Cell*k28*Dshi*W/Cell
Kcat24 = 32.344; Km24 = 35954.3Reaction: pC3G + Rap1 => pC3G + pRap1, Rate Law: Cell*Kcat24*pC3G*Rap1/(Rap1+Km24)/Cell
Kcat17b = 15.1212; Km17b = 119355.0Reaction: pAkt + pBRaf => pAkt + BRaf, Rate Law: Cell*Kcat17b*pBRaf*pAkt/(Km17b+pBRaf)/Cell
Km16b = 62464.6; Kcat16b = 0.8841Reaction: pRap1 + BRaf => pRap1 + pBRaf, Rate Law: Cell*Kcat16b*pRap1*BRaf/(BRaf+Km16b)/Cell
Kcat23a = 694.73; Km23a = 6086100.0Reaction: bEGFR + C3G => bEGFR + pC3G, Rate Law: Cell*Kcat23a*bEGFR*C3G/(C3G+Km23a)/Cell
Km22b = 100.0; Kcat22b = 48.667Reaction: pAkt => Akt, Rate Law: Cell*Kcat22b*pAkt/(Km22b+pAkt)/Cell
k18a = 0.005Reaction: pP90Rsk => P90Rsk, Rate Law: Cell*k18a*pP90Rsk/Cell
Km20 = 4.0; Kcat20 = 4.0Reaction: pPI3K + PIP2 => pPI3K + PIP3, Rate Law: Cell*Kcat20*pPI3K*PIP2/(PIP2+Km20)/Cell
Km22a = 100.0; Kcat22a = 0.33Reaction: PIP3 + Akt => PIP3 + pAkt, Rate Law: Cell*Kcat22a*PIP3*Akt/(Akt+Km22a)/Cell
Km7 = 35954.3; Kcat7 = 32.644Reaction: pSOS + Ras => pSOS + pRas, Rate Law: Cell*Kcat7*pSOS*Ras/(Ras+Km7)/Cell
Kcat13 = 9.8537; Km13 = 1007300.0Reaction: pMEK + ERK => pMEK + pERK, Rate Law: Cell*Kcat13*pMEK*ERK/(ERK+Km13)/Cell
k19c = 0.005Reaction: pPI3K => PI3K, Rate Law: Cell*k19c*pPI3K/Cell
k26b = 3.85E-4Reaction: PKCD => null, Rate Law: Cell*k26b*PKCD/Cell
Kcat27a = 0.002Reaction: pERK + GSK3B => pERK + pGSK3B, Rate Law: Cell*Kcat27a*GSK3B*pERK/Cell
Km9a = 62464.6; Kcat9a = 0.884096Reaction: pRas + Raf1 => pRas + pRaf1, Rate Law: Cell*Kcat9a*pRas*Raf1/(Raf1+Km9a)/Cell
Kcat12 = 2.8324; Km12 = 518750.0Reaction: pMEK + PP2A => MEK + PP2A, Rate Law: Cell*Kcat12*PP2A*pMEK/(pMEK+Km12)/Cell
k53 = 2.8833E-4; k52 = 3.85E-5; k54 = 1.5; k51 = 0.003465Reaction: PKCD + pERK + bEGFR + SOS => PKCD + pERK + bEGFR + pSOS, Rate Law: Cell*(k51*bEGFR+k52+k53*PKCD)/(1+pERK/k54)/Cell
k37a2 = 20.0; k37a1 = 0.01667Reaction: BCatenin + APC => APCBCatenin, Rate Law: Cell*(k37a1*APC*BCatenin-k37a2*APCBCatenin)/Cell
Kcat14 = 8.8912; Km14 = 3496500.0Reaction: pERK + PP2A => ERK + PP2A, Rate Law: Cell*Kcat14*PP2A*pERK/(pERK+Km14)/Cell
k312 = 0.01515; k311 = 0.001515Reaction: APCAxin + GSK3B => APCAxinGSK3B, Rate Law: Cell*(k311*GSK3B*APCAxin-k312*APCAxinGSK3B)/Cell
Kcat19b = 0.07711; Km19b = 272056.0Reaction: PI3K => pPI3K; pRas, Rate Law: Cell*Kcat19b*pRas*PI3K/(PI3K+Km19b)/Cell
k23b = 2.5Reaction: pC3G => C3G, Rate Law: Cell*k23b*pC3G/Cell
k15c = 0.00193Reaction: RKIP => null, Rate Law: Cell*k15c*RKIP/Cell
V37b = 0.00705Reaction: null => BCatenin, Rate Law: Cell*V37b/Cell
k37c = 4.283E-6Reaction: BCatenin => null, Rate Law: Cell*k37c*BCatenin/Cell
Km9b = 15.0; W = 0.0; k9b = 0.025Reaction: X + Raf1 => X + pRaf1, Rate Law: Cell*k9b*W*X*Raf1/(Km9b+Raf1)/Cell
Kcat6b = 1611.97; Km6b = 896896.0Reaction: pP90Rsk + pSOS => pP90Rsk + SOS, Rate Law: Cell*Kcat6b*pP90Rsk*pSOS/(pSOS+Km6b)/Cell
Kcat18b = 0.02137; Km18b = 763523.0Reaction: pERK + P90Rsk => pERK + pP90Rsk, Rate Law: Cell*Kcat18b*pERK*P90Rsk/(P90Rsk+Km18b)/Cell
k32b = 0.002217Reaction: pAPCpAxinGSK3B => APCAxinGSK3B, Rate Law: Cell*k32b*pAPCpAxinGSK3B/Cell
k22 = 0.121008; k21 = 2.18503E-5Reaction: fEGFR + EGF => bEGFR, Rate Law: Cell*(k21*EGF*fEGFR-k22*bEGFR)/Cell
V15b = 4.0Reaction: pRKIP => RKIP, Rate Law: Cell*V15b*pRKIP/Cell
k32a = 0.00445Reaction: APCAxinGSK3B => pAPCpAxinGSK3B, Rate Law: Cell*k32a*APCAxinGSK3B/Cell
Kcat21 = 5.5; Km21 = 0.08Reaction: PTEN + PIP3 => PTEN + PIP2, Rate Law: Cell*Kcat21*PTEN*PIP3/(PIP3+Km21)/Cell
Kcat10b = 15.1212; Km10b = 119355.0Reaction: pAkt + pRaf1 => pAkt + Raf1, Rate Law: Cell*Kcat10b*pAkt*pRaf1/(pRaf1+Km10b)/Cell
k11b2 = 120.0; k11b1 = 1.1167E-5Reaction: pRKIP + pRaf1 + MEK => pRKIP + pRaf1 + pMEK; RKIP, Rate Law: Cell*k11b1*pRaf1*MEK/(1+((RKIP-pRKIP)/k11b2)^2)/Cell
k342 = 2.0; k341 = 0.01667Reaction: BCatenin + pAPCpAxinGSK3B => pAPCpAxinGSK3BBCatenin, Rate Law: Cell*(k341*pAPCpAxinGSK3B*BCatenin-k342*pAPCpAxinGSK3BBCatenin)/Cell
Km10a = 1061.7; Kcat10a = 0.12633Reaction: RafPPtase + pRaf1 => RafPPtase + Raf1, Rate Law: Cell*Kcat10a*RafPPtase*pRaf1/(pRaf1+Km10a)/Cell
Kcat11a = 185.76; Km11a = 4768400.0Reaction: pBRaf + MEK => pBRaf + pMEK, Rate Law: Cell*Kcat11a*pBRaf*MEK/(MEK+Km11a)/Cell
Km19a = 184912.0; Kcat19a = 10.6737Reaction: bEGFR + PI3K => bEGFR + pPI3K, Rate Law: Cell*Kcat19a*bEGFR*PI3K/(PI3K+Km19a)/Cell
k29 = 0.003Reaction: Dsha => Dshi, Rate Law: Cell*k29*Dsha/Cell
k381 = 0.01667; k382 = 0.5Reaction: TCF + BCatenin => TCFBCatenin, Rate Law: Cell*(k381*BCatenin*TCF-k382*TCFBCatenin)/Cell
Kcat16a = 0.8841; Km16a = 62645.0Reaction: pRas + BRaf => pRas + pBRaf, Rate Law: Cell*Kcat16a*pRas*BRaf/(BRaf+Km16a)/Cell
Kcat17a = 0.12633; Km17a = 1061.71Reaction: RafPPtase + pBRaf => RafPPtase + BRaf, Rate Law: Cell*Kcat17a*RafPPtase*pBRaf/(Km17a+RafPPtase)/Cell
Km25 = 1432400.0; Kcat25 = 1509.4Reaction: Rap1Gap + pRap1 => Rap1Gap + Rap1, Rate Law: Cell*Kcat25*Rap1Gap*pRap1/(pRap1+Km25)/Cell
V1 = 100.0Reaction: pEGFR => fEGFR, Rate Law: Cell*V1/Cell
V26a = 0.00154; k26a = 20.0Reaction: GSK3B => GSK3B + PKCD, Rate Law: Cell*V26a/(1+(GSK3B/k26a)^2.5)/Cell
Kcat27d = 0.01541Reaction: pGSK3B => GSK3B, Rate Law: Cell*Kcat27d*pGSK3B/Cell
Kcat27b = 0.04596Reaction: pAkt + GSK3B => pAkt + pGSK3B, Rate Law: Cell*Kcat27b*GSK3B*pAkt/Cell
k33b = 0.002783Reaction: Axin => null, Rate Law: Cell*k33b*Axin/Cell
k30 = 8.33E-4Reaction: Dsha + APCAxinGSK3B => GSK3B + APCAxin + Dsha, Rate Law: Cell*k30*Dsha*APCAxinGSK3B/Cell
k15a = 1.3Reaction: pERK + RKIP => pERK + pRKIP, Rate Law: Cell*k15a*pERK*(RKIP-pRKIP)/Cell
k33c1 = 1.37E-6; k33c2 = 1.667E-8Reaction: BCatenin + TCFBCatenin => BCatenin + TCFBCatenin + Axin, Rate Law: Cell*(k33c1+k33c2*(TCFBCatenin+BCatenin))/Cell

States:

NameDescription
pC3G[urn:miriam:bao:0002007; Complement C3]
PIP2[urn:miriam:chebi:0018348]
APCAxin[Adenomatous polyposis coli protein; Axin-1]
pPI3K[urn:miriam:bao:0002007; urn:miriam:omit:0027264]
Akt[RAC-alpha serine/threonine-protein kinase]
bEGFR[Pro-epidermal growth factor; Epidermal growth factor receptor]
pSOS[urn:miriam:bao:0002007; Son of sevenless homolog 1]
EGF[Pro-epidermal growth factor]
RafPPtase[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
pP90Rsk[urn:miriam:bao:0002007; Ribosomal protein S6 kinase alpha-1]
BRaf[Serine/threonine-protein kinase B-raf]
BCatenin[Catenin beta-1]
pMEK[urn:miriam:bao:0002007; Dual specificity mitogen-activated protein kinase kinase 1]
pAkt[urn:miriam:bao:0002007; RAC-alpha serine/threonine-protein kinase]
PKCD[Protein kinase C delta type]
pRKIP[urn:miriam:bao:0002007; Phosphatidylethanolamine-binding protein 1]
PIP3[urn:miriam:chebi:0016618]
pEGFR[urn:miriam:bao:0002007; Epidermal growth factor receptor]
RKIP[Phosphatidylethanolamine-binding protein 1]
SOS[Son of sevenless homolog 1]
Ras[GTPase HRas]
pRaf1[urn:miriam:bao:0002007; RAF proto-oncogene serine/threonine-protein kinase]
MEK[Dual specificity mitogen-activated protein kinase kinase 1]
PI3K[urn:miriam:omit:0027264]
C3G[Rap guanine nucleotide exchange factor 1]
Rap1[Ras-related protein Rap-1A]
Dsha[urn:miriam:bao:0002007; Segment polarity protein dishevelled homolog DVL-1]
P90Rsk[Ribosomal protein S6 kinase alpha-1]
pGSK3B[urn:miriam:bao:0002007; Glycogen synthase kinase-3 beta]
Raf1[RAF proto-oncogene serine/threonine-protein kinase]
PP2A[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
pRas[urn:miriam:bao:0002007; Ras-related protein R-Ras2]
Dshi[Segment polarity protein dishevelled homolog DVL-1]
pERK[urn:miriam:bao:0002007; Mitogen-activated protein kinase 1]
APCBCatenin[Adenomatous polyposis coli protein; Catenin beta-1]
APC[Adenomatous polyposis coli protein]
GSK3B[Glycogen synthase kinase-3 beta]
ERK[Mitogen-activated protein kinase 3]
pRap1[urn:miriam:bao:0002007; Ras-related protein Rap-1A]
APCAxinGSK3B[Glycogen synthase kinase-3 beta; Adenomatous polyposis coli protein; Axin-1]
Axin[Axin-1]

Padala2017- ERK, PI3K/Akt and Wnt signalling network (PI3K mutated): BIOMD0000000652v0.0.1

Padala2017- ERK, PI3K/Akt and Wnt signalling network (PI3K mutated)Crosstalk model of the ERK, Wnt and Akt signalling pa…

Details

Perturbations in molecular signaling pathways are a result of genetic or epigenetic alterations, which may lead to malignant transformation of cells. Despite cellular robustness, specific genetic or epigenetic changes of any gene can trigger a cascade of failures, which result in the malfunctioning of cell signaling pathways and lead to cancer phenotypes. The extent of cellular robustness has a link with the architecture of the network such as feedback and feedforward loops. Perturbation in components within feedback loops causes a transition from a regulated to a persistently activated state and results in uncontrolled cell growth. This work represents the mathematical and quantitative modeling of ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk to show the dynamics of signaling responses during genetic and epigenetic changes in cancer. ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk networks include both intra and inter-pathway feedback loops which function in a controlled fashion in a healthy cell. Our results show that cancerous perturbations of components such as EGFR, Ras, B-Raf, PTEN, and components of the destruction complex cause extreme fragility in the network and constitutively activate inter-pathway positive feedback loops. We observed that the aberrant signaling response due to the failure of specific network components is transmitted throughout the network via crosstalk, generating an additive effect on cancer growth and proliferation. link: http://identifiers.org/pubmed/28367561

Parameters:

NameDescription
k27c = 1.5E-4Reaction: RKIP => RKIP + GSK3B, Rate Law: Cell*k27c*RKIP/Cell
k33a2 = 0.8333; k33a1 = 0.01667Reaction: APC + Axin => APCAxin, Rate Law: Cell*(k33a1*Axin*APC-k33a2*APCAxin)/Cell
W = 0.0; k28 = 0.003Reaction: Dshi => Dsha, Rate Law: Cell*k28*Dshi*W/Cell
Kcat24 = 32.344; Km24 = 35954.3Reaction: pC3G + Rap1 => pC3G + pRap1, Rate Law: Cell*Kcat24*pC3G*Rap1/(Rap1+Km24)/Cell
Kcat17b = 15.1212; Km17b = 119355.0Reaction: pAkt + pBRaf => pAkt + BRaf, Rate Law: Cell*Kcat17b*pBRaf*pAkt/(Km17b+pBRaf)/Cell
Km16b = 62464.6; Kcat16b = 0.8841Reaction: pRap1 + BRaf => pRap1 + pBRaf, Rate Law: Cell*Kcat16b*pRap1*BRaf/(BRaf+Km16b)/Cell
k6a = 2.5Reaction: pSOS => SOS, Rate Law: Cell*k6a*pSOS/Cell
Kcat23a = 694.73; Km23a = 6086100.0Reaction: bEGFR + C3G => bEGFR + pC3G, Rate Law: Cell*Kcat23a*bEGFR*C3G/(C3G+Km23a)/Cell
Km22b = 100.0; Kcat22b = 48.667Reaction: pAkt => Akt, Rate Law: Cell*Kcat22b*pAkt/(Km22b+pAkt)/Cell
Kcat8b = 1509.36; Km8b = 1432410.0Reaction: RasGap + pRas => RasGap + Ras, Rate Law: Cell*Kcat8b*RasGap*pRas/(pRas+Km8b)/Cell
k18a = 0.005Reaction: pP90Rsk => P90Rsk, Rate Law: Cell*k18a*pP90Rsk/Cell
Km20 = 4.0; Kcat20 = 4.0Reaction: pPI3K + PIP2 => pPI3K + PIP3, Rate Law: Cell*Kcat20*pPI3K*PIP2/(PIP2+Km20)/Cell
Km22a = 100.0; Kcat22a = 0.33Reaction: PIP3 + Akt => PIP3 + pAkt, Rate Law: Cell*Kcat22a*PIP3*Akt/(Akt+Km22a)/Cell
Km7 = 35954.3; Kcat7 = 32.644Reaction: pSOS + Ras => pSOS + pRas, Rate Law: Cell*Kcat7*pSOS*Ras/(Ras+Km7)/Cell
Kcat13 = 9.8537; Km13 = 1007300.0Reaction: pMEK + ERK => pMEK + pERK, Rate Law: Cell*Kcat13*pMEK*ERK/(ERK+Km13)/Cell
k35 = 3.433Reaction: pAPCpAxinGSK3BBCatenin => pAPCpAxinGSK3BpBCatenin, Rate Law: Cell*k35*pAPCpAxinGSK3BBCatenin/Cell
k26b = 3.85E-4Reaction: PKCD => null, Rate Law: Cell*k26b*PKCD/Cell
Kcat27a = 0.002Reaction: pERK + GSK3B => pERK + pGSK3B, Rate Law: Cell*Kcat27a*GSK3B*pERK/Cell
k36 = 3.433Reaction: pAPCpAxinGSK3BpBCatenin => pBCatenin + pAPCpAxinGSK3B, Rate Law: Cell*k36*pAPCpAxinGSK3BpBCatenin/Cell
V8a = 0.0717Reaction: null => Ras, Rate Law: Cell*V8a/Cell
Km9a = 62464.6; Kcat9a = 0.884096Reaction: pRas + Raf1 => pRas + pRaf1, Rate Law: Cell*Kcat9a*pRas*Raf1/(Raf1+Km9a)/Cell
Kcat12 = 2.8324; Km12 = 518750.0Reaction: pMEK + PP2A => MEK + PP2A, Rate Law: Cell*Kcat12*PP2A*pMEK/(pMEK+Km12)/Cell
k53 = 2.8833E-4; k52 = 3.85E-5; k54 = 1.5; k51 = 0.003465Reaction: PKCD + pERK + bEGFR + SOS => PKCD + pERK + bEGFR + pSOS, Rate Law: Cell*(k51*bEGFR+k52+k53*PKCD)/(1+pERK/k54)/Cell
k37a2 = 20.0; k37a1 = 0.01667Reaction: BCatenin + APC => APCBCatenin, Rate Law: Cell*(k37a1*APC*BCatenin-k37a2*APCBCatenin)/Cell
Kcat14 = 8.8912; Km14 = 3496500.0Reaction: pERK + PP2A => ERK + PP2A, Rate Law: Cell*Kcat14*PP2A*pERK/(pERK+Km14)/Cell
k312 = 0.01515; k311 = 0.001515Reaction: APCAxin + GSK3B => APCAxinGSK3B, Rate Law: Cell*(k311*GSK3B*APCAxin-k312*APCAxinGSK3B)/Cell
Kcat19b = 0.07711; Km19b = 272056.0Reaction: PI3K => pPI3K; pRas, Rate Law: Cell*Kcat19b*pRas*PI3K/(PI3K+Km19b)/Cell
k23b = 2.5Reaction: pC3G => C3G, Rate Law: Cell*k23b*pC3G/Cell
k15c = 0.00193Reaction: RKIP => null, Rate Law: Cell*k15c*RKIP/Cell
V37b = 0.00705Reaction: null => BCatenin, Rate Law: Cell*V37b/Cell
k37c = 4.283E-6Reaction: BCatenin => null, Rate Law: Cell*k37c*BCatenin/Cell
Km9b = 15.0; W = 0.0; k9b = 0.025Reaction: X + Raf1 => X + pRaf1, Rate Law: Cell*k9b*W*X*Raf1/(Km9b+Raf1)/Cell
Kcat6b = 1611.97; Km6b = 896896.0Reaction: pP90Rsk + pSOS => pP90Rsk + SOS, Rate Law: Cell*Kcat6b*pP90Rsk*pSOS/(pSOS+Km6b)/Cell
Kcat18b = 0.02137; Km18b = 763523.0Reaction: pERK + P90Rsk => pERK + pP90Rsk, Rate Law: Cell*Kcat18b*pERK*P90Rsk/(P90Rsk+Km18b)/Cell
k32b = 0.002217Reaction: pAPCpAxinGSK3B => APCAxinGSK3B, Rate Law: Cell*k32b*pAPCpAxinGSK3B/Cell
k22 = 0.121008; k21 = 2.18503E-5Reaction: fEGFR + EGF => bEGFR, Rate Law: Cell*(k21*EGF*fEGFR-k22*bEGFR)/Cell
V15b = 4.0Reaction: pRKIP => RKIP, Rate Law: Cell*V15b*pRKIP/Cell
k32a = 0.00445Reaction: APCAxinGSK3B => pAPCpAxinGSK3B, Rate Law: Cell*k32a*APCAxinGSK3B/Cell
Kcat21 = 5.5; Km21 = 0.08Reaction: PTEN + PIP3 => PTEN + PIP2, Rate Law: Cell*Kcat21*PTEN*PIP3/(PIP3+Km21)/Cell
Kcat10b = 15.1212; Km10b = 119355.0Reaction: pAkt + pRaf1 => pAkt + Raf1, Rate Law: Cell*Kcat10b*pAkt*pRaf1/(pRaf1+Km10b)/Cell
k11b2 = 120.0; k11b1 = 1.1167E-5Reaction: pRKIP + pRaf1 + MEK => pRKIP + pRaf1 + pMEK; RKIP, Rate Law: Cell*k11b1*pRaf1*MEK/(1+((RKIP-pRKIP)/k11b2)^2)/Cell
Km10a = 1061.7; Kcat10a = 0.12633Reaction: RafPPtase + pRaf1 => RafPPtase + Raf1, Rate Law: Cell*Kcat10a*RafPPtase*pRaf1/(pRaf1+Km10a)/Cell
k342 = 2.0; k341 = 0.01667Reaction: BCatenin + pAPCpAxinGSK3B => pAPCpAxinGSK3BBCatenin, Rate Law: Cell*(k341*pAPCpAxinGSK3B*BCatenin-k342*pAPCpAxinGSK3BBCatenin)/Cell
Km19a = 184912.0; Kcat19a = 10.6737Reaction: bEGFR + PI3K => bEGFR + pPI3K, Rate Law: Cell*Kcat19a*bEGFR*PI3K/(PI3K+Km19a)/Cell
k29 = 0.003Reaction: Dsha => Dshi, Rate Law: Cell*k29*Dsha/Cell
k381 = 0.01667; k382 = 0.5Reaction: TCF + BCatenin => TCFBCatenin, Rate Law: Cell*(k381*BCatenin*TCF-k382*TCFBCatenin)/Cell
Kcat16a = 0.8841; Km16a = 62645.0Reaction: pRas + BRaf => pRas + pBRaf, Rate Law: Cell*Kcat16a*pRas*BRaf/(BRaf+Km16a)/Cell
Kcat17a = 0.12633; Km17a = 1061.71Reaction: RafPPtase + pBRaf => RafPPtase + BRaf, Rate Law: Cell*Kcat17a*RafPPtase*pBRaf/(Km17a+RafPPtase)/Cell
Km25 = 1432400.0; Kcat25 = 1509.4Reaction: Rap1Gap + pRap1 => Rap1Gap + Rap1, Rate Law: Cell*Kcat25*Rap1Gap*pRap1/(pRap1+Km25)/Cell
V1 = 100.0Reaction: pEGFR => fEGFR, Rate Law: Cell*V1/Cell
V26a = 0.00154; k26a = 20.0Reaction: GSK3B => GSK3B + PKCD, Rate Law: Cell*V26a/(1+(GSK3B/k26a)^2.5)/Cell
Kcat27b = 0.04596Reaction: pAkt + GSK3B => pAkt + pGSK3B, Rate Law: Cell*Kcat27b*GSK3B*pAkt/Cell
k33b = 0.002783Reaction: Axin => null, Rate Law: Cell*k33b*Axin/Cell
k30 = 8.33E-4Reaction: Dsha + APCAxinGSK3B => GSK3B + APCAxin + Dsha, Rate Law: Cell*k30*Dsha*APCAxinGSK3B/Cell
k15a = 1.3Reaction: pERK + RKIP => pERK + pRKIP, Rate Law: Cell*k15a*pERK*(RKIP-pRKIP)/Cell
k33c1 = 1.37E-6; k33c2 = 1.667E-8Reaction: BCatenin + TCFBCatenin => BCatenin + TCFBCatenin + Axin, Rate Law: Cell*(k33c1+k33c2*(TCFBCatenin+BCatenin))/Cell

States:

NameDescription
pC3G[Complement C3; urn:miriam:bao:0002007]
PIP2[urn:miriam:chebi:0018348]
pPI3K[urn:miriam:omit:0027264; urn:miriam:bao:0002007; urn:miriam:omit:0010192]
Akt[RAC-alpha serine/threonine-protein kinase]
pSOS[Son of sevenless homolog 1; urn:miriam:bao:0002007]
pAPCpAxinGSK3BBCatenin[Axin-1; Glycogen synthase kinase-3 beta; Catenin beta-1; Adenomatous polyposis coli protein; urn:miriam:bao:0002007]
EGF[Pro-epidermal growth factor]
RafPPtase[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
pP90Rsk[Ribosomal protein S6 kinase alpha-1; urn:miriam:bao:0002007]
BRaf[Serine/threonine-protein kinase B-raf]
BCatenin[Catenin beta-1]
pAkt[RAC-alpha serine/threonine-protein kinase; urn:miriam:bao:0002007]
pRKIP[Phosphatidylethanolamine-binding protein 1; urn:miriam:bao:0002007]
RKIP[Phosphatidylethanolamine-binding protein 1]
PIP3[urn:miriam:chebi:0016618]
Ras[GTPase HRas]
pEGFR[Epidermal growth factor receptor; urn:miriam:bao:0002007]
PKCD[Protein kinase C delta type]
pMEK[Dual specificity mitogen-activated protein kinase kinase 1; urn:miriam:bao:0002007]
pRaf1[RAF proto-oncogene serine/threonine-protein kinase; urn:miriam:bao:0002007]
Rap1[Ras-related protein Rap-1A]
C3G[Rap guanine nucleotide exchange factor 1]
Dsha[Segment polarity protein dishevelled homolog DVL-1; urn:miriam:bao:0002007]
pAPCpAxinGSK3B[Adenomatous polyposis coli protein; Axin-1; Glycogen synthase kinase-3 beta; urn:miriam:bao:0002007]
P90Rsk[Ribosomal protein S6 kinase alpha-1]
Raf1[RAF proto-oncogene serine/threonine-protein kinase]
pGSK3B[Glycogen synthase kinase-3 beta; urn:miriam:bao:0002007]
PP2A[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
Dshi[Segment polarity protein dishevelled homolog DVL-1]
APCBCatenin[Catenin beta-1; Adenomatous polyposis coli protein]
pERK[Mitogen-activated protein kinase 1; urn:miriam:bao:0002007]
pRas[Ras-related protein R-Ras2; urn:miriam:bao:0002007]
APC[Adenomatous polyposis coli protein]
pBRaf[Serine/threonine-protein kinase B-raf; urn:miriam:bao:0002007]
GSK3B[Glycogen synthase kinase-3 beta]
ERK[Mitogen-activated protein kinase 3]
pRap1[Ras-related protein Rap-1A; urn:miriam:bao:0002007]
APCAxinGSK3B[Axin-1; Adenomatous polyposis coli protein; Glycogen synthase kinase-3 beta]
Axin[Axin-1]
pAPCpAxinGSK3BpBCatenin[Catenin beta-1; Glycogen synthase kinase-3 beta; Axin-1; Adenomatous polyposis coli protein; urn:miriam:bao:0002007]

Padala2017- ERK, PI3K/Akt and Wnt signalling network (PTEN mutation): BIOMD0000000655v0.0.1

Padala2017- ERK, PI3K/Akt and Wnt signalling network (PTEN mutation)Crosstalk model of the ERK, Wnt and Akt Signalling p…

Details

Perturbations in molecular signaling pathways are a result of genetic or epigenetic alterations, which may lead to malignant transformation of cells. Despite cellular robustness, specific genetic or epigenetic changes of any gene can trigger a cascade of failures, which result in the malfunctioning of cell signaling pathways and lead to cancer phenotypes. The extent of cellular robustness has a link with the architecture of the network such as feedback and feedforward loops. Perturbation in components within feedback loops causes a transition from a regulated to a persistently activated state and results in uncontrolled cell growth. This work represents the mathematical and quantitative modeling of ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk to show the dynamics of signaling responses during genetic and epigenetic changes in cancer. ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk networks include both intra and inter-pathway feedback loops which function in a controlled fashion in a healthy cell. Our results show that cancerous perturbations of components such as EGFR, Ras, B-Raf, PTEN, and components of the destruction complex cause extreme fragility in the network and constitutively activate inter-pathway positive feedback loops. We observed that the aberrant signaling response due to the failure of specific network components is transmitted throughout the network via crosstalk, generating an additive effect on cancer growth and proliferation. link: http://identifiers.org/pubmed/28367561

Parameters:

NameDescription
k33a2 = 0.8333; k33a1 = 0.01667Reaction: APC + Axin => APCAxin, Rate Law: Cell*(k33a1*Axin*APC-k33a2*APCAxin)/Cell
Kcat24 = 32.344; Km24 = 35954.3Reaction: pC3G + Rap1 => pC3G + pRap1, Rate Law: Cell*Kcat24*pC3G*Rap1/(Rap1+Km24)/Cell
Kcat17b = 15.1212; Km17b = 119355.0Reaction: pAkt + pBRaf => pAkt + BRaf, Rate Law: Cell*Kcat17b*pBRaf*pAkt/(Km17b+pBRaf)/Cell
Km16b = 62464.6; Kcat16b = 0.8841Reaction: pRap1 + BRaf => pRap1 + pBRaf, Rate Law: Cell*Kcat16b*pRap1*BRaf/(BRaf+Km16b)/Cell
k6a = 2.5Reaction: pSOS => SOS, Rate Law: Cell*k6a*pSOS/Cell
k3 = 0.00125Reaction: fEGFR => null, Rate Law: Cell*k3*fEGFR/Cell
Km22b = 100.0; Kcat22b = 48.667Reaction: pAkt => Akt, Rate Law: Cell*Kcat22b*pAkt/(Km22b+pAkt)/Cell
Kcat23a = 694.73; Km23a = 6086100.0Reaction: bEGFR + C3G => bEGFR + pC3G, Rate Law: Cell*Kcat23a*bEGFR*C3G/(C3G+Km23a)/Cell
Kcat8b = 1509.36; Km8b = 1432410.0Reaction: RasGap + pRas => RasGap + Ras, Rate Law: Cell*Kcat8b*RasGap*pRas/(pRas+Km8b)/Cell
k18a = 0.005Reaction: pP90Rsk => P90Rsk, Rate Law: Cell*k18a*pP90Rsk/Cell
Km20 = 4.0; Kcat20 = 4.0Reaction: pPI3K + PIP2 => pPI3K + PIP3, Rate Law: Cell*Kcat20*pPI3K*PIP2/(PIP2+Km20)/Cell
Km22a = 100.0; Kcat22a = 0.33Reaction: PIP3 + Akt => PIP3 + pAkt, Rate Law: Cell*Kcat22a*PIP3*Akt/(Akt+Km22a)/Cell
Km7 = 35954.3; Kcat7 = 32.644Reaction: pSOS + Ras => pSOS + pRas, Rate Law: Cell*Kcat7*pSOS*Ras/(Ras+Km7)/Cell
Kcat13 = 9.8537; Km13 = 1007300.0Reaction: pMEK + ERK => pMEK + pERK, Rate Law: Cell*Kcat13*pMEK*ERK/(ERK+Km13)/Cell
k35 = 3.433Reaction: pAPCpAxinGSK3BBCatenin => pAPCpAxinGSK3BpBCatenin, Rate Law: Cell*k35*pAPCpAxinGSK3BBCatenin/Cell
k19c = 0.005Reaction: pPI3K => PI3K, Rate Law: Cell*k19c*pPI3K/Cell
k26b = 3.85E-4Reaction: PKCD => null, Rate Law: Cell*k26b*PKCD/Cell
k36 = 3.433Reaction: pAPCpAxinGSK3BpBCatenin => pBCatenin + pAPCpAxinGSK3B, Rate Law: Cell*k36*pAPCpAxinGSK3BpBCatenin/Cell
Km9a = 62464.6; Kcat9a = 0.884096Reaction: pRas + Raf1 => pRas + pRaf1, Rate Law: Cell*Kcat9a*pRas*Raf1/(Raf1+Km9a)/Cell
Kcat12 = 2.8324; Km12 = 518750.0Reaction: pMEK + PP2A => MEK + PP2A, Rate Law: Cell*Kcat12*PP2A*pMEK/(pMEK+Km12)/Cell
k53 = 2.8833E-4; k52 = 3.85E-5; k54 = 1.5; k51 = 0.003465Reaction: PKCD + pERK + bEGFR + SOS => PKCD + pERK + bEGFR + pSOS, Rate Law: Cell*(k51*bEGFR+k52+k53*PKCD)/(1+pERK/k54)/Cell
k37a2 = 20.0; k37a1 = 0.01667Reaction: BCatenin + APC => APCBCatenin, Rate Law: Cell*(k37a1*APC*BCatenin-k37a2*APCBCatenin)/Cell
Kcat14 = 8.8912; Km14 = 3496500.0Reaction: pERK + PP2A => ERK + PP2A, Rate Law: Cell*Kcat14*PP2A*pERK/(pERK+Km14)/Cell
k312 = 0.01515; k311 = 0.001515Reaction: APCAxin + GSK3B => APCAxinGSK3B, Rate Law: Cell*(k311*GSK3B*APCAxin-k312*APCAxinGSK3B)/Cell
Kcat19b = 0.07711; Km19b = 272056.0Reaction: PI3K => pPI3K; pRas, Rate Law: Cell*Kcat19b*pRas*PI3K/(PI3K+Km19b)/Cell
k23b = 2.5Reaction: pC3G => C3G, Rate Law: Cell*k23b*pC3G/Cell
k37c = 4.283E-6Reaction: BCatenin => null, Rate Law: Cell*k37c*BCatenin/Cell
V37b = 0.00705Reaction: null => BCatenin, Rate Law: Cell*V37b/Cell
Km9b = 15.0; W = 0.0; k9b = 0.025Reaction: X + Raf1 => X + pRaf1, Rate Law: Cell*k9b*W*X*Raf1/(Km9b+Raf1)/Cell
Kcat6b = 1611.97; Km6b = 896896.0Reaction: pP90Rsk + pSOS => pP90Rsk + SOS, Rate Law: Cell*Kcat6b*pP90Rsk*pSOS/(pSOS+Km6b)/Cell
Kcat18b = 0.02137; Km18b = 763523.0Reaction: pERK + P90Rsk => pERK + pP90Rsk, Rate Law: Cell*Kcat18b*pERK*P90Rsk/(P90Rsk+Km18b)/Cell
k32b = 0.002217Reaction: pAPCpAxinGSK3B => APCAxinGSK3B, Rate Law: Cell*k32b*pAPCpAxinGSK3B/Cell
k22 = 0.121008; k21 = 2.18503E-5Reaction: fEGFR + EGF => bEGFR, Rate Law: Cell*(k21*EGF*fEGFR-k22*bEGFR)/Cell
V15b = 4.0Reaction: pRKIP => RKIP, Rate Law: Cell*V15b*pRKIP/Cell
k32a = 0.00445Reaction: APCAxinGSK3B => pAPCpAxinGSK3B, Rate Law: Cell*k32a*APCAxinGSK3B/Cell
Kcat10b = 15.1212; Km10b = 119355.0Reaction: pAkt + pRaf1 => pAkt + Raf1, Rate Law: Cell*Kcat10b*pAkt*pRaf1/(pRaf1+Km10b)/Cell
k11b2 = 120.0; k11b1 = 1.1167E-5Reaction: pRKIP + pRaf1 + MEK => pRKIP + pRaf1 + pMEK; RKIP, Rate Law: Cell*k11b1*pRaf1*MEK/(1+((RKIP-pRKIP)/k11b2)^2)/Cell
k342 = 2.0; k341 = 0.01667Reaction: BCatenin + pAPCpAxinGSK3B => pAPCpAxinGSK3BBCatenin, Rate Law: Cell*(k341*pAPCpAxinGSK3B*BCatenin-k342*pAPCpAxinGSK3BBCatenin)/Cell
Km10a = 1061.7; Kcat10a = 0.12633Reaction: RafPPtase + pRaf1 => RafPPtase + Raf1, Rate Law: Cell*Kcat10a*RafPPtase*pRaf1/(pRaf1+Km10a)/Cell
Km19a = 184912.0; Kcat19a = 10.6737Reaction: bEGFR + PI3K => bEGFR + pPI3K, Rate Law: Cell*Kcat19a*bEGFR*PI3K/(PI3K+Km19a)/Cell
Kcat11a = 185.76; Km11a = 4768400.0Reaction: pBRaf + MEK => pBRaf + pMEK, Rate Law: Cell*Kcat11a*pBRaf*MEK/(MEK+Km11a)/Cell
k4 = 0.2Reaction: bEGFR => null, Rate Law: Cell*k4*bEGFR/Cell
k381 = 0.01667; k382 = 0.5Reaction: TCF + BCatenin => TCFBCatenin, Rate Law: Cell*(k381*BCatenin*TCF-k382*TCFBCatenin)/Cell
Kcat16a = 0.8841; Km16a = 62645.0Reaction: pRas + BRaf => pRas + pBRaf, Rate Law: Cell*Kcat16a*pRas*BRaf/(BRaf+Km16a)/Cell
V1 = 100.0Reaction: pEGFR => fEGFR, Rate Law: Cell*V1/Cell
Km25 = 1432400.0; Kcat25 = 1509.4Reaction: Rap1Gap + pRap1 => Rap1Gap + Rap1, Rate Law: Cell*Kcat25*Rap1Gap*pRap1/(pRap1+Km25)/Cell
Kcat17a = 0.12633; Km17a = 1061.71Reaction: RafPPtase + pBRaf => RafPPtase + BRaf, Rate Law: Cell*Kcat17a*RafPPtase*pBRaf/(Km17a+RafPPtase)/Cell
V26a = 0.00154; k26a = 20.0Reaction: GSK3B => GSK3B + PKCD, Rate Law: Cell*V26a/(1+(GSK3B/k26a)^2.5)/Cell
Kcat27d = 0.01541Reaction: pGSK3B => GSK3B, Rate Law: Cell*Kcat27d*pGSK3B/Cell
k41 = 0.00695Reaction: pBCatenin => null, Rate Law: Cell*k41*pBCatenin/Cell
Kcat27b = 0.04596Reaction: pAkt + GSK3B => pAkt + pGSK3B, Rate Law: Cell*Kcat27b*GSK3B*pAkt/Cell
k33b = 0.002783Reaction: Axin => null, Rate Law: Cell*k33b*Axin/Cell
k30 = 8.33E-4Reaction: Dsha + APCAxinGSK3B => GSK3B + APCAxin + Dsha, Rate Law: Cell*k30*Dsha*APCAxinGSK3B/Cell
k15a = 1.3Reaction: pERK + RKIP => pERK + pRKIP, Rate Law: Cell*k15a*pERK*(RKIP-pRKIP)/Cell
k33c1 = 1.37E-6; k33c2 = 1.667E-8Reaction: BCatenin + TCFBCatenin => BCatenin + TCFBCatenin + Axin, Rate Law: Cell*(k33c1+k33c2*(TCFBCatenin+BCatenin))/Cell

States:

NameDescription
pC3G[Complement C3; phosphorylated]
pBCatenin[Catenin beta-1; phosphorylated]
bEGFR[Epidermal growth factor receptor; Pro-epidermal growth factor]
pPI3K[0027264; phosphorylated]
APCAxin[Axin-1; Adenomatous polyposis coli protein]
Akt[RAC-alpha serine/threonine-protein kinase]
pSOS[Son of sevenless homolog 1; phosphorylated]
pAPCpAxinGSK3BBCatenin[Axin-1; Glycogen synthase kinase-3 beta; Catenin beta-1; Adenomatous polyposis coli protein; phosphorylated]
EGF[Pro-epidermal growth factor]
pP90Rsk[Ribosomal protein S6 kinase alpha-1; phosphorylated]
pMEK[Dual specificity mitogen-activated protein kinase kinase 1; phosphorylated]
BCatenin[Catenin beta-1]
pAkt[RAC-alpha serine/threonine-protein kinase; phosphorylated]
pRKIP[Phosphatidylethanolamine-binding protein 1; phosphorylated]
BRaf[Serine/threonine-protein kinase B-raf]
RKIP[Phosphatidylethanolamine-binding protein 1]
PIP3[0016618]
pEGFR[Epidermal growth factor receptor; phosphorylated]
PKCD[Protein kinase C delta type]
pRaf1[RAF proto-oncogene serine/threonine-protein kinase; phosphorylated]
MEK[Dual specificity mitogen-activated protein kinase kinase 1]
C3G[Rap guanine nucleotide exchange factor 1]
Dsha[Segment polarity protein dishevelled homolog DVL-1; phosphorylated]
pAPCpAxinGSK3B[Adenomatous polyposis coli protein; Axin-1; Glycogen synthase kinase-3 beta; phosphorylated]
P90Rsk[Ribosomal protein S6 kinase alpha-1]
pGSK3B[Glycogen synthase kinase-3 beta; phosphorylated]
Raf1[RAF proto-oncogene serine/threonine-protein kinase]
PP2A[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
pRas[Ras-related protein R-Ras2; phosphorylated]
APCBCatenin[Catenin beta-1; Adenomatous polyposis coli protein]
pERK[Mitogen-activated protein kinase 1; phosphorylated]
fEGFR[Epidermal growth factor receptor]
APC[Adenomatous polyposis coli protein]
pBRaf[Serine/threonine-protein kinase B-raf; phosphorylated]
GSK3B[Glycogen synthase kinase-3 beta]
ERK[Mitogen-activated protein kinase 3]
pRap1[Ras-related protein Rap-1A; phosphorylated]
APCAxinGSK3B[Axin-1; Adenomatous polyposis coli protein; Glycogen synthase kinase-3 beta]
Axin[Axin-1]
pAPCpAxinGSK3BpBCatenin[Catenin beta-1; Glycogen synthase kinase-3 beta; Axin-1; Adenomatous polyposis coli protein; phosphorylated]

Padala2017- ERK, PI3K/Akt and Wnt signalling network (Ras mutated): BIOMD0000000654v0.0.1

Padala2017- ERK, PI3K/Akt and Wnt signalling network (Ras mutated)Crosstalk model of the ERK, Wnt and Akt signalling pat…

Details

Perturbations in molecular signaling pathways are a result of genetic or epigenetic alterations, which may lead to malignant transformation of cells. Despite cellular robustness, specific genetic or epigenetic changes of any gene can trigger a cascade of failures, which result in the malfunctioning of cell signaling pathways and lead to cancer phenotypes. The extent of cellular robustness has a link with the architecture of the network such as feedback and feedforward loops. Perturbation in components within feedback loops causes a transition from a regulated to a persistently activated state and results in uncontrolled cell growth. This work represents the mathematical and quantitative modeling of ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk to show the dynamics of signaling responses during genetic and epigenetic changes in cancer. ERK, PI3K/Akt, and Wnt/β-catenin signaling crosstalk networks include both intra and inter-pathway feedback loops which function in a controlled fashion in a healthy cell. Our results show that cancerous perturbations of components such as EGFR, Ras, B-Raf, PTEN, and components of the destruction complex cause extreme fragility in the network and constitutively activate inter-pathway positive feedback loops. We observed that the aberrant signaling response due to the failure of specific network components is transmitted throughout the network via crosstalk, generating an additive effect on cancer growth and proliferation. link: http://identifiers.org/pubmed/28367561

Parameters:

NameDescription
k27c = 1.5E-4Reaction: RKIP => RKIP + GSK3B, Rate Law: Cell*k27c*RKIP/Cell
k33a2 = 0.8333; k33a1 = 0.01667Reaction: APC + Axin => APCAxin, Rate Law: Cell*(k33a1*Axin*APC-k33a2*APCAxin)/Cell
W = 0.0; k28 = 0.003Reaction: Dshi => Dsha, Rate Law: Cell*k28*Dshi*W/Cell
Kcat24 = 32.344; Km24 = 35954.3Reaction: pC3G + Rap1 => pC3G + pRap1, Rate Law: Cell*Kcat24*pC3G*Rap1/(Rap1+Km24)/Cell
Kcat17b = 15.1212; Km17b = 119355.0Reaction: pAkt + pBRaf => pAkt + BRaf, Rate Law: Cell*Kcat17b*pBRaf*pAkt/(Km17b+pBRaf)/Cell
Km16b = 62464.6; Kcat16b = 0.8841Reaction: pRap1 + BRaf => pRap1 + pBRaf, Rate Law: Cell*Kcat16b*pRap1*BRaf/(BRaf+Km16b)/Cell
k6a = 2.5Reaction: pSOS => SOS, Rate Law: Cell*k6a*pSOS/Cell
Kcat23a = 694.73; Km23a = 6086100.0Reaction: bEGFR + C3G => bEGFR + pC3G, Rate Law: Cell*Kcat23a*bEGFR*C3G/(C3G+Km23a)/Cell
Km22b = 100.0; Kcat22b = 48.667Reaction: pAkt => Akt, Rate Law: Cell*Kcat22b*pAkt/(Km22b+pAkt)/Cell
k18a = 0.005Reaction: pP90Rsk => P90Rsk, Rate Law: Cell*k18a*pP90Rsk/Cell
Km20 = 4.0; Kcat20 = 4.0Reaction: pPI3K + PIP2 => pPI3K + PIP3, Rate Law: Cell*Kcat20*pPI3K*PIP2/(PIP2+Km20)/Cell
Km22a = 100.0; Kcat22a = 0.33Reaction: PIP3 + Akt => PIP3 + pAkt, Rate Law: Cell*Kcat22a*PIP3*Akt/(Akt+Km22a)/Cell
Km7 = 35954.3; Kcat7 = 32.644Reaction: pSOS + Ras => pSOS + pRas, Rate Law: Cell*Kcat7*pSOS*Ras/(Ras+Km7)/Cell
k35 = 3.433Reaction: pAPCpAxinGSK3BBCatenin => pAPCpAxinGSK3BpBCatenin, Rate Law: Cell*k35*pAPCpAxinGSK3BBCatenin/Cell
k19c = 0.005Reaction: pPI3K => PI3K, Rate Law: Cell*k19c*pPI3K/Cell
k26b = 3.85E-4Reaction: PKCD => null, Rate Law: Cell*k26b*PKCD/Cell
k36 = 3.433Reaction: pAPCpAxinGSK3BpBCatenin => pBCatenin + pAPCpAxinGSK3B, Rate Law: Cell*k36*pAPCpAxinGSK3BpBCatenin/Cell
V8a = 0.0717Reaction: null => Ras, Rate Law: Cell*V8a/Cell
Km9a = 62464.6; Kcat9a = 0.884096Reaction: pRas + Raf1 => pRas + pRaf1, Rate Law: Cell*Kcat9a*pRas*Raf1/(Raf1+Km9a)/Cell
Kcat12 = 2.8324; Km12 = 518750.0Reaction: pMEK + PP2A => MEK + PP2A, Rate Law: Cell*Kcat12*PP2A*pMEK/(pMEK+Km12)/Cell
k53 = 2.8833E-4; k52 = 3.85E-5; k54 = 1.5; k51 = 0.003465Reaction: PKCD + pERK + bEGFR + SOS => PKCD + pERK + bEGFR + pSOS, Rate Law: Cell*(k51*bEGFR+k52+k53*PKCD)/(1+pERK/k54)/Cell
k37a2 = 20.0; k37a1 = 0.01667Reaction: BCatenin + APC => APCBCatenin, Rate Law: Cell*(k37a1*APC*BCatenin-k37a2*APCBCatenin)/Cell
Kcat14 = 8.8912; Km14 = 3496500.0Reaction: pERK + PP2A => ERK + PP2A, Rate Law: Cell*Kcat14*PP2A*pERK/(pERK+Km14)/Cell
k312 = 0.01515; k311 = 0.001515Reaction: APCAxin + GSK3B => APCAxinGSK3B, Rate Law: Cell*(k311*GSK3B*APCAxin-k312*APCAxinGSK3B)/Cell
Kcat19b = 0.07711; Km19b = 272056.0Reaction: PI3K => pPI3K; pRas, Rate Law: Cell*Kcat19b*pRas*PI3K/(PI3K+Km19b)/Cell
k23b = 2.5Reaction: pC3G => C3G, Rate Law: Cell*k23b*pC3G/Cell
Km9b = 15.0; W = 0.0; k9b = 0.025Reaction: X + Raf1 => X + pRaf1, Rate Law: Cell*k9b*W*X*Raf1/(Km9b+Raf1)/Cell
Kcat6b = 1611.97; Km6b = 896896.0Reaction: pP90Rsk + pSOS => pP90Rsk + SOS, Rate Law: Cell*Kcat6b*pP90Rsk*pSOS/(pSOS+Km6b)/Cell
Kcat18b = 0.02137; Km18b = 763523.0Reaction: pERK + P90Rsk => pERK + pP90Rsk, Rate Law: Cell*Kcat18b*pERK*P90Rsk/(P90Rsk+Km18b)/Cell
k22 = 0.121008; k21 = 2.18503E-5Reaction: fEGFR + EGF => bEGFR, Rate Law: Cell*(k21*EGF*fEGFR-k22*bEGFR)/Cell
V15b = 4.0Reaction: pRKIP => RKIP, Rate Law: Cell*V15b*pRKIP/Cell
k32a = 0.00445Reaction: APCAxinGSK3B => pAPCpAxinGSK3B, Rate Law: Cell*k32a*APCAxinGSK3B/Cell
Kcat21 = 5.5; Km21 = 0.08Reaction: PTEN + PIP3 => PTEN + PIP2, Rate Law: Cell*Kcat21*PTEN*PIP3/(PIP3+Km21)/Cell
Kcat10b = 15.1212; Km10b = 119355.0Reaction: pAkt + pRaf1 => pAkt + Raf1, Rate Law: Cell*Kcat10b*pAkt*pRaf1/(pRaf1+Km10b)/Cell
k11b2 = 120.0; k11b1 = 1.1167E-5Reaction: pRKIP + pRaf1 + MEK => pRKIP + pRaf1 + pMEK; RKIP, Rate Law: Cell*k11b1*pRaf1*MEK/(1+((RKIP-pRKIP)/k11b2)^2)/Cell
Km10a = 1061.7; Kcat10a = 0.12633Reaction: RafPPtase + pRaf1 => RafPPtase + Raf1, Rate Law: Cell*Kcat10a*RafPPtase*pRaf1/(pRaf1+Km10a)/Cell
Kcat11a = 185.76; Km11a = 4768400.0Reaction: pBRaf + MEK => pBRaf + pMEK, Rate Law: Cell*Kcat11a*pBRaf*MEK/(MEK+Km11a)/Cell
Km19a = 184912.0; Kcat19a = 10.6737Reaction: bEGFR + PI3K => bEGFR + pPI3K, Rate Law: Cell*Kcat19a*bEGFR*PI3K/(PI3K+Km19a)/Cell
k29 = 0.003Reaction: Dsha => Dshi, Rate Law: Cell*k29*Dsha/Cell
k381 = 0.01667; k382 = 0.5Reaction: TCF + BCatenin => TCFBCatenin, Rate Law: Cell*(k381*BCatenin*TCF-k382*TCFBCatenin)/Cell
Kcat16a = 0.8841; Km16a = 62645.0Reaction: pRas + BRaf => pRas + pBRaf, Rate Law: Cell*Kcat16a*pRas*BRaf/(BRaf+Km16a)/Cell
Kcat17a = 0.12633; Km17a = 1061.71Reaction: RafPPtase + pBRaf => RafPPtase + BRaf, Rate Law: Cell*Kcat17a*RafPPtase*pBRaf/(Km17a+RafPPtase)/Cell
Km25 = 1432400.0; Kcat25 = 1509.4Reaction: Rap1Gap + pRap1 => Rap1Gap + Rap1, Rate Law: Cell*Kcat25*Rap1Gap*pRap1/(pRap1+Km25)/Cell
V26a = 0.00154; k26a = 20.0Reaction: GSK3B => GSK3B + PKCD, Rate Law: Cell*V26a/(1+(GSK3B/k26a)^2.5)/Cell
Kcat27d = 0.01541Reaction: pGSK3B => GSK3B, Rate Law: Cell*Kcat27d*pGSK3B/Cell
k41 = 0.00695Reaction: pBCatenin => null, Rate Law: Cell*k41*pBCatenin/Cell
Kcat27b = 0.04596Reaction: pAkt + GSK3B => pAkt + pGSK3B, Rate Law: Cell*Kcat27b*GSK3B*pAkt/Cell
Km39 = 15.0; k39 = 0.01Reaction: TCFBCatenin => X + TCFBCatenin, Rate Law: Cell*k39*TCFBCatenin^2/(Km39^2+TCFBCatenin^2)/Cell
k30 = 8.33E-4Reaction: Dsha + APCAxinGSK3B => GSK3B + APCAxin + Dsha, Rate Law: Cell*k30*Dsha*APCAxinGSK3B/Cell
k15a = 1.3Reaction: pERK + RKIP => pERK + pRKIP, Rate Law: Cell*k15a*pERK*(RKIP-pRKIP)/Cell
k33c1 = 1.37E-6; k33c2 = 1.667E-8Reaction: BCatenin + TCFBCatenin => BCatenin + TCFBCatenin + Axin, Rate Law: Cell*(k33c1+k33c2*(TCFBCatenin+BCatenin))/Cell

States:

NameDescription
pC3G[Complement C3; phosphorylated]
pBCatenin[Catenin beta-1; phosphorylated]
PIP2[0018348]
PTEN[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN]
pPI3K[0027264; phosphorylated]
bEGFR[Epidermal growth factor receptor; Pro-epidermal growth factor]
APCAxin[Axin-1; Adenomatous polyposis coli protein]
pSOS[Son of sevenless homolog 1; phosphorylated]
EGF[Pro-epidermal growth factor]
RafPPtase[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
pP90Rsk[Ribosomal protein S6 kinase alpha-1; phosphorylated]
BRaf[Serine/threonine-protein kinase B-raf]
pAkt[RAC-alpha serine/threonine-protein kinase; phosphorylated]
pRKIP[Phosphatidylethanolamine-binding protein 1; phosphorylated]
PIP3[0016618]
PKCD[Protein kinase C delta type]
RKIP[Phosphatidylethanolamine-binding protein 1]
Ras[GTPase HRas; 0010192]
SOS[Son of sevenless homolog 1]
pRaf1[RAF proto-oncogene serine/threonine-protein kinase; phosphorylated]
MEK[Dual specificity mitogen-activated protein kinase kinase 1]
PI3K[0027264]
C3G[Rap guanine nucleotide exchange factor 1]
Rap1[Ras-related protein Rap-1A]
TCFBCatenin[Lymphoid enhancer-binding factor 1; Catenin beta-1]
Dsha[Segment polarity protein dishevelled homolog DVL-1; phosphorylated]
XX
TCF[Lymphoid enhancer-binding factor 1]
P90Rsk[Ribosomal protein S6 kinase alpha-1]
Raf1[RAF proto-oncogene serine/threonine-protein kinase]
pAPCpAxinGSK3B[Adenomatous polyposis coli protein; Axin-1; Glycogen synthase kinase-3 beta; phosphorylated]
PP2A[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
Dshi[Segment polarity protein dishevelled homolog DVL-1]
pRas[Ras-related protein R-Ras2; phosphorylated; 0010192]
APCBCatenin[Catenin beta-1; Adenomatous polyposis coli protein]
APC[Adenomatous polyposis coli protein]
pBRaf[Serine/threonine-protein kinase B-raf; phosphorylated]
GSK3B[Glycogen synthase kinase-3 beta]
pRap1[Ras-related protein Rap-1A; phosphorylated]
pAPCpAxinGSK3BpBCatenin[Catenin beta-1; Glycogen synthase kinase-3 beta; Axin-1; Adenomatous polyposis coli protein; phosphorylated]
APCAxinGSK3B[Axin-1; Adenomatous polyposis coli protein; Glycogen synthase kinase-3 beta]

Paiva2020 - SEIAHRD model of transmission dynamics of COVID-19: BIOMD0000000960v0.0.1

This paper proposes a dynamic model to describe and forecast the dynamics of the coronavirus disease COVID-19 transmissi…

Details

This paper proposes a dynamic model to describe and forecast the dynamics of the coronavirus disease COVID-19 transmission. The model is based on an approach previously used to describe the Middle East Respiratory Syndrome (MERS) epidemic. This methodology is used to describe the COVID-19 dynamics in six countries where the pandemic is widely spread, namely China, Italy, Spain, France, Germany, and the USA. For this purpose, data from the European Centre for Disease Prevention and Control (ECDC) are adopted. It is shown how the model can be used to forecast new infection cases and new deceased and how the uncertainties associated to this prediction can be quantified. This approach has the advantage of being relatively simple, grouping in few mathematical parameters the many conditions which affect the spreading of the disease. On the other hand, it requires previous data from the disease transmission in the country, being better suited for regions where the epidemic is not at a very early stage. With the estimated parameters at hand, one can use the model to predict the evolution of the disease, which in turn enables authorities to plan their actions. Moreover, one key advantage is the straightforward interpretation of these parameters and their influence over the evolution of the disease, which enables altering some of them, so that one can evaluate the effect of public policy, such as social distancing. The results presented for the selected countries confirm the accuracy to perform predictions. link: http://identifiers.org/pubmed/32735581

Palini2011_Minimal_2_Feedback_Model: BIOMD0000000325v0.0.1

This is the model of the minmal 2 feedback switch described in the article: **Synthetic conversion of a graded recepto…

Details

The ability to engineer an all-or-none cellular response to a given signaling ligand is important in applications ranging from biosensing to tissue engineering. However, synthetic gene network 'switches' have been limited in their applicability and tunability due to their reliance on specific components to function. Here, we present a strategy for reversible switch design that instead relies only on a robust, easily constructed network topology with two positive feedback loops and we apply the method to create highly ultrasensitive (n(H)>20), bistable cellular responses to a synthetic ligand/receptor complex. Independent modulation of the two feedback strengths enables rational tuning and some decoupling of steady-state (ultrasensitivity, signal amplitude, switching threshold, and bistability) and kinetic (rates of system activation and deactivation) response properties. Our integrated computational and synthetic biology approach elucidates design rules for building cellular switches with desired properties, which may be of utility in engineering signal-transduction pathways. link: http://identifiers.org/pubmed/21451590

Parameters:

NameDescription
kdegR = 0.005Reaction: R =>, Rate Law: cell*kdegR*R
kdegC = 0.01Reaction: C =>, Rate Law: cell*kdegC*C
kdegX = 0.005Reaction: X =>, Rate Law: cell*kdegX*X
Rs = 3.0; BR = 0.005; KD = 200.0Reaction: => R; A, Rate Law: cell*(BR+Rs*A/(KD+A))
k3 = 45.0Reaction: X => C + A, Rate Law: cell*k3*X
kdegA = 0.005Reaction: A =>, Rate Law: cell*kdegA*A
koff = 0.05; kon = 0.001Reaction: R + L => C, Rate Law: cell*(kon*L*R-koff*C)
kdegI = 0.005Reaction: I =>, Rate Law: cell*kdegI*I
k2 = 5.0; k1 = 1.0Reaction: C + I => X, Rate Law: cell*(k1*C*I-k2*X)
BI = 0.005; TFs = 3.0; KD = 200.0Reaction: => I; A, Rate Law: cell*(BI+TFs*A/(KD+A))

States:

NameDescription
I[Transcription factor SKN7]
A[obsolete transcription activator activity; phosphorylated L-histidine; Transcription factor SKN7]
X[transmembrane histidine kinase cytokinin receptor activity; Transcription factor SKN7; Histidine kinase 4; N(6)-isopentenyladenosine]
C[transmembrane histidine kinase cytokinin receptor activity; Histidine kinase 4; N(6)-isopentenyladenosine]
L[cytokinin; N(6)-isopentenyladenosine; CHEMBL1163500]
R[Histidine kinase 4]

Palmer2008 - Negative Feedback in IL-7 mediated Jak-Stat signaling: BIOMD0000000968v0.0.1

Interleukin-7 (IL-7) is an essential cytokine for the development and homeostatic maintenance of T and B lymphocytes. Bi…

Details

Interleukin-7 (IL-7) is an essential cytokine for the development and homeostatic maintenance of T and B lymphocytes. Binding of IL-7 to its cognate receptor, the IL-7 receptor (IL-7R), activates multiple pathways that regulate lymphocyte survival, glucose uptake, proliferation and differentiation. There has been much interest in understanding how IL-7 receptor signaling is modulated at multiple interconnected network levels. This review examines how the strength of the signal through the IL-7 receptor is modulated in T and B cells, including the use of shared receptor components, signaling crosstalk, shared interaction domains, feedback loops, integrated gene regulation, multimerization and ligand competition. We discuss how these network control mechanisms could integrate to govern the properties of IL-7R signaling in lymphocytes in health and disease. Analysis of IL-7 receptor signaling at a network level in a systematic manner will allow for a comprehensive approach to understanding the impact of multiple signaling pathways on lymphocyte biology. link: http://identifiers.org/pubmed/18445337

Palmer2014 - Effect of IL-1β-Blocking therapies in T2DM - Disease Condition: BIOMD0000000620v0.0.1

Palmer2014 - Effect of IL-1β-Blocking therapies in T2DM - Disease Condition This is the model with disease state initia…

Details

Recent clinical studies suggest sustained treatment effects of interleukin-1β (IL-1β)-blocking therapies in type 2 diabetes mellitus. The underlying mechanisms of these effects, however, remain underexplored. Using a quantitative systems pharmacology modeling approach, we combined ex vivo data of IL-1β effects on β-cell function and turnover with a disease progression model of the long-term interactions between insulin, glucose, and β-cell mass in type 2 diabetes mellitus. We then simulated treatment effects of the IL-1 receptor antagonist anakinra. The result was a substantial and partly sustained symptomatic improvement in β-cell function, and hence also in HbA1C, fasting plasma glucose, and proinsulin-insulin ratio, and a small increase in β-cell mass. We propose that improved β-cell function, rather than mass, is likely to explain the main IL-1β-blocking effects seen in current clinical data, but that improved β-cell mass might result in disease-modifying effects not clearly distinguishable until >1 year after treatment. link: http://identifiers.org/pubmed/24918743

Parameters:

NameDescription
taus = 0.5Reaction: TigB =>, Rate Law: taus*TigB
Kxg = 1.6E-5Reaction: Glucose =>, Rate Law: Kxg*Glucose
placebo_on = 0.0; kplacebo = 0.00137Reaction: => IL1b, Rate Law: placebo_on*kplacebo
vfg = 4.0; tauf = 0.5; kmf = 0.021; IL1R = 0.02341920375; kf = 0.00957754; kmfg = 9.0; xfg = 4.0; vf = 0.4Reaction: => f; Glucose, Rate Law: tauf*kf*(1+vfg*Glucose^xfg/(kmfg^xfg+Glucose^xfg))*(1+vf*IL1R/(kmf+IL1R))
replication = 5.12314779E-4Reaction: => B, Rate Law: replication*B
Kxgi = 2.24E-5Reaction: Glucose => ; Insulin, Rate Law: Kxgi*Insulin*Glucose
Tgl = 0.025405Reaction: => Glucose, Rate Law: Tgl
Kglucose = 2.92E-4; lambda = 0.743; Kin = 1.05; Ktr = 0.12Reaction: rbc1 = (Kin-Ktr*rbc1)-Kglucose*Glucose^lambda*rbc1, Rate Law: (Kin-Ktr*rbc1)-Kglucose*Glucose^lambda*rbc1
Gh = 9.0; vh = 4.0Reaction: => Proinsulin; TigB, B, f, Glucose, Rate Law: f*(Glucose/Gh)^vh/(1+(Glucose/Gh)^vh)*TigB*B
apoptosis = 7.543653797E-4Reaction: B =>, Rate Law: apoptosis*B
kab = 3.94; Vp = 48.0Reaction: => Anakinra; Anakinrasc, Rate Law: kab*Anakinrasc/Vp
k1 = 0.2; placebo_on = 0.0; k2 = 0.0025Reaction: IL1b =>, Rate Law: piecewise((1-placebo_on)*k1*IL1b, time < 91, (1-placebo_on)*k2*IL1b)
k1 = 0.2; il1bH = 0.05; placebo_on = 0.0; il1b0 = 5.0; kplacebo = 0.00137; k2 = 0.0025Reaction: => IL1b, Rate Law: piecewise((1-placebo_on)*k1*il1bH, time < 91, (1-placebo_on)*k2*(il1b0+kplacebo*time))
CL = 432.0; Vp = 48.0Reaction: Anakinra =>, Rate Law: CL/Vp*Anakinra
IL1R = 0.02341920375; taus = 0.5; vs = 0.7; ks = 0.291008; kms = 0.021Reaction: => TigB, Rate Law: taus*ks*(1-vs*IL1R/(kms+IL1R))
Kglucose = 2.92E-4; lambda = 0.743; Ktr = 0.12Reaction: rbc5 = (Ktr*rbc4-Ktr*rbc5)-Kglucose*Glucose^lambda*rbc5, Rate Law: (Ktr*rbc4-Ktr*rbc5)-Kglucose*Glucose^lambda*rbc5
Kxi = 0.05Reaction: Proinsulin =>, Rate Law: 0.1*Kxi*Proinsulin
kab = 3.94Reaction: Anakinrasc =>, Rate Law: kab*Anakinrasc
tauf = 0.5Reaction: f =>, Rate Law: tauf*f

States:

NameDescription
Glucose[glucose]
f[insulin secretion]
a1c5[urn:miriam:efo:EFO%3A0004541]
a1c1[urn:miriam:efo:EFO%3A0004541]
a1c8[urn:miriam:efo:EFO%3A0004541]
a1c3[urn:miriam:efo:EFO%3A0004541]
rbc12[erythrocyte]
IL1b[Interleukin-1 beta]
rbc6[erythrocyte]
B[pancreatic beta cell]
a1c12[urn:miriam:efo:EFO%3A0004541]
rbc3[erythrocyte]
rbc1[erythrocyte]
Proinsulin[Insulin]
Anakinra[Interleukin-1 receptor antagonist protein; pharmaceutical]
a1c7[urn:miriam:efo:EFO%3A0004541]
a1c11[urn:miriam:efo:EFO%3A0004541]
rbc2[erythrocyte]
rbc11[erythrocyte]
a1c6[urn:miriam:efo:EFO%3A0004541]
rbc9[erythrocyte]
TigB[insulin secretion]
rbc5[erythrocyte]
Anakinrasc[Interleukin-1 receptor antagonist protein]
rbc7[erythrocyte]
a1c9[urn:miriam:efo:EFO%3A0004541]
Insulin[Insulin]
a1c10[urn:miriam:efo:EFO%3A0004541]
a1c4[urn:miriam:efo:EFO%3A0004541]
rbc4[erythrocyte]
rbc8[erythrocyte]
rbc10[erythrocyte]
a1c2[urn:miriam:efo:EFO%3A0004541]
hba1c[urn:miriam:efo:EFO%3A0004541]

Palmer2014 - Effect of IL-1β-Blocking therapies in T2DM - Healthy Condition: BIOMD0000000621v0.0.1

Palmer2014 - Effect of IL-1β-Blocking therapies in T2DM - Healthy Condition This is the model with healthy state initia…

Details

Recent clinical studies suggest sustained treatment effects of interleukin-1β (IL-1β)-blocking therapies in type 2 diabetes mellitus. The underlying mechanisms of these effects, however, remain underexplored. Using a quantitative systems pharmacology modeling approach, we combined ex vivo data of IL-1β effects on β-cell function and turnover with a disease progression model of the long-term interactions between insulin, glucose, and β-cell mass in type 2 diabetes mellitus. We then simulated treatment effects of the IL-1 receptor antagonist anakinra. The result was a substantial and partly sustained symptomatic improvement in β-cell function, and hence also in HbA1C, fasting plasma glucose, and proinsulin-insulin ratio, and a small increase in β-cell mass. We propose that improved β-cell function, rather than mass, is likely to explain the main IL-1β-blocking effects seen in current clinical data, but that improved β-cell mass might result in disease-modifying effects not clearly distinguishable until >1 year after treatment. link: http://identifiers.org/pubmed/24918743

Parameters:

NameDescription
taus = 0.5Reaction: TigB =>, Rate Law: taus*TigB
Kxg = 1.6E-5Reaction: Glucose =>, Rate Law: Kxg*Glucose
placebo_on = 0.0; kplacebo = 0.00137Reaction: => IL1b, Rate Law: placebo_on*kplacebo
il1b0 = 0.05; k1 = 0.2; il1bH = 0.05; placebo_on = 0.0; kplacebo = 0.00137; k2 = 0.0025Reaction: => IL1b, Rate Law: piecewise((1-placebo_on)*k1*il1bH, time < 91, (1-placebo_on)*k2*(il1b0+kplacebo*time))
Tgl = 0.025405Reaction: => Glucose, Rate Law: Tgl
Kglucose = 2.92E-4; lambda = 0.743; Kin = 1.05; Ktr = 0.12Reaction: rbc1 = (Kin-Ktr*rbc1)-Kglucose*Glucose^lambda*rbc1, Rate Law: (Kin-Ktr*rbc1)-Kglucose*Glucose^lambda*rbc1
IL1R = 3.743916136E-4; taus = 0.5; vs = 0.7; ks = 0.291008; kms = 0.021Reaction: => TigB, Rate Law: taus*ks*(1-vs*IL1R/(kms+IL1R))
Gh = 9.0; vh = 4.0Reaction: => Proinsulin; TigB, B, f, Glucose, Rate Law: f*(Glucose/Gh)^vh/(1+(Glucose/Gh)^vh)*TigB*B
kab = 3.94; Vp = 48.0Reaction: => Anakinra; Anakinrasc, Rate Law: kab*Anakinrasc/Vp
k1 = 0.2; placebo_on = 0.0; k2 = 0.0025Reaction: IL1b =>, Rate Law: piecewise((1-placebo_on)*k1*IL1b, time < 91, (1-placebo_on)*k2*IL1b)
CL = 432.0; Vp = 48.0Reaction: Anakinra =>, Rate Law: CL/Vp*Anakinra
Kxgi = 1.0E-4Reaction: Glucose => ; Insulin, Rate Law: Kxgi*Insulin*Glucose
Kglucose = 2.92E-4; lambda = 0.743; Ktr = 0.12Reaction: a1c5 = (Kglucose*Glucose^lambda*rbc5+Ktr*a1c4)-Ktr*a1c5, Rate Law: (Kglucose*Glucose^lambda*rbc5+Ktr*a1c4)-Ktr*a1c5
replication = 2.740001106E-4Reaction: => B, Rate Law: replication*B
Kxi = 0.05Reaction: Proinsulin =>, Rate Law: 0.1*Kxi*Proinsulin
vfg = 4.0; tauf = 0.5; kmf = 0.021; IL1R = 3.743916136E-4; kf = 0.00957754; kmfg = 9.0; xfg = 4.0; vf = 0.4Reaction: => f; Glucose, Rate Law: tauf*kf*(1+vfg*Glucose^xfg/(kmfg^xfg+Glucose^xfg))*(1+vf*IL1R/(kmf+IL1R))
kab = 3.94Reaction: Anakinrasc =>, Rate Law: kab*Anakinrasc
apoptosis = 2.740002397E-4Reaction: B =>, Rate Law: apoptosis*B
tauf = 0.5Reaction: f =>, Rate Law: tauf*f

States:

NameDescription
Glucose[glucose]
f[insulin secretion]
a1c5[urn:miriam:efo:EFO%3A0004541]
a1c1[urn:miriam:efo:EFO%3A0004541]
a1c8[urn:miriam:efo:EFO%3A0004541]
rbc12[erythrocyte]
a1c3[urn:miriam:efo:EFO%3A0004541]
IL1b[Interleukin-1 beta]
rbc6[erythrocyte]
B[pancreatic beta cell]
a1c12[urn:miriam:efo:EFO%3A0004541]
rbc3[erythrocyte]
rbc1[erythrocyte]
Proinsulin[Insulin]
Anakinra[Interleukin-1 receptor antagonist protein; pharmaceutical]
a1c7[urn:miriam:efo:EFO%3A0004541]
a1c11[urn:miriam:efo:EFO%3A0004541]
rbc2[erythrocyte]
rbc11[erythrocyte]
a1c6[urn:miriam:efo:EFO%3A0004541]
rbc9[erythrocyte]
rbc5[erythrocyte]
TigB[insulin secretion]
Anakinrasc[Interleukin-1 receptor antagonist protein]
a1c9[urn:miriam:efo:EFO%3A0004541]
Insulin[Insulin]
a1c2[urn:miriam:efo:EFO%3A0004541]
a1c10[urn:miriam:efo:EFO%3A0004541]
rbc10[erythrocyte]
a1c4[urn:miriam:efo:EFO%3A0004541]
rbc4[erythrocyte]
rbc7[erythrocyte]
rbc8[erythrocyte]
hba1c[urn:miriam:efo:EFO%3A0004541]

Palsson2013 - Fully-integrated immune response model (FIRM): BIOMD0000000608v0.0.1

Palsson2013 - Fully-integration immune response model (FIRM)FIRM (The Fully-integrated Immune Response Modeling) is a hy…

Details

BACKGROUND: The complexity and multiscale nature of the mammalian immune response provides an excellent test bed for the potential of mathematical modeling and simulation to facilitate mechanistic understanding. Historically, mathematical models of the immune response focused on subsets of the immune system and/or specific aspects of the response. Mathematical models have been developed for the humoral side of the immune response, or for the cellular side, or for cytokine kinetics, but rarely have they been proposed to encompass the overall system complexity. We propose here a framework for integration of subset models, based on a system biology approach. RESULTS: A dynamic simulator, the Fully-integrated Immune Response Model (FIRM), was built in a stepwise fashion by integrating published subset models and adding novel features. The approach used to build the model includes the formulation of the network of interacting species and the subsequent introduction of rate laws to describe each biological process. The resulting model represents a multi-organ structure, comprised of the target organ where the immune response takes place, circulating blood, lymphoid T, and lymphoid B tissue. The cell types accounted for include macrophages, a few T-cell lineages (cytotoxic, regulatory, helper 1, and helper 2), and B-cell activation to plasma cells. Four different cytokines were accounted for: IFN-γ, IL-4, IL-10 and IL-12. In addition, generic inflammatory signals are used to represent the kinetics of IL-1, IL-2, and TGF-β. Cell recruitment, differentiation, replication, apoptosis and migration are described as appropriate for the different cell types. The model is a hybrid structure containing information from several mammalian species. The structure of the network was built to be physiologically and biochemically consistent. Rate laws for all the cellular fate processes, growth factor production rates and half-lives, together with antibody production rates and half-lives, are provided. The results demonstrate how this framework can be used to integrate mathematical models of the immune response from several published sources and describe qualitative predictions of global immune system response arising from the integrated, hybrid model. In addition, we show how the model can be expanded to include novel biological findings. Case studies were carried out to simulate TB infection, tumor rejection, response to a blood borne pathogen and the consequences of accounting for regulatory T-cells. CONCLUSIONS: The final result of this work is a postulated and increasingly comprehensive representation of the mammalian immune system, based on physiological knowledge and susceptible to further experimental testing and validation. We believe that the integrated nature of FIRM has the potential to simulate a range of responses under a variety of conditions, from modeling of immune responses after tuberculosis (TB) infection to tumor formation in tissues. FIRM also has the flexibility to be expanded to include both complex and novel immunological response features as our knowledge of the immune system advances. link: http://identifiers.org/pubmed/24074340

Parameters:

NameDescription
q72e = 1.0E-4Reaction: x_3 => x_3 + x_35, Rate Law: q72e*x_3
volLymphT = 10.0; Rho21 = 100.0Reaction: x_11 => x_12, Rate Law: Rho21*volLymphT
w9 = 0.14; Mu9 = 0.04Reaction: x_3 => x_1 + x_3, Rate Law: Mu9*w9*x_3
q72b = 5.0E-5Reaction: x_8 => x_35 + x_8, Rate Law: q72b*x_8
Eta47 = 0.0024Reaction: x_19 => x_16 + x_43, Rate Law: Eta47*x_19
volLymphT = 10.0; Mu22 = 0.9; c22 = 3000.0Reaction: x_12 => x_12, Rate Law: x_12*Mu22/(c22+(x_12/volLymphT)^2)
Mu8 = 0.1; volLung = 1000.0; v14 = 0.0; k3 = 0.11; K3s = 50.0Reaction: x_3 + x_5 => x_3, Rate Law: Mu8*x_3*x_5*(x_5/x_3)^2/((x_5/x_3)^2+K3s^2)/x_3+x_3*x_5*volLung*v14/x_3/x_3+x_3*x_5*x_3/x_3*(x_5/x_3)^2*k3/((x_5/x_3)^2+K3s^2)/x_3
c12 = 500000.0; volLung = 1000.0; Delta12 = 0.4; cf12 = 150.0; fi12 = 2.333Reaction: x_1 + x_33 + x_39 + x_4 + x_5 => x_1 + x_2 + x_33 + x_39 + x_4 + x_5, Rate Law: Delta12*x_1*x_39/(x_39+fi12*x_33+cf12*volLung)*x_4/(c12*volLung+x_4+x_5)
Mu86 = 1.0; volLung = 1000.0; FACTOR = NaN; cf86 = 50.0; cF = 1000.0Reaction: x_29 + x_8 => x_29 + x_8, Rate Law: x_8*x_8/volLung*x_29*volLung*Mu86/(cF+x_29)/(cf86+FACTOR)
volBlood = 4500.0; Rho34 = 10.0Reaction: => x_16 + x_43, Rate Law: Rho34*volBlood
Delta11 = 0.36; volLung = 1000.0; cf11 = 100.0Reaction: x_2 + x_35 => x_1 + x_35, Rate Law: Delta11*x_2*x_35/(x_35+cf11*volLung)
Delta54 = 0.001; volLung = 1000.0Reaction: x_26 + x_29 => x_27 + x_29, Rate Law: x_26*x_29*Delta54/volLung
Delta43 = 2.4Reaction: x_20 + x_47 + x_48 => x_21 + x_47 + x_48, Rate Law: Delta43*x_20*x_47/(x_47+x_48+1E-5)
Alpha61 = 10.0Reaction: x_2 + x_29 => x_2 + x_30, Rate Law: Alpha61*x_2*x_29/(100000+x_29)
volLung = 1000.0; Mu15 = 0.02; c15 = 150000.0Reaction: x_4 => x_4 + x_6, Rate Law: volLung*x_4*Mu15/(c15*volLung+x_4)
Eta13 = 1.0Reaction: x_2 =>, Rate Law: Eta13*x_2
Delta36 = 2.4Reaction: x_16 + x_43 + x_44 => x_17 + x_44 + x_45, Rate Law: Delta36*x_16*x_44/(x_43+x_44+1E-5)
c25 = 100000.0; volLung = 1000.0; Mu25 = 0.4Reaction: x_2 + x_7 => x_2 + x_7, Rate Law: Mu25*x_7*x_2/(c25*volLung+x_2)
Gamma31 = 0.3333Reaction: x_9 => x_14, Rate Law: Gamma31*x_9
k3 = 0.11; K3s = 50.0Reaction: x_3 + x_5 => x_5, Rate Law: k3*x_3/((x_5/x_3)^2+K3s^2)*(x_5/x_3)^2
ci12 = 1000.0; Deltai12 = 0.009Reaction: x_1 + x_29 => x_2 + x_29, Rate Law: Deltai12*x_1*x_29/(ci12+x_29)
volLung = 1000.0; MuI = 9.0; cF = 1000.0; cI = 50.0; Gamma17 = 0.2Reaction: x_29 + x_6 => x_29, Rate Law: x_6/volLung*x_29*volLung*MuI*Gamma17/(cF+x_29)/(cI+x_29/(cF+x_29))
Gamma32 = 0.9Reaction: x_14 => x_15, Rate Law: Gamma32*x_14
Eta16 = 0.01Reaction: x_6 =>, Rate Law: Eta16*x_6
Mu8 = 0.1Reaction: x_3 + x_5 => x_3 + x_5, Rate Law: Mu8*x_3*x_5/x_3
volLung = 1000.0; c4 = 0.15; Alpha4 = 0.5Reaction: x_3 + x_5 + x_8 => x_3 + x_8, Rate Law: x_3*x_5*x_3/x_3*x_8*Alpha4/x_3/(x_8/x_3+c4*volLung)/x_3
fi27 = 4.1; Delta27 = 0.1; fii27 = 4.8; volLung = 1000.0; cf27 = 30.0Reaction: x_33 + x_35 + x_36 + x_38 + x_7 => x_33 + x_35 + x_36 + x_38 + x_8, Rate Law: x_36*x_38*Delta27/volLung*x_7/(x_36+fi27*x_33+fii27*x_35+cf27*volLung)
Alpha5 = 1.25E-7; volLung = 1000.0Reaction: x_2 + x_4 => x_2, Rate Law: x_2*x_4*Alpha5/volLung
Rho19 = 1000.0; volLymphT = 10.0Reaction: => x_11, Rate Law: Rho19*volLymphT
Delta38 = 2.4Reaction: x_18 + x_47 + x_48 => x_19 + x_48, Rate Law: Delta38*x_18*x_47/(x_47+x_48+1E-5)
q68a = 0.0029Reaction: x_7 => x_33 + x_7, Rate Law: q68a*x_7
Eta53 = 0.02Reaction: x_26 =>, Rate Law: Eta53*x_26
volLung = 1000.0; Rho21 = 100.0; MuI = 9.0; cF = 1000.0; cI = 50.0; Rho50 = 100.0Reaction: x_29 => x_29 + x_6, Rate Law: x_29*volLung*Rho21*MuI/(cF+x_29)/(cI+x_29/(cF+x_29))+x_29*volLung*Rho50*MuI/(cF+x_29)/(cI+x_29/(cF+x_29))
volLung = 1000.0; k7 = 1.0E-7Reaction: x_4 + x_6 => x_6, Rate Law: x_4*x_6*k7/volLung
Eta10 = 0.05Reaction: x_1 =>, Rate Law: Eta10*x_1
c24 = 15000.0; volLung = 1000.0; Gamma24 = 0.9Reaction: x_13 + x_2 => x_2 + x_7, Rate Law: Gamma24*x_13*x_2/(c24*volLung+x_2)
volLung = 1000.0; v14 = 0.0; k3 = 0.11; K3s = 50.0Reaction: x_3 + x_5 => x_3 + x_4 + x_5, Rate Law: volLung*x_5*v14/x_3+k3*x_3*1/((x_5/x_3)^2+K3s^2)*x_5/x_3*(x_5/x_3)^2
volLymphT = 10.0; Delta21 = 1.0E-4Reaction: x_10 + x_11 => x_10 + x_12, Rate Law: x_10*x_11*Delta21/volLymphT
Mui9 = 125000.0; volLung = 1000.0; MuI = 9.0; cF = 1000.0; cI = 50.0Reaction: x_29 => x_1 + x_29, Rate Law: x_29*volLung*Mui9*MuI/(cF+x_29)/(cI+x_29/(cF+x_29))
K93 = 100.0; Beta90 = 1000.0Reaction: x_52 => x_4 + x_51, Rate Law: Beta90*x_52/K93
q68b = 0.0218Reaction: x_9 => x_33 + x_9, Rate Law: q68b*x_9
Gamma40 = 0.9Reaction: x_18 + x_47 => x_20, Rate Law: Gamma40*x_18
q72d = 1.0E-4Reaction: x_7 => x_35 + x_7, Rate Law: q72d*x_7
volLung = 1000.0; Rho80 = 700.0; c80 = 5000.0; ci80 = 50.0Reaction: x_38 + x_4 + x_5 => x_38 + x_39 + x_4 + x_5, Rate Law: Rho80*volLung*x_38*x_4/(ci80*volLung+x_38)/(c80*volLung+x_4+x_5)+Rho80*volLung*x_38*x_5/(ci80*volLung+x_38)/(c80*volLung+x_4+x_5)
volLung = 1000.0; Rho9 = 5000.0Reaction: => x_1, Rate Law: Rho9*volLung
k2 = 0.4; c2 = 1000000.0; volLung = 1000.0Reaction: x_1 + x_4 => x_3, Rate Law: k2*x_1*x_4/(x_4+c2*volLung)
volLung = 1000.0; Beta90 = 1000.0Reaction: x_4 + x_51 => x_52, Rate Law: Beta90*x_4*x_51/volLung
Deltai27 = 0.001Reaction: x_2 + x_30 + x_7 => x_2 + x_30 + x_7 + x_8, Rate Law: Deltai27*x_2*x_30*x_7/(10000000+x_30)
v67 = 0.0; volLung = 1000.0Reaction: x_32 =>, Rate Law: v67*volLung
Eta28 = 0.3333Reaction: x_8 =>, Rate Law: Eta28*x_8
Mu9 = 0.04Reaction: x_2 => x_1 + x_2, Rate Law: Mu9*x_2
volLung = 1000.0; cf29 = 2.0; Delta29 = 0.05; fi29 = 0.12Reaction: x_33 + x_39 + x_7 => x_33 + x_39 + x_9, Rate Law: Delta29*x_7*x_33/(x_33+fi29*x_39+cf29*volLung)
volLung = 1000.0; v30 = 0.0Reaction: x_9 =>, Rate Law: v30*volLung
volLung = 1000.0; c17 = 10000.0; Gamma17 = 0.2Reaction: x_4 + x_6 => x_10 + x_4, Rate Law: Gamma17*x_6*x_4/(c17*volLung+x_4)
volLung = 1000.0; c52 = 15000.0; Gamma52 = 0.9Reaction: x_2 + x_25 => x_2 + x_26, Rate Law: Gamma52*x_25*x_2/(c52*volLung+x_2)
Eta33 = 0.3333Reaction: x_15 =>, Rate Law: Eta33*x_15
Gamma23 = 0.9Reaction: x_12 => x_13, Rate Law: Gamma23*x_12
volLung = 1000.0; Mu55 = 1.0; FACTOR = NaN; cF = 1000.0Reaction: x_27 + x_29 + x_8 => x_27 + x_29 + x_8, Rate Law: x_8*x_27/volLung*x_29*volLung*Mu55/(cF+x_29)/(cF+FACTOR)
ci72 = 0.05; volLung = 1000.0; c72 = 51.0; q72a = 0.006Reaction: x_2 + x_35 + x_39 => x_2 + x_35 + x_39, Rate Law: c72*q72a*x_2*1/(x_35+ci72*x_39+c72*volLung)
Eta26 = 0.3333Reaction: x_7 =>, Rate Law: Eta26*x_7
v41 = 0.0; volLymphB = 150.0Reaction: x_20 => x_18 + x_47, Rate Law: v41*volLymphB
volLung = 1000.0; cii80 = 100000.0; q80 = 0.02Reaction: x_2 + x_8 => x_2 + x_39 + x_8, Rate Law: volLung*x_2*x_8*q80/(cii80*volLung+x_2)

States:

NameDescription
x 6x_6
x 3x_3
x 33x_33
x 29x_29
x 22x_22
x 11x_11
x 17x_17
x 5x_5
x 15x_15
x 18x_18
x 32x_32
x 14x_14
x 16x_16
x 35x_35
x 1x_1
x 8x_8
x 21x_21
x 7x_7
x 4x_4
x 19x_19
x 9x_9
x 10x_10
x 12x_12
x 27x_27
x 20x_20
x 2x_2
x 26x_26
x 13x_13

Pandey2018-reversible transition between quiescence and proliferation: BIOMD0000000954v0.0.1

Cells switch between quiescence and proliferation states for maintaining tissue homeostasis and regeneration. At the res…

Details

Cells switch between quiescence and proliferation states for maintaining tissue homeostasis and regeneration. At the restriction point (R-point), cells become irreversibly committed to the completion of the cell cycle independent of mitogen. The mechanism involving hyper-phosphorylation of retinoblastoma (Rb) and activation of transcription factor E2F is linked to the R-point passage. However, stress stimuli trigger exit from the cell cycle back to the mitogen-sensitive quiescent state after Rb hyper-phosphorylation but only until APC/CCdh1 inactivation. In this study, we developed a mathematical model to investigate the reversible transition between quiescence and proliferation in mammalian cells with respect to mitogen and stress signals. The model integrates the current mechanistic knowledge and accounts for the recent experimental observations with cells exiting quiescence and proliferating cells. We show that Cyclin E:Cdk2 couples Rb-E2F and APC/CCdh1 bistable switches and temporally segregates the R-point and the G1/S transition. A redox-dependent mutual antagonism between APC/CCdh1 and its inhibitor Emi1 makes the inactivation of APC/CCdh1 bistable. We show that the levels of Cdk inhibitor (CKI) and mitogen control the reversible transition between quiescence and proliferation. Further, we propose that shifting of the mitogen-induced transcriptional program to G2-phase in proliferating cells might result in an intermediate Cdk2 activity at the mitotic exit and in the immediate inactivation of APC/CCdh1. Our study builds a coherent framework and generates hypotheses that can be further explored by experiments. link: http://identifiers.org/pubmed/29856829

Pandit2003_VentricularMyocytes: MODEL8685104549v0.0.1

This a model from the article: A mathematical model of the electrophysiological alterations in rat ventricular myocyte…

Details

Our mathematical model of the rat ventricular myocyte (Pandit et al., 2001) was utilized to explore the ionic mechanism(s) that underlie the altered electrophysiological characteristics associated with the short-term model of streptozotocin-induced, type-I diabetes. The simulations show that the observed reductions in the Ca(2+)-independent transient outward K(+) current (I(t)) and the steady-state outward K(+) current (I(ss)), along with slowed inactivation of the L-type Ca(2+) current (I(CaL)), can result in the prolongation of the action potential duration, a well-known experimental finding. In addition, the model demonstrates that the slowed reactivation kinetics of I(t) in diabetic myocytes can account for the more pronounced rate-dependent action potential duration prolongation in diabetes, and that a decrease in the electrogenic Na(+)-K(+) pump current (I(NaK)) results in a small depolarization in the resting membrane potential (V(rest)). This depolarization reduces the availability of the Na(+) channels (I(Na)), thereby resulting in a slower upstroke (dV/dt(max)) of the diabetic action potential. Additional simulations suggest that a reduction in the magnitude of I(CaL), in combination with impaired sarcoplasmic reticulum uptake can lead to a decreased sarcoplasmic reticulum Ca(2+) load. These factors contribute to characteristic abnormal Ca(2+) homeostasis (reduced peak systolic value and rate of decay) in myocytes from diabetic animals. In combination, these simulation results provide novel information and integrative insights concerning plausible ionic mechanisms for the observed changes in cardiac repolarization and excitation-contraction coupling in rat ventricular myocytes in the setting of streptozotocin-induced, type-I diabetes. link: http://identifiers.org/pubmed/12547767

PanRTK model for single cell line: MODEL1708210000v0.0.1

Hass2017-PanRTK model for single cell lineThe model structure comprises heterodimerization and receptor trafficking as d…

Details

Targeted therapies have shown significant patient benefit in about 5-10% of solid tumors that are addicted to a single oncogene. Here, we explore the idea of ligand addiction as a driver of tumor growth. High ligand levels in tumors have been shown to be associated with impaired patient survival, but targeted therapies have not yet shown great benefit in unselected patient populations. Using an approach of applying Bagged Decision Trees (BDT) to high-dimensional signaling features derived from a computational model, we can predict ligand dependent proliferation across a set of 58 cell lines. This mechanistic, multi-pathway model that features receptor heterodimerization, was trained on seven cancer cell lines and can predict signaling across two independent cell lines by adjusting only the receptor expression levels for each cell line. Interestingly, for patient samples the predicted tumor growth response correlates with high growth factor expression in the tumor microenvironment, which argues for a co-evolution of both factors in vivo. link: http://identifiers.org/pubmed/28944080

Panteleev2002_TFPImechanism_schmema3: BIOMD0000000359v0.0.1

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

Details

We have analyzed several mathematical models that describe inhibition of the factor VIIa-tissue factor complex (VIIa-TF) by tissue factor pathway inhibitor (TFPI). At the core of these models is a common mechanism of TFPI action suggesting that only the Xa-TFPI complex is the inhibitor of the extrinsic tenase activity. However, the model based on this hypothesis could not explain well all the available experimental data. Here, we show that a good quantitative description of all experimental data could be achieved in a model that contains two more assumptions. The first assumption is based on the hypothesis originally proposed by Baugh et al. [Baugh, R.J., Broze, G.J. Jr & Krishnaswamy, S. (1998) J. Biol. Chem. 273, 4378-4386], which suggests that TFPI could inhibit the enzyme-product complex Xa-VIIa-TF. The second assumption proposes an interaction between the X-VIIa-TF complex and the factor Xa-TFPI complex. Experiments to test these hypotheses are suggested. link: http://identifiers.org/pubmed/11985578

Parameters:

NameDescription
k1=6.0; k2=0.02Reaction: VIIa_TF_Xa + TFPI => VIIa_TF_Xa_TFPI, Rate Law: compartment*(k1*VIIa_TF_Xa*TFPI-k2*VIIa_TF_Xa_TFPI)
k2=0.02; k1=0.054Reaction: Xa + TFPI => Xa_TFPI, Rate Law: compartment*(k1*Xa*TFPI-k2*Xa_TFPI)
k1=420.0Reaction: VIIa_TF_X => VIIa_TF_Xa, Rate Law: compartment*k1*VIIa_TF_X
k1=0.44; k2=0.0Reaction: VIIa_TF + Xa_TFPI => Xa_TFPI_VIIa_TF, Rate Law: compartment*(k1*VIIa_TF*Xa_TFPI-k2*Xa_TFPI_VIIa_TF)
k2=770.0; k1=5.0Reaction: X + VIIa_TF => VIIa_TF_X, Rate Law: compartment*(k1*X*VIIa_TF-k2*VIIa_TF_X)
k2=5.0; k1=770.0Reaction: VIIa_TF_Xa => Xa + VIIa_TF, Rate Law: compartment*(k1*VIIa_TF_Xa-k2*Xa*VIIa_TF)
k1=0.0; k2=0.0Reaction: VIIa_TF_Xa_TFPI => Xa_TFPI_VIIa_TF, Rate Law: compartment*(k1*VIIa_TF_Xa_TFPI-k2*Xa_TFPI_VIIa_TF)
k1=20.0; k2=0.0Reaction: VIIa_TF_X + Xa_TFPI => X + VIIa_TF_Xa_TFPI, Rate Law: compartment*(k1*VIIa_TF_X*Xa_TFPI-k2*X*VIIa_TF_Xa_TFPI)

States:

NameDescription
Xa TFPI[Tissue factor pathway inhibitor; Coagulation factor X]
Xa TFPI VIIa TF[Tissue factor; Coagulation factor VII; Tissue factor pathway inhibitor; Coagulation factor X]
VIIa TF X[Coagulation factor X; Tissue factor; Coagulation factor VII]
X[Coagulation factor X]
TFPI[Tissue factor pathway inhibitor]
VIIa TF Xa TFPI[Tissue factor; Coagulation factor VII; Coagulation factor X; Tissue factor pathway inhibitor]
VIIa TF[Tissue factor; Coagulation factor VII]
Xa[Coagulation factor X]
VIIa TF Xa[Coagulation factor X; Tissue factor; Coagulation factor VII]

Panteleev2010 - Blood Coagulation: Full Model: BIOMD0000000740v0.0.1

Full and reduced mathematical model of blood coagulation focusing on fibrin formation and the response to varied TF and…

Details

Analysis of complex time-dependent biological networks is an important challenge in the current postgenomic era. We propose a middle-out approach for decomposition and analysis of complex time-dependent biological networks based on: 1), creation of a detailed mechanism-driven mathematical model of the network; 2), network response decomposition into several physiologically relevant subtasks; and 3), subsequent decomposition of the model, with the help of task-oriented necessity and sensitivity analysis into several modules that each control a single specific subtask, which is followed by further simplification employing temporal hierarchy reduction. The technique is tested and illustrated by studying blood coagulation. Five subtasks (threshold, triggering, control by blood flow velocity, spatial propagation, and localization), together with responsible modules, can be identified for the coagulation network. We show that the task of coagulation triggering is completely regulated by a two-step pathway containing a single positive feedback of factor V activation by thrombin. These theoretical predictions are experimentally confirmed by studies of fibrin generation in normal, factor V-, and factor VIII-deficient plasmas. The function of the factor V-dependent feedback is to minimize temporal and parametrical intervals of fibrin clot instability. We speculate that this pathway serves to lessen possibility of fibrin clot disruption by flow and subsequent thromboembolism. link: http://identifiers.org/pubmed/20441738

Parameters:

NameDescription
k_01 = 1.1; k03 = 0.4; k02 = 0.0014; k01 = 4.2Reaction: VII_TF = ((k01*VII*TF-k_01*VII_TF)-k02*VII_TF*IIa_F)-k03*VII_TF*Xa_F, Rate Law: ((k01*VII*TF-k_01*VII_TF)-k02*VII_TF*IIa_F)-k03*VII_TF*Xa_F
k15 = 54.0; K15 = 147.0; h14 = 0.35Reaction: VIIIa = k15*VIII*IIa_F/(K15+IIa_F)-h14*VIIIa, Rate Law: k15*VIII*IIa_F/(K15+IIa_F)-h14*VIIIa
h17 = 2.6E-5; h20 = 1.4E-4; k17 = 0.03; h18 = 6.0E-6; h16 = 1.9E-5; h19 = 0.0054Reaction: XIa = k17*Phospholipid*XI*IIa_F-(h16*AT_III+h17*alpha2_antiplasmin+h18*alpha1_antitrypsin+h19*ProteinC_Inhibitor+h20*C1_inhibitor)*XIa, Rate Law: k17*Phospholipid*XI*IIa_F-(h16*AT_III+h17*alpha2_antiplasmin+h18*alpha1_antitrypsin+h19*ProteinC_Inhibitor+h20*C1_inhibitor)*XIa
K26 = 470.0; n25 = 16000.0; K25 = 320.0Reaction: X_B = X*Phospholipid*n25/(K25*(1+X/K25+II/K26)), Rate Law: missing
K05 = 200.0; k04 = 15.0; K04 = 210.0; k05 = 5.8Reaction: IX = (-k04/K04)*IX*VIIa_TF_F-k05*IX*XIa/(K05+IX), Rate Law: (-k04/K04)*IX*VIIa_TF_F-k05*IX*XIa/(K05+IX)
k07 = 0.06; k08 = 6350.0; k06 = 435.0; K06 = 238.0; K07 = 230.0; K09 = 278.0Reaction: X = ((-k06/K06)*X*VIIa_TF_F-k07*IXa_B_F*X_B/(Phospholipid*K07))-k08*IXa_B_F*VIIIa_B_F*X_B/(Phospholipid^2*K09*k08), Rate Law: ((-k06/K06)*X*VIIa_TF_F-k07*IXa_B_F*X_B/(Phospholipid*K07))-k08*IXa_B_F*VIIIa_B_F*X_B/(Phospholipid^2*K09*k08)
h21 = 6.0E-6; h22 = 6.0E-6; h23 = 7.0E-7; k18 = 2.0E-5; h24 = 3.9E-4Reaction: APC = k18*PC*IIa_F-(h21*alpha2_macroglobulin+h22*alpha2_antiplasmin+h23*alpha1_antitrypsin+h24*ProteinC_Inhibitor)*APC, Rate Law: k18*PC*IIa_F-(h21*alpha2_macroglobulin+h22*alpha2_antiplasmin+h23*alpha1_antitrypsin+h24*ProteinC_Inhibitor)*APC
K20 = 2.57; n20 = 260.0Reaction: IXa_B_F = IXa*Phospholipid*n20/(K20+IXa), Rate Law: missing
k_01 = 1.1; k02 = 0.0014; k01 = 4.2Reaction: VII = (-(k01*VII*TF-k_01*VII_TF))-k02*VII*IIa_F, Rate Law: (-(k01*VII*TF-k_01*VII_TF))-k02*VII*IIa_F
k11 = 0.052; k_11 = 0.02; h02 = 6.0Reaction: TFPI = (-(k11*Xa_F*TFPI-k_11*Xa_TFPI))-h02*Xa_VIIa_TF*TFPI, Rate Law: (-(k11*Xa_F*TFPI-k_11*Xa_TFPI))-h02*Xa_VIIa_TF*TFPI
K21 = 1.5; K10 = 1655.0; K22 = 150.0; n21 = 750.0Reaction: VIIIa_B_F = VIIIa*Phospholipid*n21/((K21+VIIIa)*(1+X_B/(Phospholipid*K10)*(1+ProteinS_inhibitor/K22))), Rate Law: missing
k_01 = 1.1; h01 = 0.44; k03 = 0.4; k02 = 0.0014; k01 = 4.2; h02 = 6.0Reaction: VIIa_TF = (((k01*VIIa*TF-k_01*VIIa_TF_F)+k02*VII_TF*IIa_F+k03*VII_TF*Xa_F)-h01*VIIa_TF_F*Xa_TFPI)-h02*Xa_VIIa_TF*TFPI, Rate Law: (((k01*VIIa*TF-k_01*VIIa_TF_F)+k02*VII_TF*IIa_F+k03*VII_TF*Xa_F)-h01*VIIa_TF_F*Xa_TFPI)-h02*Xa_VIIa_TF*TFPI
k15 = 54.0; K15 = 147.0Reaction: VIII = (-k15)*VIII*IIa_F/(K15+IIa_F), Rate Law: (-k15)*VIII*IIa_F/(K15+IIa_F)
n27 = 2700.0; K27 = 2.9Reaction: Va_B = Va*Phospholipid*n27/(K27+Va), Rate Law: missing
h03 = 8.2E-6; h08 = 2.2E-5; h04 = 1.5E-4; h09 = 4.1E-4; h16 = 1.9E-5Reaction: AT_III = (-(h03*IXa+h04*Xa_F+h08*Xa_Va_b+h09*IIa_F+h16*XIa))*AT_III, Rate Law: (-(h03*IXa+h04*Xa_F+h08*Xa_Va_b+h09*IIa_F+h16*XIa))*AT_III
k_01 = 1.1; k01 = 4.2Reaction: TF = (-(k01*VIIa*TF-k_01*VIIa_TF_F))-(k01*VII*TF-k_01*VII_TF), Rate Law: (-(k01*VIIa*TF-k_01*VIIa_TF_F))-(k01*VII*TF-k_01*VII_TF)
K23 = 0.118; K24 = 200.0Reaction: Xa_Va_b = Xa*Va_B/(K23*(1+ProteinS_inhibitor/K24+Xa/K23)+Va_B), Rate Law: missing
k17 = 0.03Reaction: XI = (-k17)*Phospholipid*XI*IIa_F, Rate Law: (-k17)*Phospholipid*XI*IIa_F
K14 = 7200.0Reaction: IIa_F = IIa/(1+(fibrin+fibrinogen)/K14), Rate Law: missing
k11 = 0.052; h08 = 2.2E-5; h07 = 0.0012; k_11 = 0.02; K06 = 238.0; K07 = 230.0; h05 = 4.0E-5; k07 = 0.06; k08 = 6350.0; h04 = 1.5E-4; k06 = 435.0; h06 = 1.36E-5; K09 = 278.0Reaction: Xa = (((k06/K06*X*VIIa_TF_F+k07*IXa_B_F*X_B/(Phospholipid*K07)+k08*IXa_B_F*VIIIa_B_F*X_B/(Phospholipid^2*K09*k08))-(k11*Xa_F*TFPI-k_11*Xa_TFPI))-(h04*AT_III+h05*alpha2_macroglobulin+h06*alpha1_antitrypsin+h07*ProteinC_Inhibitor)*Xa_F)-h08*AT_III*Xa_Va_b, Rate Law: (((k06/K06*X*VIIa_TF_F+k07*IXa_B_F*X_B/(Phospholipid*K07)+k08*IXa_B_F*VIIIa_B_F*X_B/(Phospholipid^2*K09*k08))-(k11*Xa_F*TFPI-k_11*Xa_TFPI))-(h04*AT_III+h05*alpha2_macroglobulin+h06*alpha1_antitrypsin+h07*ProteinC_Inhibitor)*Xa_F)-h08*AT_III*Xa_Va_b
k11 = 0.052; h01 = 0.44; k_11 = 0.02Reaction: Xa_TFPI = (k11*Xa_F*TFPI-k_11*Xa_TFPI)-h01*VIIa_TF_F*Xa_TFPI, Rate Law: (k11*Xa_F*TFPI-k_11*Xa_TFPI)-h01*VIIa_TF_F*Xa_TFPI
k16 = 14.0; K16 = 71.7Reaction: V = (-k16)*V*IIa_F/(K16+IIa_F), Rate Law: (-k16)*V*IIa_F/(K16+IIa_F)
K06 = 238.0; K04 = 210.0Reaction: VIIa_TF_F = VIIa_TF/(1+IX/K04+X/K06), Rate Law: missing
k13 = 1.44; k12 = 45.0Reaction: II = (-k12)*Phospholipid*Xa_F*II-k13*Xa_Va_b*II_B/Phospholipid, Rate Law: (-k12)*Phospholipid*Xa_F*II-k13*Xa_Va_b*II_B/Phospholipid
k18 = 2.0E-5Reaction: PC = (-k18)*PC*IIa_F, Rate Law: (-k18)*PC*IIa_F
k14 = 5040.0; K14 = 7200.0Reaction: fibrin = k14/K14*fibrinogen*IIa_F, Rate Law: k14/K14*fibrinogen*IIa_F
k06 = 435.0; k_19 = 770.0; K06 = 238.0Reaction: Xa_VIIa_TF = k06/(K06*k_19)*X*VIIa_TF_F, Rate Law: missing
h03 = 8.2E-6; K05 = 200.0; k04 = 15.0; K04 = 210.0; k05 = 5.8Reaction: IXa = (k04/K04*IX*VIIa_TF_F+k05*IX*XIa/(K05+IX))-h03*Xa_TFPI*IXa, Rate Law: (k04/K04*IX*VIIa_TF_F+k05*IX*XIa/(K05+IX))-h03*Xa_TFPI*IXa
k16 = 14.0; K16 = 71.7; h15 = 7.7Reaction: Va = k16*V*IIa_F/(K16+IIa_F)-h15*APC*Va_B_F, Rate Law: k16*V*IIa_F/(K16+IIa_F)-h15*APC*Va_B_F
k13 = 1.44; h10 = 1.0E-4; h12 = 3.7E-4; h11 = 3.0E-6; h09 = 4.1E-4; k12 = 45.0; h13 = 6.3E-5Reaction: IIa = (k12*Phospholipid*Xa_F*II+k13*Xa_Va_b*II_B/Phospholipid)-(h09*AT_III+h10*alpha2_macroglobulin+h11*alpha1_antitrypsin+h12*ProteinC_Inhibitor+h13*heparin_cofactor2)*IIa_F, Rate Law: (k12*Phospholipid*Xa_F*II+k13*Xa_Va_b*II_B/Phospholipid)-(h09*AT_III+h10*alpha2_macroglobulin+h11*alpha1_antitrypsin+h12*ProteinC_Inhibitor+h13*heparin_cofactor2)*IIa_F

States:

NameDescription
fibrin[Fibrin]
VIII[Coagulation Factor VIII]
TFPI[TFPI Gene]
X B[Coagulation Factor X]
II B[Thrombin]
V[Coagulation Factor V]
Xa VIIa TF[Coagulation Factor VII; Coagulation Factor X; Tissue Factor; Coagulation Factor VII; Coagulation Factor X]
Xa[Coagulation Factor X]
Va B[Coagulation Factor V]
VIIIa B F[Coagulation Factor VIII]
PC[Protein C]
VII TF[Coagulation Factor VII; Tissue Factor]
TF[Tissue Factor]
XIa[121660]
X[Coagulation Factor X]
IIa F[Thrombin]
VIIIa[Coagulation Factor VIII]
AT III[Therapeutic Human Antithrombin-III]
Va[Coagulation Factor V]
IIa[Thrombin]
Xa TFPI[Coagulation Factor X; TFPI Gene; Coagulation Factor X]
VIIa[Coagulation Factor VII]
Xa Va b[Coagulation Factor V; Coagulation Factor X]
fibrinogen[Fibrinogen]
XI[121660]
APC[Protein C]
VIIa TF F[Tissue Factor; Coagulation Factor VII]
VIIa TF[Coagulation Factor VII; Tissue Factor]
IXa[Coagulation Factor IX]
Xa F[Coagulation Factor X]
Va B F[Coagulation Factor V]
VII[Coagulation Factor VII]
II[Thrombin]
IX[Coagulation Factor IX]
IXa B F[Coagulation Factor IX]

Pappalardo2016 - PI3K/AKT and MAPK Signaling Pathways in Melanoma Cancer: BIOMD0000000666v0.0.1

Pappalardo2016 - PI3K/AKT and MAPK Signaling Pathways in Melanoma CancerThis model is described in the article: [Comput…

Details

Malignant melanoma is an aggressive tumor of the skin and seems to be resistant to current therapeutic approaches. Melanocytic transformation is thought to occur by sequential accumulation of genetic and molecular alterations able to activate the Ras/Raf/MEK/ERK (MAPK) and/or the PI3K/AKT (AKT) signalling pathways. Specifically, mutations of B-RAF activate MAPK pathway resulting in cell cycle progression and apoptosis prevention. According to these findings, MAPK and AKT pathways may represent promising therapeutic targets for an otherwise devastating disease.Here we show a computational model able to simulate the main biochemical and metabolic interactions in the PI3K/AKT and MAPK pathways potentially involved in melanoma development. Overall, this computational approach may accelerate the drug discovery process and encourages the identification of novel pathway activators with consequent development of novel antioncogenic compounds to overcome tumor cell resistance to conventional therapeutic agents. The source code of the various versions of the model are available as S1 Archive. link: http://identifiers.org/pubmed/27015094

Parameters:

NameDescription
Kcat=8.8912; km=3496490.0Reaction: species_10 => species_11; species_26, Rate Law: compartment_0*Kcat*species_26*species_10/(km+species_10)
km=62464.6; Kcat=0.884096Reaction: species_7 => species_6; species_4, Rate Law: compartment_0*Kcat*species_4*species_7/(km+species_7)
Kcat=10.6737; km=184912.0Reaction: species_15 => species_14; species_0, Rate Law: compartment_0*Kcat*species_0*species_15/(km+species_15)
Kcat=3.19E13; km=3200.0Reaction: bRafMutated => bRafMutatedInactive; Dabrafenib, Rate Law: compartment_0*Kcat*Dabrafenib*bRafMutated/(km+bRafMutated)
Kcat=0.0213697; km=763523.0Reaction: species_2 => species_3; species_10, Rate Law: compartment_0*Kcat*species_10*species_2/(km+species_2)
Kcat=0.126329; km=1061.71Reaction: species_6 => species_7; species_27, Rate Law: compartment_0*Kcat*species_27*species_6/(km+species_6)
Kcat=2.83243; km=518753.0Reaction: species_8 => species_9; species_26, Rate Law: compartment_0*Kcat*species_26*species_8/(km+species_8)
v=100.0Reaction: probRafMutated => bRafMutated, Rate Law: compartment_0*v
km=896896.0; Kcat=1611.97Reaction: species_2 => species_3; species_12, Rate Law: compartment_0*Kcat*species_12*species_2/(km+species_2)
Kcat=185.759; km=4768350.0Reaction: species_9 => species_8; species_6, Rate Law: compartment_0*Kcat*species_6*species_9/(km+species_9)
k1=2.18503E-5; k2=0.121008Reaction: species_25 + species_1 => species_0, Rate Law: compartment_0*(k1*species_25*species_1-k2*species_0)
km=1432410.0; Kcat=1509.36Reaction: species_4 => species_5; species_28, Rate Law: compartment_0*Kcat*species_28*species_4/(km+species_4)
k1=1.92527E-5Reaction: Dabrafenib =>, Rate Law: compartment_0*k1*Dabrafenib
Kcat=32.344; km=35954.3Reaction: species_5 => species_4; species_2, Rate Law: compartment_0*Kcat*species_2*species_5/(km+species_5)
Kcat=9.85367; km=1007340.0Reaction: species_11 => species_10; species_8, Rate Law: compartment_0*Kcat*species_8*species_11/(km+species_11)
k1=2.5Reaction: species_19 => species_20, Rate Law: compartment_0*k1*species_19
k1=0.2Reaction: species_0 =>, Rate Law: compartment_0*k1*species_0
k1=0.005Reaction: species_12 => species_13, Rate Law: compartment_0*k1*species_12
km=6086070.0; Kcat=694.731Reaction: species_20 => species_19; species_0, Rate Law: compartment_0*Kcat*species_0*species_20/(km+species_20)
Kcat=15.1212; km=119355.0Reaction: species_6 => species_7; species_16, Rate Law: compartment_0*Kcat*species_16*species_6/(km+species_6)
Kcat=0.0566279; km=653951.0Reaction: species_17 => species_16; species_14, Rate Law: compartment_0*Kcat*species_14*species_17/(km+species_17)
k1=0.00125Reaction: species_1 =>, Rate Law: compartment_0*k1*species_1
Kcat=0.0771067; km=272056.0Reaction: species_15 => species_14; species_4, Rate Law: compartment_0*Kcat*species_4*species_15/(km+species_15)

States:

NameDescription
species 9[Dual specificity mitogen-activated protein kinase kinase 1; K04368; Protein kinase byr1]
species 1[Receptor Tyrosine Kinase; Protein sevenless]
species 20[Rap guanine nucleotide exchange factor 1; K06277]
species 4[K07829; Ras-related protein R-Ras]
species 16[AKT kinase; Putative serine/threonine-protein kinase-like protein CCR3]
PIP3Active[Phosphatidylinositol-3,4,5-trisphosphate]
PDK1Inactive[[Pyruvate dehydrogenase (acetyl-transferring)] kinase isozyme 1, mitochondrial; Probable [pyruvate dehydrogenase (acetyl-transferring)] kinase, mitochondrial; K12077]
IRS1Active[K16172; Insulin receptor substrate 1]
species 0[Receptor Tyrosine Kinase; Protein sevenless]
species 21[Ras-related protein Rap-1A; K04353]
species 8[Dual specificity mitogen-activated protein kinase kinase 1; Protein kinase byr1; K04368]
species 17[Putative serine/threonine-protein kinase-like protein CCR3; AKT kinase]
species 12[ribosomal protein S6 kinase alpha; Putative serine/threonine-protein kinase-like protein CCR3]
species 25[Growth Factor]
species 5[K07829; Ras-related protein R-Ras]
species 15[K00914; Phosphatidylinositol 3-kinase age-1]
S6K1Active[Ribosomal protein S6 kinase beta-1; Putative serine/threonine-protein kinase-like protein CCR3; K04688]
Dabrafenib[D10064; dabrafenib]
species 2[K03099; Son of sevenless homolog 1]
species 6[Putative serine/threonine-protein kinase-like protein CCR3; RAF proto-oncogene serine/threonine-protein kinase; K04366]
mTORC1Inactive[Serine/threonine-protein kinase mTOR; TORC1 complex]
species 19[Rap guanine nucleotide exchange factor 1; K06277]
species 10[Mitogen-activated protein kinase 1; K05111]
S6K1Inactive[Ribosomal protein S6 kinase beta-1; Putative serine/threonine-protein kinase-like protein CCR3; K04688]
species 11[Mitogen-activated protein kinase 1; K05111]
bRafMutatedInactive[Putative serine/threonine-protein kinase-like protein CCR3; K04365; Serine/threonine-protein kinase B-raf; BRAF Gene Mutation]
species 30proRTK
IRS1Inactive[Insulin receptor substrate 1; K16172]
mTORC1Active[Serine/threonine-protein kinase mTOR; TORC1 complex]
species 14[Phosphatidylinositol 3-kinase age-1; K00914]
species 22[K04353; Ras-related protein Rap-1A]
PDK1Active[K12077; [Pyruvate dehydrogenase (acetyl-transferring)] kinase isozyme 1, mitochondrial; Probable [pyruvate dehydrogenase (acetyl-transferring)] kinase, mitochondrial]
bRafMutated[K04365; Serine/threonine-protein kinase B-raf; Putative serine/threonine-protein kinase-like protein CCR3; BRAF Gene Mutation]
species 3[Son of sevenless homolog 1; K03099]
PIP3Inactive[Phosphatidylinositol-3,4,5-trisphosphate]
species 7[RAF proto-oncogene serine/threonine-protein kinase; K04366; Putative serine/threonine-protein kinase-like protein CCR3]
probRafMutatedprobRafMutated
species 13[Putative serine/threonine-protein kinase-like protein CCR3; ribosomal protein S6 kinase alpha]

Park2019 - Cetuximab resistance in colorectal cancer: MODEL1909300004v0.0.1

It's an experimental + mathematical paper explaining probable targets for Cetixumab resistance in colorectal cancer.

Details

Cetuximab (CTX), a monoclonal antibody against epidermal growth factor receptor, is being widely used for colorectal cancer (CRC) with wild-type (WT) KRAS. However, its responsiveness is still very limited and WT KRAS is not enough to indicate such responsiveness. Here, by analyzing the gene expression data of CRC patients treated with CTX monotherapy, we have identified DUSP4, ETV5, GNB5, NT5E, and PHLDA1 as potential targets to overcome CTX resistance. We found that knockdown of any of these five genes can increase CTX sensitivity in KRAS WT cells. Interestingly, we further found that GNB5 knockdown can increase CTX sensitivity even for KRAS mutant cells. We unraveled that GNB5 overexpression contributes to CTX resistance by modulating the Akt signaling pathway from experiments and mathematical simulation. Overall, these results indicate that GNB5 might be a promising target for combination therapy with CTX irrespective of KRAS mutation. link: http://identifiers.org/pubmed/30719834

Park2019 - IL7 receptor signaling in T cells: BIOMD0000000803v0.0.1

This model is an attempt to provide a mathematical description of IL-7 dependent T cell homeostasis at the molecular and…

Details

Interleukin-7 (IL7) plays a nonredundant role in T cell survival and homeostasis, which is illustrated in the severe T cell lymphopenia of IL7-deficient mice, or demonstrated in animals or humans that lack expression of either the IL7Rα or γ c chain, the two subunits that constitute the functional IL7 receptor. Remarkably, IL7 is not expressed by T cells themselves, but produced in limited amounts by radio-resistant stromal cells. Thus, T cells need to constantly compete for IL7 to survive. How T cells maintain homeostasis and further maximize the size of the peripheral T cell pool in face of such competition are important questions that have fascinated both immunologists and mathematicians for a long time. Exceptionally, IL7 downregulates expression of its own receptor, so that IL7-signaled T cells do not consume extracellular IL7, and thus, the remaining extracellular IL7 can be shared among unsignaled T cells. Such an altruistic behavior of the IL7Rα chain is quite unique among members of the γ c cytokine receptor family. However, the consequences of this altruistic signaling behavior at the molecular, single cell and population levels are less well understood and require further investigation. In this regard, mathematical modeling of how a limited resource can be shared, while maintaining the clonal diversity of the T cell pool, can help decipher the molecular or cellular mechanisms that regulate T cell homeostasis. Thus, the current review aims to provide a mathematical modeling perspective of IL7-dependent T cell homeostasis at the molecular, cellular and population levels, in the context of recent advances in our understanding of the IL7 biology. This article is categorized under: Models of Systems Properties and Processes > Organ, Tissue, and Physiological Models Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Mechanistic Models Analytical and Computational Methods > Computational Methods. link: http://identifiers.org/pubmed/31137085

Parameters:

NameDescription
k_f_4 = 1.66054E-5Reaction: IL15 + IL15Ru => IL15Rb, Rate Law: compartment*k_f_4*IL15*IL15Ru
k_f_3 = 1.66054E-4Reaction: IL7 + IL7Ru => IL7Rb, Rate Law: compartment*k_f_3*IL7*IL7Ru
k_f_2 = 1.66054E-4Reaction: IL15Rbeta + gamma_c => IL15Ru, Rate Law: compartment*k_f_2*IL15Rbeta*gamma_c
k_r_4 = 0.1Reaction: IL15Rb => IL15 + IL15Ru, Rate Law: compartment*k_r_4*IL15Rb
k_r_3 = 0.1Reaction: IL7Rb => IL7 + IL7Ru, Rate Law: compartment*k_r_3*IL7Rb
k_r_2 = 0.1Reaction: IL15Ru => IL15Rbeta + gamma_c, Rate Law: compartment*k_r_2*IL15Ru
k_r_1 = 0.1Reaction: IL7Ru => IL7Ra + gamma_c, Rate Law: compartment*k_r_1*IL7Ru
k_f_1 = 1.66054E-4Reaction: IL7Ra + gamma_c => IL7Ru, Rate Law: compartment*k_f_1*IL7Ra*gamma_c

States:

NameDescription
IL7Ra[Interleukin-7 Receptor Subunit Alpha]
IL15Ru[Interleukin-15 Receptor]
IL15Rbeta[Interleukin-2 Receptor Subunit Beta]
gamma c[PR:P31785]
IL15Rb[interleukin-15 receptor complex]
IL7Ru[159734]
IL15[Interleukin-15]
IL7[Interleukin-7]
IL7Rb[interleukin-7 receptor complex]

Parra_Guillen2013 - Mathematical model approach to describe tumour response in mice after vaccine administration_model1: BIOMD0000000914v0.0.1

Mathematical model approach to describe tumour response in mice after vaccine administration and its applicability to im…

Details

Immunotherapy is a growing therapeutic strategy in oncology based on the stimulation of innate and adaptive immune systems to induce the death of tumour cells. In this paper, we have developed a population semi-mechanistic model able to characterize the mechanisms implied in tumour growth dynamic after the administration of CyaA-E7, a vaccine able to target antigen to dendritic cells, thus triggering a potent immune response. The mathematical model developed presented the following main components: (1) tumour progression in the animals without treatment was described with a linear model, (2) vaccine effects were modelled assuming that vaccine triggers a non-instantaneous immune response inducing cell death. Delayed response was described with a series of two transit compartments, (3) a resistance effect decreasing vaccine efficiency was also incorporated through a regulator compartment dependent upon tumour size, and (4) a mixture model at the level of the elimination of the induced signal vaccine (k 2) to model tumour relapse after treatment, observed in a small percentage of animals (15.6%). The proposed model structure was successfully applied to describe antitumor effect of IL-12, suggesting its applicability to different immune-stimulatory therapies. In addition, a simulation exercise to evaluate in silico the impact on tumour size of possible combination therapies has been shown. This type of mathematical approaches may be helpful to maximize the information obtained from experiments in mice, reducing the number of animals and the cost of developing new antitumor immunotherapies. link: http://identifiers.org/pubmed/23605806

Parameters:

NameDescription
k1 = 0.0907Reaction: VAC =>, Rate Law: compartment*k1*VAC
gamma = 5.24; REG_50 = 3.18; k3 = 1.08Reaction: Ts => ; REG, SVAC, Rate Law: compartment*k3*REG_50^gamma/(REG_50^gamma+REG^gamma)*Ts*SVAC
k2_pop2 = 0.0907Reaction: SVAC =>, Rate Law: compartment*k2_pop2*SVAC
gamma = 5.24Reaction: => Ts, Rate Law: compartment*gamma
k4 = 0.039Reaction: => REG; Ts, Rate Law: compartment*k4*Ts

States:

NameDescription
VAC[Vaccine]
Ts[Tumor Mass]
SVAC[Signal; Vaccine; Signal]
TRANTRAN
REG[Regulator]

Parton2018 - A model of Atherosclerosis and atheroma formation.: MODEL1812100001v0.0.1

SBML and SBGN-ML models of atherosclerosis and atheroma formation. This model is described in the publication: New mod…

Details

Motivation Atherosclerosis is amongst the leading causes of death globally. However, it is challenging to study in vivo or in vitro and no detailed, openly-available computational models exist. Clinical studies hint that pharmaceutical therapy may be possible. Here we develop the first detailed, computational model of atherosclerosis and use it to develop multi-drug therapeutic hypotheses.

Results We assembled a network describing atheroma development from the literature. Maps and mathematical models were produced using the Systems Biology Graphical Notation (SBGN) and Systems Biology Markup Language (SBML), respectively. The model was constrained against clinical and laboratory data. We identified five drugs that together potentially reverse advanced atheroma formation.

Availability and Implementation The map is available in the supplementary information in SBGN-ML format. The model is available in the supplementary material and from BioModels, a repository of SBML models, containing CellDesigner markup.

Supplementary Information Available from Bioinformatics online. link: http://identifiers.org/doi/10.1093/bioinformatics/bty980

Pasek2006_VentricularCardioMyocytes: MODEL0406553884v0.0.1

This a model from the article: The functional role of cardiac T-tubules explored in a model of rat ventricular myocyte…

Details

The morphology of the cardiac transverse-axial tubular system (TATS) has been known for decades, but its function has received little attention. To explore the possible role of this system in the physiological modulation of electrical and contractile activity, we have developed a mathematical model of rat ventricular cardiomyocytes in which the TATS is described as a single compartment. The geometrical characteristics of the TATS, the biophysical characteristics of ion transporters and their distribution between surface and tubular membranes were based on available experimental data. Biophysically realistic values of mean access resistance to the tubular lumen and time constants for ion exchange with the bulk extracellular solution were included. The fraction of membrane in the TATS was set to 56%. The action potentials initiated in current-clamp mode are accompanied by transient K+ accumulation and transient Ca2+ depletion in the TATS lumen. The amplitude of these changes relative to external ion concentrations was studied at steady-state stimulation frequencies of 1-5 Hz. Ca2+ depletion increased from 7 to 13.1% with stimulation frequency, while K+ accumulation decreased from 4.1 to 2.7%. These ionic changes (particularly Ca2+ depletion) implicated significant decrease of intracellular Ca2+ load at frequencies natural for rat heart. link: http://identifiers.org/pubmed/16608703

Pasek2008_VentricularCardioMyocyte: MODEL0406793751v0.0.1

This a model from the article: A model of the guinea-pig ventricular cardiac myocyte incorporating a transverse-axial…

Details

A model of the guinea-pig cardiac ventricular myocyte has been developed that includes a representation of the transverse-axial tubular system (TATS), including heterogeneous distribution of ion flux pathways between the surface and tubular membranes. The model reproduces frequency-dependent changes of action potential shape and intracellular ion concentrations and can replicate experimental data showing ion diffusion between the tubular lumen and external solution in guinea-pig myocytes. The model is stable at rest and during activity and returns to rested state after perturbation. Theoretical analysis and model simulations show that, due to tight electrical coupling, tubular and surface membranes behave as a homogeneous whole during voltage and current clamp (maximum difference 0.9 mV at peak tubular INa of -38 nA). However, during action potentials, restricted diffusion and ionic currents in TATS cause depletion of tubular Ca2+ and accumulation of tubular K+ (up to -19.8% and +3.4%, respectively, of bulk extracellular values, at 6 Hz). These changes, in turn, decrease ion fluxes across the TATS membrane and decrease sarcoplasmic reticulum (SR) Ca2+ load. Thus, the TATS plays a potentially important role in modulating the function of guinea-pig ventricular myocyte in physiological conditions. link: http://identifiers.org/pubmed/17888503

Passos2010_DNAdamage_CellularSenescence: BIOMD0000000287v0.0.1

This is the model described in: **Feedback between p21 and reactive oxygen production is necessary for cell senescence.*…

Details

Cellular senescence–the permanent arrest of cycling in normally proliferating cells such as fibroblasts–contributes both to age-related loss of mammalian tissue homeostasis and acts as a tumour suppressor mechanism. The pathways leading to establishment of senescence are proving to be more complex than was previously envisaged. Combining in-silico interactome analysis and functional target gene inhibition, stochastic modelling and live cell microscopy, we show here that there exists a dynamic feedback loop that is triggered by a DNA damage response (DDR) and, which after a delay of several days, locks the cell into an actively maintained state of 'deep' cellular senescence. The essential feature of the loop is that long-term activation of the checkpoint gene CDKN1A (p21) induces mitochondrial dysfunction and production of reactive oxygen species (ROS) through serial signalling through GADD45-MAPK14(p38MAPK)-GRB2-TGFBR2-TGFbeta. These ROS in turn replenish short-lived DNA damage foci and maintain an ongoing DDR. We show that this loop is both necessary and sufficient for the stability of growth arrest during the establishment of the senescent phenotype. link: http://identifiers.org/pubmed/20160708

Parameters:

NameDescription
kdegp53 = 8.25E-4Reaction: Mdm2_p53 => Mdm2, Rate Law: kdegp53*Mdm2_p53
kdephosp38 = 0.1Reaction: p38_P => p38, Rate Law: kdephosp38*p38_P
krepair = 6.0E-5Reaction: damDNA => Sink, Rate Law: krepair*damDNA
kphosp38 = 0.008Reaction: p38 + GADD45 => p38_P + GADD45, Rate Law: kphosp38*p38*GADD45
krelMdm2p53 = 1.155E-6Reaction: Mdm2_p53 => p53 + Mdm2, Rate Law: krelMdm2p53*Mdm2_p53
kactATM = 2.0E-5Reaction: damDNA + ATMI => damDNA + ATMA, Rate Law: kactATM*damDNA*ATMI
kdegMdm2 = 4.33E-4Reaction: Mdm2 => Sink, Rate Law: kdegMdm2*Mdm2
kdam = 0.007Reaction: IR => IR + damDNA, Rate Law: kdam*IR
kdephosMdm2 = 0.5Reaction: Mdm2_P => Mdm2, Rate Law: kdephosMdm2*Mdm2_P
kdamROS = 1.0E-5Reaction: ROS => ROS + damDNA, Rate Law: kdamROS*ROS
kdephosp53 = 0.5Reaction: p53_P => p53, Rate Law: kdephosp53*p53_P
ksynp21mRNAp53P = 6.0E-6Reaction: p53_P => p53_P + p21_mRNA, Rate Law: ksynp21mRNAp53P*p53_P
ksynp21step3 = 4.0E-5Reaction: p21step2 => p21, Rate Law: ksynp21step3*p21step2
kdegp53mdm2ind = 8.25E-7Reaction: p53 => Sink, Rate Law: kdegp53mdm2ind*p53
ksynp21mRNAp53 = 6.0E-8Reaction: p53 => p53 + p21_mRNA, Rate Law: ksynp21mRNAp53*p53
kbinMdm2p53 = 0.001155Reaction: p53 + Mdm2 => Mdm2_p53, Rate Law: kbinMdm2p53*p53*Mdm2
kremROS = 3.83E-4Reaction: ROS => Sink, Rate Law: kremROS*ROS
kinactATM = 5.0E-4Reaction: ATMA => ATMI, Rate Law: kinactATM*ATMA
kGADD45 = 4.0E-6Reaction: p21 => p21 + GADD45, Rate Law: kGADD45*p21
kdamBasalROS = 1.0E-9Reaction: basalROS => basalROS + damDNA, Rate Law: kdamBasalROS*basalROS
kgenROSp38 = 4.5E-4; kp38ROS = 1.0Reaction: p38_P => p38_P + ROS, Rate Law: kgenROSp38*p38_P*kp38ROS
ksynp21step1 = 4.0E-4Reaction: p21_mRNA => p21_mRNA + p21step1, Rate Law: ksynp21step1*p21_mRNA
ksynp53 = 0.006Reaction: p53_mRNA => p53 + p53_mRNA, Rate Law: ksynp53*p53_mRNA
kphosMdm2 = 2.0Reaction: Mdm2 + ATMA => Mdm2_P + ATMA, Rate Law: kphosMdm2*Mdm2*ATMA
kdegGADD45 = 1.0E-5Reaction: GADD45 => Sink, Rate Law: kdegGADD45*GADD45
kdegMdm2mRNA = 1.0E-4Reaction: Mdm2_mRNA => Sink, Rate Law: kdegMdm2mRNA*Mdm2_mRNA
kdegATMMdm2 = 4.0E-4Reaction: Mdm2_P => Sink, Rate Law: kdegATMMdm2*Mdm2_P
ksynp53mRNA = 0.001Reaction: Source => p53_mRNA, Rate Law: ksynp53mRNA*Source
ksynMdm2 = 4.95E-4Reaction: Mdm2_mRNA => Mdm2_mRNA + Mdm2, Rate Law: ksynMdm2*Mdm2_mRNA
kphosp53 = 0.006Reaction: p53 + ATMA => p53_P + ATMA, Rate Law: kphosp53*p53*ATMA
ksynp21step2 = 4.0E-5Reaction: p21step1 => p21step2, Rate Law: ksynp21step2*p21step1
kdegp53mRNA = 1.0E-4Reaction: p53_mRNA => Sink, Rate Law: kdegp53mRNA*p53_mRNA
kdegp21 = 1.9E-4Reaction: p21 => Sink, Rate Law: kdegp21*p21
ksynMdm2mRNA = 1.0E-4Reaction: p53 => p53 + Mdm2_mRNA, Rate Law: ksynMdm2mRNA*p53
kdegp21mRNA = 2.4E-5Reaction: p21_mRNA => Sink, Rate Law: kdegp21mRNA*p21_mRNA

States:

NameDescription
Mdm2 P[E3 ubiquitin-protein ligase Mdm2]
p21 mRNA[Cyclin-dependent kinase inhibitor 1]
basalROS[reactive oxygen species]
GADD45[Growth arrest and DNA damage-inducible protein GADD45 alpha; Growth arrest and DNA damage-inducible protein GADD45 beta; Growth arrest and DNA damage-inducible protein GADD45 gamma]
p38 P[Mitogen-activated protein kinase 14]
p53[Cellular tumor antigen p53]
SourceSource
p53 P[Cellular tumor antigen p53]
IRIR
Mdm2[E3 ubiquitin-protein ligase Mdm2]
ROS[reactive oxygen species]
damDNA[deoxyribonucleic acid]
p53 mRNA[Cellular tumor antigen p53]
p38[Mitogen-activated protein kinase 14]
ATMA[Serine-protein kinase ATM]
p21step2[Cyclin-dependent kinase inhibitor 1]
Mdm2 p53[E3 ubiquitin-protein ligase Mdm2; Cellular tumor antigen p53]
p21[Cyclin-dependent kinase inhibitor 1]
ATMI[Serine-protein kinase ATM]
SinkSink
Mdm2 mRNA[E3 ubiquitin-protein ligase Mdm2]
p21step1[Cyclin-dependent kinase inhibitor 1]

Pastick2009 - Genome-scale metabolic network of Streptococcus thermophilus (iMP429): MODEL1507180063v0.0.1

Pastick2009 - Genome-scale metabolic network of Streptococcus thermophilus (iMP429)This model is described in the articl…

Details

In this report, we describe the amino acid metabolism and amino acid dependency of the dairy bacterium Streptococcus thermophilus LMG18311 and compare them with those of two other characterized lactic acid bacteria, Lactococcus lactis and Lactobacillus plantarum. Through the construction of a genome-scale metabolic model of S. thermophilus, the metabolic differences between the three bacteria were visualized by direct projection on a metabolic map. The comparative analysis revealed the minimal amino acid auxotrophy (only histidine and methionine or cysteine) of S. thermophilus LMG18311 and the broad variety of volatiles produced from amino acids compared to the other two bacteria. It also revealed the limited number of pyruvate branches, forcing this strain to use the homofermentative metabolism for growth optimization. In addition, some industrially relevant features could be identified in S. thermophilus, such as the unique pathway for acetaldehyde (yogurt flavor) production and the absence of a complete pentose phosphate pathway. link: http://identifiers.org/pubmed/19346354

Pathak2013 - MAPK activation in response to various abiotic stresses: BIOMD0000000491v0.0.1

Pathak2013 - MAPK activation in response to various abiotic stressesMAPK activation mechanism in response to various abi…

Details

Mitogen-Activated Protein Kinases (MAPKs) cascade plays an important role in regulating plant growth and development, generating cellular responses to the extracellular stimuli. MAPKs cascade mainly consist of three sub-families i.e. mitogen-activated protein kinase kinase kinase (MAPKKK), mitogen-activated protein kinase kinase (MAPKK) and mitogen activated protein kinase (MAPK), several cascades of which are activated by various abiotic and biotic stresses. In this work we have modeled the holistic molecular mechanisms essential to MAPKs activation in response to several abiotic and biotic stresses through a system biology approach and performed its simulation studies. As extent of abiotic and biotic stresses goes on increasing, the process of cell division, cell growth and cell differentiation slow down in time dependent manner. The models developed depict the combinatorial and multicomponent signaling triggered in response to several abiotic and biotic factors. These models can be used to predict behavior of cells in event of various stresses depending on their time and exposure through activation of complex signaling cascades. link: http://identifiers.org/pubmed/23847397

Parameters:

NameDescription
kass_re81 = 1.0 s^(-1); kdiss_re81 = 1.0 s^(-1)Reaction: s42 => s57, Rate Law: kass_re81*s42-kdiss_re81*s57
kass_re52 = 1.0 s^(-1); kdiss_re52 = 1.0 s^(-1)Reaction: s47 => s48, Rate Law: kass_re52*s47-kdiss_re52*s48
kdiss_re38 = 1.0 s^(-1); kass_re38 = 1.0 s^(-1)Reaction: s28 => s30, Rate Law: kass_re38*s28-kdiss_re38*s30
kdiss_re78 = 1.0 s^(-1); kass_re78 = 1.0 s^(-1)Reaction: s48 => s57, Rate Law: kass_re78*s48-kdiss_re78*s57
kass_re31 = 1.0 s^(-1); kdiss_re31 = 1.0 s^(-1)Reaction: s18 => s26, Rate Law: kass_re31*s18-kdiss_re31*s26
kdiss_re55 = 1.0 s^(-1); kass_re55 = 1.0 s^(-1)Reaction: s29 => s37, Rate Law: kass_re55*s29-kdiss_re55*s37
kdiss_re64 = 1.0 s^(-1); kass_re64 = 1.0 s^(-1)Reaction: s32 => s45, Rate Law: kass_re64*s32-kdiss_re64*s45
kass_re30 = 1.0 s^(-1); kdiss_re30 = 1.0 s^(-1)Reaction: s18 => s25, Rate Law: kass_re30*s18-kdiss_re30*s25
kass_re35 = 1.0 s^(-1); kdiss_re35 = 1.0 s^(-1)Reaction: s15 => s20, Rate Law: kass_re35*s15-kdiss_re35*s20
kass_re68 = 1.0 s^(-1); kdiss_re68 = 1.0 s^(-1)Reaction: s28 => s51, Rate Law: kass_re68*s28-kdiss_re68*s51
kass_re48 = 1.0 s^(-1); kdiss_re48 = 1.0 s^(-1)Reaction: s39 => s40, Rate Law: kass_re48*s39-kdiss_re48*s40
kass_re23 = 1.0 s^(-1); kdiss_re23 = 1.0 s^(-1)Reaction: s14 => s17, Rate Law: kass_re23*s14-kdiss_re23*s17
kass_re25 = 1.0 s^(-1); kdiss_re25 = 1.0 s^(-1)Reaction: s18 => s20, Rate Law: kass_re25*s18-kdiss_re25*s20
kdiss_re47 = 1.0 s^(-1); kass_re47 = 1.0 s^(-1)Reaction: s37 => s38, Rate Law: kass_re47*s37-kdiss_re47*s38
kass_re58 = 1.0 s^(-1); kdiss_re58 = 1.0 s^(-1)Reaction: s30 => s41, Rate Law: kass_re58*s30-kdiss_re58*s41
kass_re49 = 1.0 s^(-1); kdiss_re49 = 1.0 s^(-1)Reaction: s41 => s42, Rate Law: kass_re49*s41-kdiss_re49*s42
kass_re40 = 1.0 s^(-1); kdiss_re40 = 1.0 s^(-1)Reaction: s28 => s32, Rate Law: kass_re40*s28-kdiss_re40*s32
kdiss_re57 = 1.0 s^(-1); kass_re57 = 1.0 s^(-1)Reaction: s30 => s35, Rate Law: kass_re57*s30-kdiss_re57*s35
kdiss_re67 = 1.0 s^(-1); kass_re67 = 1.0 s^(-1)Reaction: s28 => s49, Rate Law: kass_re67*s28-kdiss_re67*s49
kdiss_re69 = 1.0 s^(-1); kass_re69 = 1.0 s^(-1)Reaction: s28 => s53, Rate Law: kass_re69*s28-kdiss_re69*s53
kass_re84 = 1.0 s^(-1); kdiss_re84 = 1.0 s^(-1)Reaction: s36 => s57, Rate Law: kass_re84*s36-kdiss_re84*s57
kass_re62 = 1.0 s^(-1); kdiss_re62 = 1.0 s^(-1)Reaction: s31 => s39, Rate Law: kass_re62*s31-kdiss_re62*s39
kass_re44 = 1.0 s^(-1); kdiss_re44 = 1.0 s^(-1)Reaction: s26 => s30, Rate Law: kass_re44*s26-kdiss_re44*s30
kdiss_re76 = 1.0 s^(-1); kass_re76 = 1.0 s^(-1)Reaction: s50 => s57, Rate Law: kass_re76*s50-kdiss_re76*s57
kass_re15 = 1.0 s^(-1); kdiss_re15 = 1.0 s^(-1)Reaction: s9 => s13, Rate Law: kass_re15*s9-kdiss_re15*s13
kdiss_re29 = 1.0 s^(-1); kass_re29 = 1.0 s^(-1)Reaction: s18 => s24, Rate Law: kass_re29*s18-kdiss_re29*s24
kdiss_re79 = 1.0 s^(-1); kass_re79 = 1.0 s^(-1)Reaction: s30 => s43, Rate Law: kass_re79*s30-kdiss_re79*s43
kdiss_re60 = 1.0 s^(-1); kass_re60 = 1.0 s^(-1)Reaction: s31 => s33, Rate Law: kass_re60*s31-kdiss_re60*s33
kass_re46 = 1.0 s^(-1); kdiss_re46 = 1.0 s^(-1)Reaction: s35 => s36, Rate Law: kass_re46*s35-kdiss_re46*s36
kdiss_re83 = 1.0 s^(-1); kass_re83 = 1.0 s^(-1)Reaction: s38 => s57, Rate Law: kass_re83*s38-kdiss_re83*s57
kass_re36 = 1.0 s^(-1); kdiss_re36 = 1.0 s^(-1)Reaction: s16 => s26, Rate Law: kass_re36*s16-kdiss_re36*s26
kdiss_re24 = 1.0 s^(-1); kass_re24 = 1.0 s^(-1)Reaction: s18 => s19, Rate Law: kass_re24*s18-kdiss_re24*s19
kdiss_re32 = 1.0 s^(-1); kass_re32 = 1.0 s^(-1)Reaction: s27 => s28, Rate Law: kass_re32*s27-kdiss_re32*s28
kdiss_re28 = 1.0 s^(-1); kass_re28 = 1.0 s^(-1)Reaction: s18 => s23, Rate Law: kass_re28*s18-kdiss_re28*s23
kass_re61 = 1.0 s^(-1); kdiss_re61 = 1.0 s^(-1)Reaction: s31 => s45, Rate Law: kass_re61*s31-kdiss_re61*s45
kdiss_re65 = 1.0 s^(-1); kass_re65 = 1.0 s^(-1)Reaction: s32 => s35, Rate Law: kass_re65*s32-kdiss_re65*s35
kdiss_re19 = 1.0 s^(-1); kass_re19 = 1.0 s^(-1)Reaction: s14 => s16, Rate Law: kass_re19*s14-kdiss_re19*s16
kass_re71 = 1.0 s^(-1); kdiss_re71 = 1.0 s^(-1)Reaction: s28 => s55, Rate Law: kass_re71*s28-kdiss_re71*s55
kass_re85 = 1.0 s^(-1); kdiss_re85 = 1.0 s^(-1)Reaction: s34 => s57, Rate Law: kass_re85*s34-kdiss_re85*s57
kass_re66 = 1.0 s^(-1); kdiss_re66 = 1.0 s^(-1)Reaction: s28 => s56, Rate Law: kass_re66*s28-kdiss_re66*s56
kass_re17 = 1.0 s^(-1); kdiss_re17 = 1.0 s^(-1)Reaction: s14 => s15, Rate Law: kass_re17*s14-kdiss_re17*s15
kdiss_re22 = 1.0 s^(-1); kass_re22 = 1.0 s^(-1)Reaction: s17 => s18, Rate Law: kass_re22*s17-kdiss_re22*s18
kass_re26 = 1.0 s^(-1); kdiss_re26 = 1.0 s^(-1)Reaction: s18 => s21, Rate Law: kass_re26*s18-kdiss_re26*s21
kass_re50 = 1.0 s^(-1); kdiss_re50 = 1.0 s^(-1)Reaction: s43 => s44, Rate Law: kass_re50*s43-kdiss_re50*s44
kass_re39 = 1.0 s^(-1); kdiss_re39 = 1.0 s^(-1)Reaction: s28 => s31, Rate Law: kass_re39*s28-kdiss_re39*s31
kass_re54 = 1.0 s^(-1); kdiss_re54 = 1.0 s^(-1)Reaction: s51 => s52, Rate Law: kass_re54*s51-kdiss_re54*s52
kass_re11 = 1.0 s^(-1); kdiss_re11 = 1.0 s^(-1)Reaction: s5 => s7, Rate Law: kass_re11*s5-kdiss_re11*s7
kass_re86 = 1.0 s^(-1); kdiss_re86 = 1.0 s^(-1)Reaction: s46 => s57, Rate Law: kass_re86*s46-kdiss_re86*s57
kdiss_re45 = 1.0 s^(-1); kass_re45 = 1.0 s^(-1)Reaction: s33 => s34, Rate Law: kass_re45*s33-kdiss_re45*s34
kdiss_re33 = 1.0 s^(-1); kass_re33 = 1.0 s^(-1)Reaction: s18 => s27, Rate Law: kass_re33*s18-kdiss_re33*s27
kass_re70 = 1.0 s^(-1); kdiss_re70 = 1.0 s^(-1)Reaction: s28 => s54, Rate Law: kass_re70*s28-kdiss_re70*s54
kass_re59 = 1.0 s^(-1); kdiss_re59 = 1.0 s^(-1)Reaction: s30 => s47, Rate Law: kass_re59*s30-kdiss_re59*s47
kdiss_re21 = 1.0 s^(-1); kass_re21 = 1.0 s^(-1)Reaction: s12 => s16, Rate Law: kass_re21*s12-kdiss_re21*s16
kdiss_re34 = 1.0 s^(-1); kass_re34 = 1.0 s^(-1)Reaction: s15 => s19, Rate Law: kass_re34*s15-kdiss_re34*s19
kdiss_re2 = 1.0 s^(-1); kass_re2 = 1.0 s^(-1)Reaction: s2 => s7, Rate Law: kass_re2*s2-kdiss_re2*s7
kass_re43 = 1.0 s^(-1); kdiss_re43 = 1.0 s^(-1)Reaction: s20 => s32, Rate Law: kass_re43*s20-kdiss_re43*s32
kass_re27 = 1.0 s^(-1); kdiss_re27 = 1.0 s^(-1)Reaction: s18 => s22, Rate Law: kass_re27*s18-kdiss_re27*s22
kdiss_re1 = 1.0 s^(-1); kass_re1 = 1.0 s^(-1)Reaction: s1 => s7, Rate Law: kass_re1*s1-kdiss_re1*s7
kdiss_re42 = 1.0 s^(-1); kass_re42 = 1.0 s^(-1)Reaction: s20 => s31, Rate Law: kass_re42*s20-kdiss_re42*s31
kass_re53 = 1.0 s^(-1); kdiss_re53 = 1.0 s^(-1)Reaction: s49 => s50, Rate Law: kass_re53*s49-kdiss_re53*s50
kass_re20 = 1.0 s^(-1); kdiss_re20 = 1.0 s^(-1)Reaction: s11 => s16, Rate Law: kass_re20*s11-kdiss_re20*s16
kass_re37 = 1.0 s^(-1); kdiss_re37 = 1.0 s^(-1)Reaction: s28 => s29, Rate Law: kass_re37*s28-kdiss_re37*s29
kdiss_re10 = 1.0 s^(-1); kass_re10 = 1.0 s^(-1)Reaction: s4 => s7, Rate Law: kass_re10*s4-kdiss_re10*s7
kdiss_re18 = 1.0 s^(-1); kass_re18 = 1.0 s^(-1)Reaction: s7 => s15, Rate Law: kass_re18*s7-kdiss_re18*s15
kdiss_re63 = 1.0 s^(-1); kass_re63 = 1.0 s^(-1)Reaction: s32 => s47, Rate Law: kass_re63*s32-kdiss_re63*s47
kdiss_re51 = 1.0 s^(-1); kass_re51 = 1.0 s^(-1)Reaction: s45 => s46, Rate Law: kass_re51*s45-kdiss_re51*s46
kdiss_re82 = 1.0 s^(-1); kass_re82 = 1.0 s^(-1)Reaction: s44 => s57, Rate Law: kass_re82*s44-kdiss_re82*s57
kass_re56 = 1.0 s^(-1); kdiss_re56 = 1.0 s^(-1)Reaction: s29 => s33, Rate Law: kass_re56*s29-kdiss_re56*s33
kass_re72 = 1.0 s^(-1); kdiss_re72 = 1.0 s^(-1)Reaction: s40 => s57, Rate Law: kass_re72*s40-kdiss_re72*s57

States:

NameDescription
s5[cellular response to metal ion]
s14[Mitogen-activated protein kinase kinase kinase 5]
s18[Mitogen-activated protein kinase kinase 1]
s37[Probable WRKY transcription factor 8]
s40[Probable WRKY transcription factor 25]
s20[Mitogen-activated protein kinase kinase 2]
s35[Probable WRKY transcription factor 12]
s44[Probable WRKY transcription factor 29]
s57[cellular response to stress]
s43[Probable WRKY transcription factor 29]
s19[Mitogen-activated protein kinase kinase 1]
s31[Mitogen-activated protein kinase 4]
s36[Probable WRKY transcription factor 12]
s34[WRKY transcription factor 1]
s50[ATMYB2At2g47190MYB transcription factorMYB transcription factor (Atmyb2)MYB transcription factor Atmyb2Myb domain protein 2]
s38[Probable WRKY transcription factor 8]
s47[Probable WRKY transcription factor 28]
s32[Mitogen-activated protein kinase 6]
s46[Probable WRKY transcription factor 33]
s15[Mitogen-activated protein kinase kinase kinase 1]
s51[Transcription repressor MYB4]
s45[Probable WRKY transcription factor 33]
s1[decreased temperature]
s48[Probable WRKY transcription factor 28]
s17[Mitogen-activated protein kinase kinase 1]
s41[WRKY transcription factor 22]
s25[Dual specificity mitogen-activated protein kinase kinase 7]
s13[Mitogen-activated protein kinase kinase kinase 5]
s2[sodium chloride]
s49[ATMYB2At2g47190MYB transcription factorMYB transcription factor (Atmyb2)MYB transcription factor Atmyb2Myb domain protein 2]
s33[WRKY transcription factor 1]
s16[Serine/threonine-protein kinase CTR1]
s4[hydrogen peroxide]
s30[Mitogen-activated protein kinase 3]
s26[Dual specificity mitogen-activated protein kinase kinase 1]
s42[WRKY transcription factor 22]
s28[Mitogen-activated protein kinase 3]
s39[Probable WRKY transcription factor 25]
s29[Mitogen-activated protein kinase]
s27[Mitogen-activated protein kinase 3]

Pathak2013 - MAPK activation in response to various biotic stresses: BIOMD0000000492v0.0.1

Pathak2013 - MAPK activation in response to various biotic stressesMAPK activation mechanism in response to various biot…

Details

Mitogen-Activated Protein Kinases (MAPKs) cascade plays an important role in regulating plant growth and development, generating cellular responses to the extracellular stimuli. MAPKs cascade mainly consist of three sub-families i.e. mitogen-activated protein kinase kinase kinase (MAPKKK), mitogen-activated protein kinase kinase (MAPKK) and mitogen activated protein kinase (MAPK), several cascades of which are activated by various abiotic and biotic stresses. In this work we have modeled the holistic molecular mechanisms essential to MAPKs activation in response to several abiotic and biotic stresses through a system biology approach and performed its simulation studies. As extent of abiotic and biotic stresses goes on increasing, the process of cell division, cell growth and cell differentiation slow down in time dependent manner. The models developed depict the combinatorial and multicomponent signaling triggered in response to several abiotic and biotic factors. These models can be used to predict behavior of cells in event of various stresses depending on their time and exposure through activation of complex signaling cascades. link: http://identifiers.org/pubmed/23847397

Parameters:

NameDescription
kass_re81 = 1.0 s^(-1); kdiss_re81 = 1.0 s^(-1)Reaction: s37 => s52, Rate Law: kass_re81*s37-kdiss_re81*s52
kdiss_re38 = 1.0 s^(-1); kass_re38 = 1.0 s^(-1)Reaction: s16 => s22, Rate Law: kass_re38*s16-kdiss_re38*s22
kass_re31 = 1.0 s^(-1); kdiss_re31 = 1.0 s^(-1)Reaction: s15 => s20, Rate Law: kass_re31*s15-kdiss_re31*s20
kdiss_re55 = 1.0 s^(-1); kass_re55 = 1.0 s^(-1)Reaction: s22 => s28, Rate Law: kass_re55*s22-kdiss_re55*s28
kass_re5 = 1.0 s^(-1); kdiss_re5 = 1.0 s^(-1)Reaction: s2 => s5, Rate Law: kass_re5*s2-kdiss_re5*s5
kass_re30 = 1.0 s^(-1); kdiss_re30 = 1.0 s^(-1)Reaction: s20 => s21, Rate Law: kass_re30*s20-kdiss_re30*s21
kass_re35 = 1.0 s^(-1); kdiss_re35 = 1.0 s^(-1)Reaction: s21 => s25, Rate Law: kass_re35*s21-kdiss_re35*s25
kass_re68 = 1.0 s^(-1); kdiss_re68 = 1.0 s^(-1)Reaction: s25 => s36, Rate Law: kass_re68*s25-kdiss_re68*s36
kass_re14 = 1.0 s^(-1); kdiss_re14 = 1.0 s^(-1)Reaction: s8 => s11, Rate Law: kass_re14*s8-kdiss_re14*s11
kass_re23 = 1.0 s^(-1); kdiss_re23 = 1.0 s^(-1)Reaction: s15 => s17, Rate Law: kass_re23*s15-kdiss_re23*s17
kass_re48 = 1.0 s^(-1); kdiss_re48 = 1.0 s^(-1)Reaction: s34 => s35, Rate Law: kass_re48*s34-kdiss_re48*s35
kdiss_re13 = 1.0 s^(-1); kass_re13 = 1.0 s^(-1)Reaction: s8 => s10, Rate Law: kass_re13*s8-kdiss_re13*s10
kass_re25 = 1.0 s^(-1); kdiss_re25 = 1.0 s^(-1)Reaction: s15 => s19, Rate Law: kass_re25*s15-kdiss_re25*s19
kdiss_re47 = 1.0 s^(-1); kass_re47 = 1.0 s^(-1)Reaction: s32 => s33, Rate Law: kass_re47*s32-kdiss_re47*s33
kass_re49 = 1.0 s^(-1); kdiss_re49 = 1.0 s^(-1)Reaction: s36 => s37, Rate Law: kass_re49*s36-kdiss_re49*s37
kass_re40 = 1.0 s^(-1); kdiss_re40 = 1.0 s^(-1)Reaction: s17 => s23, Rate Law: kass_re40*s17-kdiss_re40*s23
kdiss_re69 = 1.0 s^(-1); kass_re69 = 1.0 s^(-1)Reaction: s21 => s30, Rate Law: kass_re69*s21-kdiss_re69*s30
kdiss_re41 = 1.0 s^(-1); kass_re41 = 1.0 s^(-1)Reaction: s18 => s23, Rate Law: kass_re41*s18-kdiss_re41*s23
kass_re62 = 1.0 s^(-1); kdiss_re62 = 1.0 s^(-1)Reaction: s25 => s46, Rate Law: kass_re62*s25-kdiss_re62*s46
kass_re12 = 1.0 s^(-1); kdiss_re12 = 1.0 s^(-1)Reaction: s8 => s9, Rate Law: kass_re12*s8-kdiss_re12*s9
kass_re44 = 1.0 s^(-1); kdiss_re44 = 1.0 s^(-1)Reaction: s18 => s25, Rate Law: kass_re44*s18-kdiss_re44*s25
kdiss_re76 = 1.0 s^(-1); kass_re76 = 1.0 s^(-1)Reaction: s31 => s52, Rate Law: kass_re76*s31-kdiss_re76*s52
kass_re15 = 1.0 s^(-1); kdiss_re15 = 1.0 s^(-1)Reaction: s8 => s12, Rate Law: kass_re15*s8-kdiss_re15*s12
kdiss_re29 = 1.0 s^(-1); kass_re29 = 1.0 s^(-1)Reaction: s11 => s19, Rate Law: kass_re29*s11-kdiss_re29*s19
kass_re6 = 1.0 s^(-1); kdiss_re6 = 1.0 s^(-1)Reaction: s2 => s6, Rate Law: kass_re6*s2-kdiss_re6*s6
kass_re74 = 1.0 s^(-1); kdiss_re74 = 1.0 s^(-1)Reaction: s24 => s34, Rate Law: kass_re74*s24-kdiss_re74*s34
kass_re16 = 1.0 s^(-1); kdiss_re16 = 1.0 s^(-1)Reaction: s6 => s9, Rate Law: kass_re16*s6-kdiss_re16*s9
kdiss_re88 = 1.0 s^(-1); kass_re88 = 1.0 s^(-1)Reaction: s33 => s52, Rate Law: kass_re88*s33-kdiss_re88*s52
kass_re36 = 1.0 s^(-1); kdiss_re36 = 1.0 s^(-1)Reaction: s21 => s26, Rate Law: kass_re36*s21-kdiss_re36*s26
kdiss_re24 = 1.0 s^(-1); kass_re24 = 1.0 s^(-1)Reaction: s15 => s18, Rate Law: kass_re24*s15-kdiss_re24*s18
kdiss_re32 = 1.0 s^(-1); kass_re32 = 1.0 s^(-1)Reaction: s21 => s22, Rate Law: kass_re32*s21-kdiss_re32*s22
kdiss_re28 = 1.0 s^(-1); kass_re28 = 1.0 s^(-1)Reaction: s9 => s18, Rate Law: kass_re28*s9-kdiss_re28*s18
kdiss_re19 = 1.0 s^(-1); kass_re19 = 1.0 s^(-1)Reaction: s5 => s11, Rate Law: kass_re19*s5-kdiss_re19*s11
kass_re66 = 1.0 s^(-1); kdiss_re66 = 1.0 s^(-1)Reaction: s25 => s44, Rate Law: kass_re66*s25-kdiss_re66*s44
kass_re71 = 1.0 s^(-1); kdiss_re71 = 1.0 s^(-1)Reaction: s21 => s49, Rate Law: kass_re71*s21-kdiss_re71*s49
kass_re17 = 1.0 s^(-1); kdiss_re17 = 1.0 s^(-1)Reaction: s8 => s13, Rate Law: kass_re17*s8-kdiss_re17*s13
kass_re73 = 1.0 s^(-1); kdiss_re73 = 1.0 s^(-1)Reaction: s21 => s50, Rate Law: kass_re73*s21-kdiss_re73*s50
kass_re3 = 1.0 s^(-1); kdiss_re3 = 1.0 s^(-1)Reaction: s1 => s5, Rate Law: kass_re3*s1-kdiss_re3*s5
kdiss_re22 = 1.0 s^(-1); kass_re22 = 1.0 s^(-1)Reaction: s15 => s16, Rate Law: kass_re22*s15-kdiss_re22*s16
kass_re26 = 1.0 s^(-1); kdiss_re26 = 1.0 s^(-1)Reaction: s9 => s16, Rate Law: kass_re26*s9-kdiss_re26*s16
kass_re50 = 1.0 s^(-1); kdiss_re50 = 1.0 s^(-1)Reaction: s38 => s39, Rate Law: kass_re50*s38-kdiss_re50*s39
kass_re39 = 1.0 s^(-1); kdiss_re39 = 1.0 s^(-1)Reaction: s17 => s22, Rate Law: kass_re39*s17-kdiss_re39*s22
kass_re11 = 1.0 s^(-1); kdiss_re11 = 1.0 s^(-1)Reaction: s6 => s7, Rate Law: kass_re11*s6-kdiss_re11*s7
kdiss_re33 = 1.0 s^(-1); kass_re33 = 1.0 s^(-1)Reaction: s21 => s23, Rate Law: kass_re33*s21-kdiss_re33*s23
kass_re8 = 1.0 s^(-1); kdiss_re8 = 1.0 s^(-1)Reaction: s3 => s7, Rate Law: kass_re8*s3-kdiss_re8*s7
kass_re70 = 1.0 s^(-1); kdiss_re70 = 1.0 s^(-1)Reaction: s21 => s48, Rate Law: kass_re70*s21-kdiss_re70*s48
kass_re59 = 1.0 s^(-1); kdiss_re59 = 1.0 s^(-1)Reaction: s24 => s38, Rate Law: kass_re59*s24-kdiss_re59*s38
kdiss_re7 = 1.0 s^(-1); kass_re7 = 1.0 s^(-1)Reaction: s7 => s8, Rate Law: kass_re7*s7-kdiss_re7*s8
kdiss_re21 = 1.0 s^(-1); kass_re21 = 1.0 s^(-1)Reaction: s8 => s14, Rate Law: kass_re21*s8-kdiss_re21*s14
kdiss_re34 = 1.0 s^(-1); kass_re34 = 1.0 s^(-1)Reaction: s21 => s24, Rate Law: kass_re34*s21-kdiss_re34*s24
kdiss_re2 = 1.0 s^(-1); kass_re2 = 1.0 s^(-1)Reaction: s1 => s4, Rate Law: kass_re2*s1-kdiss_re2*s4
kass_re43 = 1.0 s^(-1); kdiss_re43 = 1.0 s^(-1)Reaction: s16 => s24, Rate Law: kass_re43*s16-kdiss_re43*s24
kass_re27 = 1.0 s^(-1); kdiss_re27 = 1.0 s^(-1)Reaction: s9 => s17, Rate Law: kass_re27*s9-kdiss_re27*s17
kass_re9 = 1.0 s^(-1); kdiss_re9 = 1.0 s^(-1)Reaction: s4 => s7, Rate Law: kass_re9*s4-kdiss_re9*s7
kdiss_re1 = 1.0 s^(-1); kass_re1 = 1.0 s^(-1)Reaction: s1 => s3, Rate Law: kass_re1*s1-kdiss_re1*s3
kdiss_re42 = 1.0 s^(-1); kass_re42 = 1.0 s^(-1)Reaction: s17 => s25, Rate Law: kass_re42*s17-kdiss_re42*s25
kass_re20 = 1.0 s^(-1); kdiss_re20 = 1.0 s^(-1)Reaction: s14 => s15, Rate Law: kass_re20*s14-kdiss_re20*s15
kass_re37 = 1.0 s^(-1); kdiss_re37 = 1.0 s^(-1)Reaction: s21 => s27, Rate Law: kass_re37*s21-kdiss_re37*s27
kdiss_re10 = 1.0 s^(-1); kass_re10 = 1.0 s^(-1)Reaction: s5 => s7, Rate Law: kass_re10*s5-kdiss_re10*s7
kdiss_re18 = 1.0 s^(-1); kass_re18 = 1.0 s^(-1)Reaction: s5 => s13, Rate Law: kass_re18*s5-kdiss_re18*s13
kdiss_re63 = 1.0 s^(-1); kass_re63 = 1.0 s^(-1)Reaction: s25 => s32, Rate Law: kass_re63*s25-kdiss_re63*s32
kass_re56 = 1.0 s^(-1); kdiss_re56 = 1.0 s^(-1)Reaction: s24 => s28, Rate Law: kass_re56*s24-kdiss_re56*s28
kdiss_re4 = 1.0 s^(-1); kass_re4 = 1.0 s^(-1)Reaction: s2 => s4, Rate Law: kass_re4*s2-kdiss_re4*s4
kass_re72 = 1.0 s^(-1); kdiss_re72 = 1.0 s^(-1)Reaction: s21 => s51, Rate Law: kass_re72*s21-kdiss_re72*s51

States:

NameDescription
s8[Mitogen-activated protein kinase kinase kinase 5]
s5[LRR receptor-like serine/threonine-protein kinase FLS2]
s7[Mitogen-activated protein kinase kinase kinase 5]
s14[Mannosyl-oligosaccharide 1,2-alpha-mannosidase MNS2]
s18[Dual specificity mitogen-activated protein kinase kinase 5]
s20[Mitogen-activated protein kinase 3]
s23[Mitogen-activated protein kinase 3]
s24[Mitogen-activated protein kinase 4]
s37[Transcription repressor MYB4]
s9[Mitogen-activated protein kinase kinase kinase 1]
s19[Dual specificity mitogen-activated protein kinase kinase 1]
s31[ATMYB2At2g47190MYB transcription factorMYB transcription factor (Atmyb2)MYB transcription factor Atmyb2Myb domain protein 2]
s10[Mitogen-activated protein kinase kinase kinase 18]
s34[WRKY transcription factor 6]
s36[Transcription repressor MYB4]
s38[Probable WRKY transcription factor 25]
s6[Probable leucine-rich repeat receptor-like serine/threonine-protein kinase At3g14840]
s32[Probable WRKY transcription factor 33]
s22[Mitogen-activated protein kinase]
s11[Mitogen-activated protein kinase kinase kinase 19Protein kinase-like protein]
s15[Mannosyl-oligosaccharide 1,2-alpha-mannosidase MNS2]
s3[LysM domain-containing GPI-anchored protein 1]
s1[173629; pathogen]
s17[Dual specificity mitogen-activated protein kinase kinase 4]
s13[Serine/threonine-protein kinase EDR1]
s25[Mitogen-activated protein kinase 6]
s2[Bacteria Latreille et al. 1825; pathogen]
s4[Pinoresinol reductase 1]
s33[Probable WRKY transcription factor 33]
s16[Mitogen-activated protein kinase kinase 2]
s21[Mitogen-activated protein kinase 3]
s28[WRKY transcription factor 1]
s39[Probable WRKY transcription factor 25]

Pawelek2016 - Within-Host Models of High and Low Pathogenic Influenza Virus Infections: MODEL1812040006v0.0.1

a possible mechanism of MP in determining HP versus LP outcomes, and how different interventions might affect infection…

Details

The World Health Organization identifies influenza as a major public health problem. While the strains commonly circulating in humans usually do not cause severe pathogenicity in healthy adults, some strains that have infected humans, such as H5N1, can cause high morbidity and mortality. Based on the severity of the disease, influenza viruses are sometimes categorized as either being highly pathogenic (HP) or having low pathogenicity (LP). The reasons why some strains are LP and others HP are not fully understood. While there are likely multiple mechanisms of interaction between the virus and the immune response that determine LP versus HP outcomes, we focus here on one component, namely macrophages (MP). There is some evidence that MP may both help fight the infection and become productively infected with HP influenza viruses. We developed mathematical models for influenza infections which explicitly included the dynamics and action of MP. We fit these models to viral load and macrophage count data from experimental infections of mice with LP and HP strains. Our results suggest that MP may not only help fight an influenza infection but may contribute to virus production in infections with HP viruses. We also explored the impact of combination therapies with antivirals and anti-inflammatory drugs on HP infections. Our study suggests a possible mechanism of MP in determining HP versus LP outcomes, and how different interventions might affect infection dynamics. link: http://identifiers.org/pubmed/26918620

Peng2013-melatonin--effects of light and routes of administration: MODEL2003190006v0.0.1

Physiologically based pharmacokinetic (PBPK) models were developed using MATLAB Simulink(®) to predict diurnal variation…

Details

Physiologically based pharmacokinetic (PBPK) models were developed using MATLAB Simulink(®) to predict diurnal variations of endogenous melatonin with light as well as pharmacokinetics of exogenous melatonin via different routes of administration. The model was structured using whole body, including pineal and saliva compartments, and parameterized based on the literature values for endogenous melatonin. It was then optimized by including various intensities of light and various dosage and formulation of melatonin. The model predictions generally have a good fit with available experimental data as evaluated by mean squared errors and ratios between model-predicted and observed values considering large variations in melatonin secretion and pharmacokinetics as reported in the literature. It also demonstrates the capability and usefulness in simulating plasma and salivary concentrations of melatonin under different light conditions and the interaction of endogenous melatonin with the pharmacokinetics of exogenous melatonin. Given the mechanistic approach and programming flexibility of MATLAB Simulink(®), the PBPK model could provide predictions of endogenous melatonin rhythms and pharmacokinetic changes in response to environmental (light) and experimental (dosage and route of administration) conditions. Furthermore, the model may be used to optimize the combined treatment using light exposure and exogenous melatonin for maximal phase advances or delays. link: http://identifiers.org/pubmed/24120727

Pepke2010_Full_Ca2/CaM_mCaMKII: MODEL1001150000v0.0.1

This the full model from the article: A dynamic model of interactions of Ca2+, calmodulin, and catalytic subunits of C…

Details

During the acquisition of memories, influx of Ca2+ into the postsynaptic spine through the pores of activated N-methyl-D-aspartate-type glutamate receptors triggers processes that change the strength of excitatory synapses. The pattern of Ca2+influx during the first few seconds of activity is interpreted within the Ca2+-dependent signaling network such that synaptic strength is eventually either potentiated or depressed. Many of the critical signaling enzymes that control synaptic plasticity,including Ca2+/calmodulin-dependent protein kinase II (CaMKII), are regulated by calmodulin, a small protein that can bindup to 4 Ca2+ ions. As a first step toward clarifying how the Ca2+-signaling network decides between potentiation or depression, we have created a kinetic model of the interactions of Ca2+, calmodulin, and CaMKII that represents our best understanding of the dynamics of these interactions under conditions that resemble those in a postsynaptic spine. We constrained parameters of the model from data in the literature, or from our own measurements, and then predicted time courses of activation and autophosphorylation of CaMKII under a variety of conditions. Simulations showed that species of calmodulin with fewer than four bound Ca2+ play a significant role in activation of CaMKII in the physiological regime,supporting the notion that processing of Ca2+ signals in a spine involves competition among target enzymes for binding to unsaturated species of CaM in an environment in which the concentration of Ca2+ is fluctuating rapidly. Indeed, we showed that dependence of activation on the frequency of Ca2+ transients arises from the kinetics of interaction of fluctuating Ca2+with calmodulin/CaMKII complexes. We used parameter sensitivity analysis to identify which parameters will be most beneficial to measure more carefully to improve the accuracy of predictions. This model provides a quantitative base from which to build more complex dynamic models of postsynaptic signal transduction during learning. link: http://identifiers.org/pubmed/20168991

Pereira2018 - Genome-scale metabolic model for Actinobacillus succinogenes 130Z: MODEL1804130001v0.0.1

The model, iBP722, was reconstructed based on the functional reannotation of the complete genome sequence of A. succinog…

Details

Actinobacillus succinogenes is a promising bacterial catalyst for the bioproduction of succinic acid from low-cost raw materials. In this work, a genome-scale metabolic model was reconstructed and used to assess the metabolic capabilities of this microorganism under producing conditions.The model, iBP722, was reconstructed based on the functional reannotation of the complete genome sequence of A. succinogenes 130Z and manual inspection of metabolic pathways, covering 1072 enzymatic reactions associated with 722 metabolic genes that involve 713 metabolites. The highly curated model was effective in capturing the growth of A. succinogenes on various carbon sources, as well as the SA production under various growth conditions with fair agreement between experimental and predicted data. Calculated flux distributions under different conditions show that a number of metabolic pathways are affected by the activity of some metabolic enzymes at key nodes in metabolism, including the transport mechanism of carbon sources and the ability to fix carbon dioxide.The established genome-scale metabolic model can be used for model-driven strain design and medium alteration to improve succinic acid yields. link: http://identifiers.org/pubmed/29843739

Perelson1993 - HIVinfection_CD4Tcells_ModelA: BIOMD0000000874v0.0.1

This a model from the article: Dynamics of HIV infection of CD4+ T cells. Perelson AS, Kirschner DE, De Boer R. Math…

Details

We examine a model for the interaction of HIV with CD4+ T cells that considers four populations: uninfected T cells, latently infected T cells, actively infected T cells, and free virus. Using this model we show that many of the puzzling quantitative features of HIV infection can be explained simply. We also consider effects of AZT on viral growth and T-cell population dynamics. The model exhibits two steady states, an uninfected state in which no virus is present and an endemically infected state, in which virus and infected T cells are present. We show that if N, the number of infectious virions produced per actively infected T cell, is less a critical value, Ncrit, then the uninfected state is the only steady state in the nonnegative orthant, and this state is stable. For N > Ncrit, the uninfected state is unstable, and the endemically infected state can be either stable, or unstable and surrounded by a stable limit cycle. Using numerical bifurcation techniques we map out the parameter regimes of these various behaviors. oscillatory behavior seems to lie outside the region of biologically realistic parameter values. When the endemically infected state is stable, it is characterized by a reduced number of T cells compared with the uninfected state. Thus T-cell depletion occurs through the establishment of a new steady state. The dynamics of the establishment of this new steady state are examined both numerically and via the quasi-steady-state approximation. We develop approximations for the dynamics at early times in which the free virus rapidly binds to T cells, during an intermediate time scale in which the virus grows exponentially, and a third time scale on which viral growth slows and the endemically infected steady state is approached. Using the quasi-steady-state approximation the model can be simplified to two ordinary differential equations the summarize much of the dynamical behavior. We compute the level of T cells in the endemically infected state and show how that level varies with the parameters in the model. The model predicts that different viral strains, characterized by generating differing numbers of infective virions within infected T cells, can cause different amounts of T-cell depletion and generate depletion at different rates. Two versions of the model are studied. In one the source of T cells from precursors is constant, whereas in the other the source of T cells decreases with viral load, mimicking the infection and killing of T-cell precursors.(ABSTRACT TRUNCATED AT 400 WORDS) link: http://identifiers.org/pubmed/8096155

Parameters:

NameDescription
N = 1000.0; mu_b = 0.24Reaction: => V; T_2, Rate Law: COMpartment*N*mu_b*T_2
k_1 = 2.4E-5; mu_V = 2.4Reaction: V => ; T, Rate Law: COMpartment*(k_1*V*T+mu_V*V)
s = 10.0; r = 0.03Reaction: => T, Rate Law: COMpartment*(s+r*T)
k_1 = 2.4E-5Reaction: => T_1; V, T, Rate Law: COMpartment*k_1*V*T
mu_T = 0.02; k_2 = 0.003Reaction: T_1 =>, Rate Law: COMpartment*(mu_T*T_1+k_2*T_1)
k_2 = 0.003Reaction: => T_2; T_1, Rate Law: COMpartment*k_2*T_1
k_1 = 2.4E-5; mu_T = 0.02; T_max = 1500.0; r = 0.03Reaction: T => ; V, T_1, T_2, Rate Law: COMpartment*(mu_T*T+k_1*V*T+r*T*(T+T_1+T_2)/T_max)
mu_b = 0.24Reaction: T_2 =>, Rate Law: COMpartment*mu_b*T_2

States:

NameDescription
T[P01730]
T 2[P01730]
T 1[P01730]
VV

Perelson1993_HIVinfection_CD4Tcells_ModelB: MODEL1006230093v0.0.1

This a model from the article: Dynamics of HIV infection of CD4+ T cells. Perelson AS, Kirschner DE, De Boer R. Math…

Details

We examine a model for the interaction of HIV with CD4+ T cells that considers four populations: uninfected T cells, latently infected T cells, actively infected T cells, and free virus. Using this model we show that many of the puzzling quantitative features of HIV infection can be explained simply. We also consider effects of AZT on viral growth and T-cell population dynamics. The model exhibits two steady states, an uninfected state in which no virus is present and an endemically infected state, in which virus and infected T cells are present. We show that if N, the number of infectious virions produced per actively infected T cell, is less a critical value, Ncrit, then the uninfected state is the only steady state in the nonnegative orthant, and this state is stable. For N > Ncrit, the uninfected state is unstable, and the endemically infected state can be either stable, or unstable and surrounded by a stable limit cycle. Using numerical bifurcation techniques we map out the parameter regimes of these various behaviors. oscillatory behavior seems to lie outside the region of biologically realistic parameter values. When the endemically infected state is stable, it is characterized by a reduced number of T cells compared with the uninfected state. Thus T-cell depletion occurs through the establishment of a new steady state. The dynamics of the establishment of this new steady state are examined both numerically and via the quasi-steady-state approximation. We develop approximations for the dynamics at early times in which the free virus rapidly binds to T cells, during an intermediate time scale in which the virus grows exponentially, and a third time scale on which viral growth slows and the endemically infected steady state is approached. Using the quasi-steady-state approximation the model can be simplified to two ordinary differential equations the summarize much of the dynamical behavior. We compute the level of T cells in the endemically infected state and show how that level varies with the parameters in the model. The model predicts that different viral strains, characterized by generating differing numbers of infective virions within infected T cells, can cause different amounts of T-cell depletion and generate depletion at different rates. Two versions of the model are studied. In one the source of T cells from precursors is constant, whereas in the other the source of T cells decreases with viral load, mimicking the infection and killing of T-cell precursors.(ABSTRACT TRUNCATED AT 400 WORDS) link: http://identifiers.org/pubmed/8096155

Perelson1993_HIVinfection_CD4Tcells_ModelC: MODEL1006230075v0.0.1

This a model from the article: Dynamics of HIV infection of CD4+ T cells. Perelson AS, Kirschner DE, De Boer R. Math…

Details

We examine a model for the interaction of HIV with CD4+ T cells that considers four populations: uninfected T cells, latently infected T cells, actively infected T cells, and free virus. Using this model we show that many of the puzzling quantitative features of HIV infection can be explained simply. We also consider effects of AZT on viral growth and T-cell population dynamics. The model exhibits two steady states, an uninfected state in which no virus is present and an endemically infected state, in which virus and infected T cells are present. We show that if N, the number of infectious virions produced per actively infected T cell, is less a critical value, Ncrit, then the uninfected state is the only steady state in the nonnegative orthant, and this state is stable. For N > Ncrit, the uninfected state is unstable, and the endemically infected state can be either stable, or unstable and surrounded by a stable limit cycle. Using numerical bifurcation techniques we map out the parameter regimes of these various behaviors. oscillatory behavior seems to lie outside the region of biologically realistic parameter values. When the endemically infected state is stable, it is characterized by a reduced number of T cells compared with the uninfected state. Thus T-cell depletion occurs through the establishment of a new steady state. The dynamics of the establishment of this new steady state are examined both numerically and via the quasi-steady-state approximation. We develop approximations for the dynamics at early times in which the free virus rapidly binds to T cells, during an intermediate time scale in which the virus grows exponentially, and a third time scale on which viral growth slows and the endemically infected steady state is approached. Using the quasi-steady-state approximation the model can be simplified to two ordinary differential equations the summarize much of the dynamical behavior. We compute the level of T cells in the endemically infected state and show how that level varies with the parameters in the model. The model predicts that different viral strains, characterized by generating differing numbers of infective virions within infected T cells, can cause different amounts of T-cell depletion and generate depletion at different rates. Two versions of the model are studied. In one the source of T cells from precursors is constant, whereas in the other the source of T cells decreases with viral load, mimicking the infection and killing of T-cell precursors.(ABSTRACT TRUNCATED AT 400 WORDS) link: http://identifiers.org/pubmed/8096155

Perelson1993_HIVinfection_CD4Tcells_ModelD: MODEL1006230035v0.0.1

This a model from the article: Dynamics of HIV infection of CD4+ T cells. Perelson AS, Kirschner DE, De Boer R. Math…

Details

We examine a model for the interaction of HIV with CD4+ T cells that considers four populations: uninfected T cells, latently infected T cells, actively infected T cells, and free virus. Using this model we show that many of the puzzling quantitative features of HIV infection can be explained simply. We also consider effects of AZT on viral growth and T-cell population dynamics. The model exhibits two steady states, an uninfected state in which no virus is present and an endemically infected state, in which virus and infected T cells are present. We show that if N, the number of infectious virions produced per actively infected T cell, is less a critical value, Ncrit, then the uninfected state is the only steady state in the nonnegative orthant, and this state is stable. For N > Ncrit, the uninfected state is unstable, and the endemically infected state can be either stable, or unstable and surrounded by a stable limit cycle. Using numerical bifurcation techniques we map out the parameter regimes of these various behaviors. oscillatory behavior seems to lie outside the region of biologically realistic parameter values. When the endemically infected state is stable, it is characterized by a reduced number of T cells compared with the uninfected state. Thus T-cell depletion occurs through the establishment of a new steady state. The dynamics of the establishment of this new steady state are examined both numerically and via the quasi-steady-state approximation. We develop approximations for the dynamics at early times in which the free virus rapidly binds to T cells, during an intermediate time scale in which the virus grows exponentially, and a third time scale on which viral growth slows and the endemically infected steady state is approached. Using the quasi-steady-state approximation the model can be simplified to two ordinary differential equations the summarize much of the dynamical behavior. We compute the level of T cells in the endemically infected state and show how that level varies with the parameters in the model. The model predicts that different viral strains, characterized by generating differing numbers of infective virions within infected T cells, can cause different amounts of T-cell depletion and generate depletion at different rates. Two versions of the model are studied. In one the source of T cells from precursors is constant, whereas in the other the source of T cells decreases with viral load, mimicking the infection and killing of T-cell precursors.(ABSTRACT TRUNCATED AT 400 WORDS) link: http://identifiers.org/pubmed/8096155

Perez-Garcia19 - Computational design of improved standardized chemotherapy protocols for grade 2 oligodendrogliomas: BIOMD0000000814v0.0.1

This is a model built by COPASI4.24(Build 197) This a model from the article: Computational design of improved standa…

Details

Here we put forward a mathematical model describing the response of low-grade (WHO grade II) oligodendrogliomas (LGO) to temozolomide (TMZ). The model describes the longitudinal volumetric dynamics of tumor response to TMZ of a cohort of 11 LGO patients treated with TMZ. After finding patient-specific parameters, different therapeutic strategies were tried computationally on the 'in-silico twins' of those patients. Chemotherapy schedules with larger-than-standard rest periods between consecutive cycles had either the same or better long-term efficacy than the standard 28-day cycles. The results were confirmed in a large trial of 2000 virtual patients. These long-cycle schemes would also have reduced toxicity and defer the appearance of resistances. On the basis of those results, a combination scheme consisting of five induction TMZ cycles given monthly plus 12 maintenance cycles given every three months was found to provide substantial survival benefits for the in-silico twins of the 11 LGO patients (median 5.69 years, range: 0.67 to 68.45 years) and in a large virtual trial including 2000 patients. We used 220 sets of experiments in-silico to show that a clinical trial incorporating 100 patients per arm (standard intensive treatment versus 5 + 12 scheme) could demonstrate the superiority of the novel scheme after a follow-up period of 10 years. Thus, the proposed treatment plan could be the basis for a standardized TMZ treatment for LGO patients with survival benefits. link: http://identifiers.org/pubmed/31306418

Parameters:

NameDescription
K = 261.799 m^3; kappa = 1.0; rho = 0.002931927433 1/dReaction: Damaged_Tumor_Cells_D => ; Tumor_Cell_Population_P, Rate Law: compartment*rho*Damaged_Tumor_Cells_D*(1-(Tumor_Cell_Population_P+Damaged_Tumor_Cells_D)/K)/kappa
lambda = 8.3184 1/dReaction: Drug_Concentration_C =>, Rate Law: compartment*lambda*Drug_Concentration_C
alpha_2 = 0.1396877593Reaction: Tumor_Cell_Population_P => ; Drug_Concentration_C, Rate Law: compartment*alpha_2*Tumor_Cell_Population_P*Drug_Concentration_C
K = 261.799 m^3; rho = 0.002931927433 1/dReaction: => Tumor_Cell_Population_P; Damaged_Tumor_Cells_D, Rate Law: compartment*rho*Tumor_Cell_Population_P*(1-(Tumor_Cell_Population_P+Damaged_Tumor_Cells_D)/K)
alpha_1 = 0.1027971308Reaction: Tumor_Cell_Population_P => Damaged_Tumor_Cells_D; Drug_Concentration_C, Rate Law: compartment*alpha_1*Tumor_Cell_Population_P*Drug_Concentration_C

States:

NameDescription
Tumor Cell Population P[cancer]
Drug Concentration C[Chemotherapy; Concentration]
Damaged Tumor Cells D[cancer; Abnormal]

PetelenzKuehn_osmoadaptation_gpd1D: BIOMD0000000610v0.0.1

Petelenz-kurdzeil2013 - Osmo adaptation gpd1DThis model is described in the article: [Quantitative analysis of glycerol…

Details

We provide an integrated dynamic view on a eukaryotic osmolyte system, linking signaling with regulation of gene expression, metabolic control and growth. Adaptation to osmotic changes enables cells to adjust cellular activity and turgor pressure to an altered environment. The yeast Saccharomyces cerevisiae adapts to hyperosmotic stress by activating the HOG signaling cascade, which controls glycerol accumulation. The Hog1 kinase stimulates transcription of genes encoding enzymes required for glycerol production (Gpd1, Gpp2) and glycerol import (Stl1) and activates a regulatory enzyme in glycolysis (Pfk26/27). In addition, glycerol outflow is prevented by closure of the Fps1 glycerol facilitator. In order to better understand the contributions to glycerol accumulation of these different mechanisms and how redox and energy metabolism as well as biomass production are maintained under such conditions we collected an extensive dataset. Over a period of 180 min after hyperosmotic shock we monitored in wild type and different mutant cells the concentrations of key metabolites and proteins relevant for osmoadaptation. The dataset was used to parameterize an ODE model that reproduces the generated data very well. A detailed computational analysis using time-dependent response coefficients showed that Pfk26/27 contributes to rerouting glycolytic flux towards lower glycolysis. The transient growth arrest following hyperosmotic shock further adds to redirecting almost all glycolytic flux from biomass towards glycerol production. Osmoadaptation is robust to loss of individual adaptation pathways because of the existence and upregulation of alternative routes of glycerol accumulation. For instance, the Stl1 glycerol importer contributes to glycerol accumulation in a mutant with diminished glycerol production capacity. In addition, our observations suggest a role for trehalose accumulation in osmoadaptation and that Hog1 probably directly contributes to the regulation of the Fps1 glycerol facilitator. Taken together, we elucidated how different metabolic adaptation mechanisms cooperate and provide hypotheses for further experimental studies. link: http://identifiers.org/pubmed/23762021

Parameters:

NameDescription
volchangespeed = 6.30627442832138E-7Reaction: cin => ; cellvol, Rate Law: intra*cin*volchangespeed/cellvol
CellSurface = 0.0296468313433281; Turgor = -0.580000000000118; vV_2 = 0.00116532; OsmoE = 0.355586; vV_T = 298.5; vV_R = 8.314; vV_1 = 3.56294E-5Reaction: => cellvol; glycerol_e, glycerol_i, cin, Rate Law: intra*vV_1*CellSurface*(Turgor-vV_2*vV_R*vV_T*((glycerol_e+OsmoE)-(glycerol_i+cin)))
kv7_2 = 0.317879; kv7_1 = 0.00983997Reaction: trioseP => pyruvate, Rate Law: intra*kv7_1*trioseP/(kv7_2+trioseP)
kv23r_1 = 2.09875E-4Reaction: AOG => AOGi, Rate Law: intra*kv23r_1*AOG
v10speed = 1.30212691282784E-11; initcellnum = 6954722.464; cellnum = 1.11543272115466E7Reaction: => trehalose_e, Rate Law: extra*(v10speed*cellnum/initcellnum-v10speed)
v13aspeed = 5.0275596254809E-8; initcellnum = 6954722.464; cellnum = 1.11543272115466E7Reaction: => glycerol_e, Rate Law: extra*(v13aspeed*cellnum/initcellnum-v13aspeed)
v1speed = 5.33353293880484E-7; initcellnum = 6954722.464; cellnum = 1.11543272115466E7Reaction: glucose_e =>, Rate Law: extra*(v1speed*cellnum/initcellnum-v1speed)
kv18r_1 = 1.32549E-4Reaction: Gpd1 =>, Rate Law: intra*kv18r_1*Gpd1
kv16r_1VARIABLE = 0.444296Reaction: Hog1PP => Hog1, Rate Law: intra*kv16r_1VARIABLE*Hog1PP
kv13a_1 = 6.28899E-6; CellSurface = 0.0296468313433281; kDiff=1.0Reaction: glycerol_i => glycerol_e; Fps1r, Rate Law: kv13a_1*CellSurface*Fps1r*(glycerol_i-kDiff*glycerol_e)
kv20r_1 = 7.05933E-4Reaction: stl1mRNA =>, Rate Law: intra*kv20r_1*stl1mRNA
kv13b_2 = 3.69196E-7; kv13b_1 = 1.27001E-7Reaction: glycerol_e => glycerol_i; Stl1, Rate Law: glycerol_e*kv13b_1*Stl1/(kv13b_2+glycerol_e)
kv4_3 = 0.00171631; kv4_1 = 0.0628885; kv4_4 = 2.67143; kv4_2 = 0.00230714; kv4_5 = 0.583865Reaction: G6P => F16DP; F26DP, Rate Law: intra*(kv4_2*(1-F26DP^kv4_5/(F26DP+kv4_3)^kv4_5)+kv4_1*F26DP^kv4_5/(F26DP+kv4_3)^kv4_5)*(G6P/kv4_4)^2/(1+(G6P/kv4_4)^2)
kv15r_2 = 3.3187E-5; kv15r_1 = 1.84829E-7Reaction: F26DP => G6P, Rate Law: intra*kv15r_1*F26DP/(kv15r_2+F26DP)
kv2_1 = 0.00303855; kv2_2 = 0.40864Reaction: glucose_i => G6P, Rate Law: intra*kv2_1*glucose_i/(kv2_2+glucose_i)
kv19r_1 = 0.0605655Reaction: Pfk2627a => Pfk2627i, Rate Law: intra*kv19r_1*Pfk2627a
kv22_2 = 0.0215179; Turgor = -0.580000000000118; kv22_1 = 8.0075; kv22_3 = 0.0554729Reaction: => Fps1r; Hog1PP, Rate Law: intra*(kv22_1*(-Turgor)/(kv22_3+(-Turgor))*1.5*(1-Hog1PP/(Hog1PP+kv22_2))-kv22_1*Fps1r)
kv8_2 = 1.50827; kv8_1 = 0.0135676Reaction: pyruvate => acetate_i, Rate Law: intra*kv8_1*pyruvate/(kv8_2+pyruvate)
Vm = 4.80000000000001E-4; kv16f_3 = 14.9448; OsmoE = 0.355586; kv16f_1 = 0.156118; kv16f_2 = 4.52424E-4Reaction: Hog1 => Hog1PP, Rate Law: intra*Hog1*kv16f_1*OsmoE*(kv16f_2/Vm)^kv16f_3
kv19f_1 = 0.299127Reaction: Pfk2627i => Pfk2627a; Hog1PP, Rate Law: intra*kv19f_1*Hog1PP*Pfk2627i
v11speed = 9.21581643247704E-8; initcellnum = 6954722.464; cellnum = 1.11543272115466E7Reaction: => acetate_e, Rate Law: extra*(v11speed*cellnum/initcellnum-v11speed)
kv18f_1 = 0.00646553Reaction: => Gpd1; gpd1mRNA, Rate Law: intra*kv18f_1*gpd1mRNA
kv23f_1 = 8.80535E-6; Vm = 4.80000000000001E-4; kv23f_3 = 6.95727; kv23f_2 = 5.1235E-4Reaction: AOGi => AOG, Rate Law: intra*AOGi*kv23f_1*(kv23f_2/Vm)^kv23f_3
kv1_2 = 0.899814; kv1_1 = 5.05249E-6Reaction: glucose_e => glucose_i, Rate Law: kv1_1*glucose_e/(kv1_2+glucose_e)
kv5_1 = 0.00383315; kv5_2 = 1.74463; kv5_3 = 0.00656128; kv5_4 = 1.13994Reaction: F16DP => trioseP, Rate Law: intra*(kv5_1*F16DP/kv5_2/(1+F16DP/kv5_2)-kv5_3*trioseP/kv5_4/(1+trioseP/kv5_4))
kv17r_1 = 0.00151498Reaction: gpd1mRNA =>, Rate Law: intra*kv17r_1*gpd1mRNA
kv9_1 = 0.214937; kv9_2 = 0.923665Reaction: pyruvate => ethanol_i, Rate Law: intra*kv9_1*pyruvate/(kv9_2+pyruvate)
CellSurface = 0.0296468313433281; kv12_2 = 0.148586; kv12_1 = 1.00927E-5Reaction: ethanol_i => ethanol_e, Rate Law: kv12_1*CellSurface*(ethanol_i-kv12_2*ethanol_e)
kv14_5 = 1.23049; kv14_2 = 6.05922E-6; OsmoE = 0.355586; kv14_4 = 0.420621; kv14_1 = 0.808051; kv14_3 = 2.05157Reaction: G6P => biomass; cellvol, Rate Law: intra*kv14_1*cellvol^kv14_3/(cellvol^kv14_3+kv14_2)*(1-OsmoE/(OsmoE+kv14_4))*G6P/kv14_5/(1+G6P/kv14_5)
kv20f_3 = 4.05843E-6; kv20f_2 = 0.0167845; kv20f_x = 1.55858; kv20f_1 = 9.81887E-5Reaction: => stl1mRNA; Hog1PP, Rate Law: intra*(kv20f_1*Hog1PP^kv20f_x/(Hog1PP^kv20f_x+kv20f_2)+kv20f_3)
kv6b_4 = 4.61918E-5; kv6b_x = 28.5; kv6b_5 = 0.292627Reaction: trioseP => glycerol_i, Rate Law: intra*kv6b_x*kv6b_4*trioseP^2/kv6b_5/(1+trioseP^2/kv6b_5)
initcellnum = 6954722.464; cellnum = 1.11543272115466E7; v13bspeed = 1.59327705289657E-11Reaction: glycerol_e =>, Rate Law: extra*(v13bspeed*cellnum/initcellnum-v13bspeed)
kv21r_1 = 2.14247E-4Reaction: Stl1 =>, Rate Law: intra*kv21r_1*Stl1
kv10_1 = 1.83291E-7; CellSurface = 0.0296468313433281; kv10_2 = 4.26512Reaction: trehalose => trehalose_e, Rate Law: kv10_1*CellSurface*(trehalose-kv10_2*trehalose_e)
kv15f_2 = 6.95877; kv15f_1 = 4.99507E-5Reaction: G6P => F26DP; Pfk2627a, Rate Law: intra*G6P*kv15f_1*Pfk2627a/(kv15f_2+G6P)
kv21f_1 = 0.00121673Reaction: => Stl1; stl1mRNA, Rate Law: intra*kv21f_1*stl1mRNA
kv3_4 = 0.166996; kv3_1 = 6.17387E-6; kv3_3 = 7.37808E-4; kv3_2 = 0.81114Reaction: G6P => trehalose, Rate Law: intra*(kv3_1*G6P/kv3_2-kv3_3*trehalose/kv3_4)/(1+G6P/kv3_2+trehalose/kv3_4)
CellSurface = 0.0296468313433281; kv11_2 = 1.17279; kv11_1 = 3.2863E-6Reaction: acetate_i => acetate_e, Rate Law: kv11_1*CellSurface*(acetate_i-kv11_2*acetate_e)
initcellnum = 6954722.464; cellnum = 1.11543272115466E7; v12speed = 2.88652220351019E-6Reaction: => ethanol_e, Rate Law: extra*(v12speed*cellnum/initcellnum-v12speed)

States:

NameDescription
glucose i[glucose; intracellular]
acetate e[acetate]
trehalose e[trehalose]
gpd1mRNA[S000002180]
Stl1[Sugar transporter STL1]
Pfk2627i[S000005496; S000001369]
Hog1PP[Mitogen-activated protein kinase HOG1]
stl1mRNA[S000002944]
glucose e[glucose]
F26DP[105021]
glycerol i[glycerol]
pyruvate[pyruvate]
ethanol e[ethanol]
Pfk2627a[S000005496; S000001369]
AOG[positive regulation of transcription, DNA-templated]
acetate i[acetate]
Hog1[Mitogen-activated protein kinase HOG1]
F16DP[keto-D-fructose 1,6-bisphosphate]
cellvolcellvol
biomassbiomass
ethanol i[ethanol]
trioseP[4643300; 729]
G6P[alpha-D-glucose 6-phosphate]
AOGi[positive regulation of transcription, DNA-templated]
Fps1r[Glycerol uptake/efflux facilitator protein]
cin[osmolyte]
glycerol e[glycerol]
trehalose[trehalose]
Gpd1[Glycerol-3-phosphate dehydrogenase [NAD(+)] 1]

Peterson2010 - Integrated calcium homeostasis and bone remodelling: BIOMD0000000613v0.0.1

&lt;notes xmlns=&quot;http://www.sbml.org/sbml/level3/version1/core&quot;&gt; &lt;body xmlns=&quot;http://www.w3.…

Details

Bone biology is physiologically complex and intimately linked to calcium homeostasis. The literature provides a wealth of qualitative and/or quantitative descriptions of cellular mechanisms, bone dynamics, associated organ dynamics, related disease sequela, and results of therapeutic interventions. We present a physiologically based mathematical model of integrated calcium homeostasis and bone biology constructed from literature data. The model includes relevant cellular aspects with major controlling mechanisms for bone remodeling and calcium homeostasis and appropriately describes a broad range of clinical and therapeutic conditions. These include changes in plasma parathyroid hormone (PTH), calcitriol, calcium and phosphate (PO4), and bone-remodeling markers as manifested by hypoparathyroidism and hyperparathyroidism, renal insufficiency, daily PTH 1-34 administration, and receptor activator of NF-kappaB ligand (RANKL) inhibition. This model highlights the utility of systems approaches to physiologic modeling in the bone field. The presented bone and calcium homeostasis model provides an integrated mathematical construct to conduct hypothesis testing of influential system aspects, to visualize elements of this complex endocrine system, and to continue to build upon iteratively with the results of ongoing scientific research. link: http://identifiers.org/pubmed/19732857

Parameters:

NameDescription
J14 = NaNReaction: Q => P, Rate Law: J14
koutRNK = 0.00323667Reaction: RNK => ; RNK, Rate Law: koutRNK*RNK
k2 = 0.112013Reaction: N => ; N, Rate Law: k2*N
kinOC2 = NaNReaction: => OC, Rate Law: kinOC2
kbslow = NaNReaction: OBslow => ; OBslow, Rate Law: kbslow*OBslow
k3 = 6.24E-6Reaction: L + RNK => M; RNK, L, Rate Law: k3*RNK*L
pO = NaNReaction: => O, Rate Law: pO
TERIPK = NaNReaction: TERISC => PTH, Rate Law: TERIPK
koutL = 0.00293273Reaction: L => ; L, Rate Law: koutL*L
kO = 15.8885Reaction: O => ; O, Rate Law: kO*O
crebKout = 0.00279513Reaction: CREB => ; CREB, Rate Law: crebKout*CREB
T76 = NaNReaction: => S; S, Rate Law: (1-S)*T76
J48 = NaNReaction: ECCPhos =>, Rate Law: J48
RX2Kout = NaNReaction: RX2 => ; RX2, Rate Law: RX2Kout*RX2
RX2Kin = NaNReaction: => RX2, Rate Law: RX2Kin
kout = NaNReaction: PTH => ; PTH, Rate Law: kout*PTH
J40 = NaNReaction: T => P, Rate Law: J40
J53 = NaNReaction: PhosGut => ECCPhos, Rate Law: J53
OralCa = NaN; F11 = NaNReaction: => T, Rate Law: OralCa*F11
KPT = NaNReaction: ROB1 => ; ROB1, Rate Law: KPT*ROB1
crebKin = NaNReaction: => CREB, Rate Law: crebKin
F12 = 0.7; OralPhos = NaNReaction: => PhosGut, Rate Law: OralPhos*F12
IPTHint = 0.0Reaction: => SC, Rate Law: IPTHint
koutTGFact = NaNReaction: TGFBact => ; TGFBact, Rate Law: koutTGFact*TGFBact
SPTH = NaNReaction: => PTH, Rate Law: SPTH
J14a = NaNReaction: Qbone => Q, Rate Law: J14a
J15 = NaNReaction: P => Q, Rate Law: J15
D = NaN; FracOBfast = 0.797629; Frackb = 0.313186; PicOB = NaN; bigDb = NaNReaction: => OBslow, Rate Law: bigDb/PicOB*D*(1-FracOBfast)*Frackb
J42 = NaNReaction: ECCPhos =>, Rate Law: J42
koutTGFeqn = NaNReaction: TGFB => TGFBact, Rate Law: koutTGFeqn
kinL = NaNReaction: => L, Rate Law: kinL
J41 = NaNReaction: => ECCPhos, Rate Law: J41
kbfast = NaNReaction: OBfast => ; OBfast, Rate Law: kbfast*OBfast
J27 = NaNReaction: P =>, Rate Law: J27
SE = NaNReaction: => A, Rate Law: SE
J15a = NaNReaction: Q => Qbone, Rate Law: J15a
PTin = NaNReaction: => PTmax, Rate Law: PTin
PTout = 1.604E-4Reaction: PTmax => ; PTmax, Rate Law: PTout*PTmax
T64 = 0.05Reaction: A => ; A, Rate Law: T64*A
kinRNKgam = 0.151825; kinRNK = NaNReaction: => RNK; TGFBact, TGFBact, Rate Law: kinRNK*TGFBact^kinRNKgam
kLShap = NaNReaction: HAp => ; HAp, Rate Law: kLShap*HAp
T36 = NaNReaction: => R; R, Rate Law: T36*(1-R)
T37 = NaNReaction: R => ; R, Rate Law: T37*R
ROBin = NaNReaction: => ROB1, Rate Law: ROBin
J56 = NaNReaction: IntraPO => ECCPhos, Rate Law: J56
k4 = 0.112013Reaction: M => L + RNK; M, Rate Law: k4*M
D = NaN; FracOBfast = 0.797629; PicOB = NaN; bigDb = NaN; Frackb2 = NaNReaction: => OBfast, Rate Law: bigDb/PicOB*D*FracOBfast*Frackb2
J54 = NaNReaction: ECCPhos => IntraPO, Rate Law: J54
bcl2Kout = 0.693Reaction: BCL2 => ; BCL2, Rate Law: bcl2Kout*BCL2
k1 = 6.24E-6Reaction: => N; O, L, O, L, Rate Law: k1*O*L
T69 = 0.1Reaction: B => ; B, Rate Law: T69*B
Osteoblast = NaN; kinTGF = NaN; OB0 = NaN; OBtgfGAM = 0.0111319Reaction: => TGFB, Rate Law: kinTGF*(Osteoblast/OB0)^OBtgfGAM
Osteoblast = NaN; kHApIn = NaNReaction: => HAp, Rate Law: kHApIn*Osteoblast
T75 = NaNReaction: S => ; S, Rate Law: S*T75
bcl2Kin = NaNReaction: => BCL2, Rate Law: bcl2Kin
KLSoc = NaNReaction: OC => ; OC, Rate Law: KLSoc*OC

States:

NameDescription
Q[calcium(2+); intracellular]
TGFB[Transforming growth factor beta-1]
IntraPO[phosphate ion]
T[calcium(2+)]
RNK[Tumor necrosis factor receptor superfamily member 11A]
P[calcium(2+)]
L[Tumor necrosis factor ligand superfamily member 11]
PTH[Parathyroid hormone]
OC[osteoclast]
O[Tumor necrosis factor receptor superfamily member 11B]
TGFBact[Transforming growth factor beta-1; active]
B[calcitriol]
M[protein complex; Tumor necrosis factor ligand superfamily member 11; Tumor necrosis factor receptor superfamily member 11A]
N[protein complex; Tumor necrosis factor ligand superfamily member 11; Tumor necrosis factor receptor superfamily member 11B]
ECCPhos[phosphate ion]
A[25-hydroxyvitamin D-1 alpha hydroxylase, mitochondrial]
CREB[Cyclic AMP-responsive element-binding protein 1]
SC[subcutaneous adipose tissue; Parathyroid hormone; pharmaceutical]
RX2[Runt-related transcription factor 2]
BCL2[Apoptosis regulator Bcl-2]
TERISC[16132393]
OBslow[osteoblast]
PTmax[Parathyroid hormone]
S[Parathyroid hormone]
OBfast[osteoblast]
Qbone[calcium(2+); extracellular region]
HAp[apatite]
ROB1[osteoclast; urn:miriam:pato:PATO%3A0000487+]
R[intestine; calcium(2+)]
PhosGut[phosphate ion]

Petrov2018 - C-547 a 6-methyluracil derivative with long-lasting binding and rebinding on acetylcholinesterase: MODEL1910240001v0.0.1

C-547, a candidate drug, is a potent slow-binding inhibitor of acetyl-cholinesterase, and the focus of this PK/PD model,…

Details

C-547, a potent slow-binding inhibitor of acetylcholinesterase (AChE) was intravenously administered to rat (0.05 mg/kg). Pharmacokinetic profiles were determined in blood and different organs: extensor digitorum longus muscle, heart, liver, lungs and kidneys as a function of time. Pharmacokinetics (PK) was studied using non-compartmental and compartmental analyses. A 3-compartment model describes PK in blood. Most of injected C-547 binds to albumin in the bloodstream. The steady-state volume of distribution (3800 ml/kg) is 15 times larger than the distribution volume, indicating a good tissue distribution. C-547 is slowly eliminated (kel = 0.17 h-1; T1/2 = 4 h) from the bloodstream. Effect of C-547 on animal model of myasthenia gravis persists for more than 72 h, even though the drug is not analytically detectable in the blood. A PK/PD model was built to account for such a pharmacodynamical (PD) effect. Long-lasting effect results from micro-PD mechanisms: the slow-binding nature of inhibition, high affinity for AChE and long residence time on target at neuromuscular junction (NMJ). In addition, NMJ spatial constraints i.e. high concentration of AChE in a small volume, and slow diffusion rate of free C-547 out of NMJ, make possible effective rebinding of ligand. Thus, compared to other cholinesterase inhibitors used for palliative treatment of myasthenia gravis, C-547 is the most selective drug, displays a slow pharmacokinetics, and has the longest duration of action. This makes C-547 a promising drug leader for treatment of myasthenia gravis, and a template for development of other drugs against neurological diseases and for neuroprotection. link: http://identifiers.org/pubmed/29277489

Peyraud2016 - Metabolic reconstruction (iRP1476) of Ralstonia solanacearum GMI1000: MODEL1612020000v0.0.1

Peyraud2016 - Metabolic reconstruction (iRP1476) of Ralstonia solanacearum GMI1000This model is described in the article…

Details

Bacterial pathogenicity relies on a proficient metabolism and there is increasing evidence that metabolic adaptation to exploit host resources is a key property of infectious organisms. In many cases, colonization by the pathogen also implies an intensive multiplication and the necessity to produce a large array of virulence factors, which may represent a significant cost for the pathogen. We describe here the existence of a resource allocation trade-off mechanism in the plant pathogen R. solanacearum. We generated a genome-scale reconstruction of the metabolic network of R. solanacearum, together with a macromolecule network module accounting for the production and secretion of hundreds of virulence determinants. By using a combination of constraint-based modeling and metabolic flux analyses, we quantified the metabolic cost for production of exopolysaccharides, which are critical for disease symptom production, and other virulence factors. We demonstrated that this trade-off between virulence factor production and bacterial proliferation is controlled by the quorum-sensing-dependent regulatory protein PhcA. A phcA mutant is avirulent but has a better growth rate than the wild-type strain. Moreover, a phcA mutant has an expanded metabolic versatility, being able to metabolize 17 substrates more than the wild-type. Model predictions indicate that metabolic pathways are optimally oriented towards proliferation in a phcA mutant and we show that this enhanced metabolic versatility in phcA mutants is to a large extent a consequence of not paying the cost for virulence. This analysis allowed identifying candidate metabolic substrates having a substantial impact on bacterial growth during infection. Interestingly, the substrates supporting well both production of virulence factors and growth are those found in higher amount within the plant host. These findings also provide an explanatory basis to the well-known emergence of avirulent variants in R. solanacearum populations in planta or in stressful environments. link: http://identifiers.org/pubmed/27732672

Pfeiffer2001_ATP-ProducingPathways_CooperationCompetition: BIOMD0000000337v0.0.1

This model is from the article: Cooperation and Competition in the Evolution of ATP-Producing Pathways Thomas Pfeiff…

Details

Heterotrophic organisms generally face a trade-off between rate and yield of adenosine triphosphate (ATP) production. This trade-off may result in an evolutionary dilemma, because cells with a higher rate but lower yield of ATP production may gain a selective advantage when competing for shared energy resources. Using an analysis of model simulations and biochemical observations, we show that ATP production with a low rate and high yield can be viewed as a form of cooperative resource use and may evolve in spatially structured environments. Furthermore, we argue that the high ATP yield of respiration may have facilitated the evolutionary transition from unicellular to undifferentiated multicellular organisms. link: http://identifiers.org/pubmed/11283355

Parameters:

NameDescription
v = 10.0 dimensionlessReaction: => S, Rate Law: v
d = 1.0 dimensionlessReaction: N1 =>, Rate Law: d*N1

States:

NameDescription
S[energy]
N1[cell]
N2[cell]

Phan2017 - innate immune in oncolytic virotherapy: BIOMD0000000748v0.0.1

The paper describes a model on the key components for tumor–immune dynamics in multiple myeloma. Created by COPASI 4.2…

Details

The complexity of the immune responses is a major challenge in current virotherapy. This study incorporates the innate immune response into our basic model for virotherapy and investigates how the innate immunity affects the outcome of virotherapy. The viral therapeutic dynamics is largely determined by the viral burst size, relative innate immune killing rate, and relative innate immunity decay rate. The innate immunity may complicate virotherapy in the way of creating more equilibria when the viral burst size is not too big, while the dynamics is similar to the system without innate immunity when the viral burst size is big. link: http://identifiers.org/pubmed/29379572

Parameters:

NameDescription
c = 0.48 1Reaction: y => ; z, Rate Law: tumor_microenvironment*c*y*z
e = 0.2 1Reaction: v =>, Rate Law: tumor_microenvironment*e*v
m = 0.6 1Reaction: => z; y, Rate Law: tumor_microenvironment*m*y*z
a = 0.11 1Reaction: x + v => y, Rate Law: tumor_microenvironment*a*x*v
d = 0.16 1Reaction: v => ; z, Rate Law: tumor_microenvironment*d*v*z
n = 0.036 1Reaction: z =>, Rate Law: tumor_microenvironment*n*z
r = 0.36 1Reaction: => x, Rate Law: tumor_microenvironment*r*x
b = 9.0 1Reaction: => v; y, Rate Law: tumor_microenvironment*b*y

States:

NameDescription
v[Oncolytic Virus]
x[neoplastic cell]
z[Effector Immune Cell]
y[neoplastic cell]

Phillips2003 - The Mechanism of Ras GTPase Activation by Neurofibromin: BIOMD0000000692v0.0.1

Phillips2003 - The Mechanism of Ras GTPase Activation by NeurofibrominA mathematical model for Ras-GTP activation by neu…

Details

Individual rate constants have been determined for each step of the Ras.GTP hydrolysis mechanism, activated by neurofibromin. Fluorescence intensity and anisotropy stopped-flow measurements used the fluorescent GTP analogue, mantGTP (2'(3')-O-(N-methylanthraniloyl)GTP), to determine rate constants for binding and release of neurofibromin. Quenched flow measurements provided the kinetics of the hydrolytic cleavage step. The fluorescent phosphate sensor, MDCC-PBP was used to measure phosphate release kinetics. Phosphate-water oxygen exchange, using (18)O-substituted GTP and inorganic phosphate (P(i)), was used to determine the extent of reversal of the hydrolysis step and of P(i) binding. The data show that neurofibromin and P(i) dissociate from the NF1.Ras.GDP.P(i) complex with identical kinetics, which are 3-fold slower than the preceding cleavage step. A model is presented in which the P(i) release is associated with the change of Ras from "GTP" to "GDP" conformation. In this model, the conformation change on P(i) release causes the large change in affinity of neurofibromin, which then dissociates rapidly. link: http://identifiers.org/pubmed/12667087

Parameters:

NameDescription
kf=1.02102E-11; kb=1.15192E-13Reaction: RasGTP_minus_NF1_star_ => RasGDP_minus_NF1_Pi, Rate Law: geometry*(kf*RasGTP_minus_NF1_star_-kb*RasGDP_minus_NF1_Pi)/geometry
kb=2.8798E-12; kf=2.18865E-10Reaction: RasGTP_minus_NF1 => RasGTP_minus_NF1_star_, Rate Law: geometry*(kf*RasGTP_minus_NF1-kb*RasGTP_minus_NF1_star_)/geometry
kb=5.65482E-17; kf=2.0944E-11Reaction: RasGDP_minus_NF1_Pi => Pi + RasGDP_NF1, Rate Law: geometry*(kf*RasGDP_minus_NF1_Pi-kb*Pi*RasGDP_NF1)/geometry
kf=6.28318E-13; kb=3.3301E-12Reaction: RasGTP + NF1 => RasGTP_minus_NF1, Rate Law: geometry*(kf*RasGTP*NF1-kb*RasGTP_minus_NF1)/geometry
kb=6.28318E-13; kf=2.43474E-11Reaction: RasGDP_NF1 => RasGDP + NF1, Rate Law: geometry*(kf*RasGDP_NF1-kb*RasGDP*NF1)/geometry

States:

NameDescription
RasGDP[GDP; 43873]
RasGDP minus NF1 Pi[GDP; inorganic phosphate; K08052; 43873]
RasGDP NF1[K08052; GDP; 43873]
Pi[inorganic phosphate]
NF1[K08052]
RasGTP[GTP; 43873]
RasGTP minus NF1[K08052; GTP; 43873]
RasGTP minus NF1 star[K08052; GTP; 43873]

Phillips2007_AscendingArousalSystem_SleepWakeDynamics: BIOMD0000000917v0.0.1

This a model from the article: A quantitative model of sleep-wake dynamics based on the physiology of the brainstem as…

Details

A quantitative, physiology-based model of the ascending arousal system is developed, using continuum neuronal population modeling, which involves averaging properties such as firing rates across neurons in each population. The model includes the ventrolateral preoptic area (VLPO), where circadian and homeostatic drives enter the system, the monoaminergic and cholinergic nuclei of the ascending arousal system, and their interconnections. The human sleep-wake cycle is governed by the activities of these nuclei, which modulate the behavioral state of the brain via diffuse neuromodulatory projections. The model parameters are not free since they correspond to physiological observables. Approximate parameter bounds are obtained by requiring consistency with physiological and behavioral measures, and the model replicates the human sleep-wake cycle, with physiologically reasonable voltages and firing rates. Mutual inhibition between the wake-promoting monoaminergic group and sleep-promoting VLPO causes ;;flip-flop'' behavior, with most time spent in 2 stable steady states corresponding to wake and sleep, with transitions between them on a timescale of a few minutes. The model predicts hysteresis in the sleep-wake cycle, with a region of bistability of the wake and sleep states. Reducing the monoaminergic-VLPO mutual inhibition results in a smaller hysteresis loop. This makes the model more prone to wake-sleep transitions in both directions and makes the states less distinguishable, as in narcolepsy. The model behavior is robust across the constrained parameter ranges, but with sufficient flexibility to describe a wide range of observed phenomena. link: http://identifiers.org/pubmed/17440218

Parameters:

NameDescription
chi = 10.8; Qm = 4.74258731775668; mu = 3.6Reaction: => Somnogen_level_H, Rate Law: COMpartment*(mu*Qm-Somnogen_level_H)/chi
Qm = 4.74258731775668; tau_v = 10.0; D = -10.7; v_vm = -1.9Reaction: => Ventrolateral_preopticarea__VLPO__voltage, Rate Law: COMpartment*((v_vm*Qm+D)-Ventrolateral_preopticarea__VLPO__voltage)/(tau_v/3600)
v_mv = -1.9; Qv = 0.127101626308136; v_maQao = 1.0; tau_m = 10.0Reaction: => Monoaminergic__MA__voltage, Rate Law: COMpartment*((v_maQao+v_mv*Qv)-Monoaminergic__MA__voltage)/(tau_m/3600)

States:

NameDescription
Ventrolateral preopticarea VLPO voltage[OMIT_0027571; OMIT_0026787; Signal; C70813]
Somnogen level H[C207]
Monoaminergic MA voltage[C70813; OMIT_0026787; C73238; C62025; Signal; C2321]

Phillips2008_AscendingArousalSystem_Baseline: MODEL1006230110v0.0.1

This a model from the article: Sleep deprivation in a quantitative physiologically based model of the ascending arousa…

Details

A physiologically based quantitative model of the human ascending arousal system is used to study sleep deprivation after being calibrated on a small set of experimentally based criteria. The model includes the sleep-wake switch of mutual inhibition between nuclei which use monoaminergic neuromodulators, and the ventrolateral preoptic area. The system is driven by the circadian rhythm and sleep homeostasis. We use a small number of experimentally derived criteria to calibrate the model for sleep deprivation, then investigate model predictions for other experiments, demonstrating the scope of application. Calibration gives an improved parameter set, in which the form of the homeostatic drive is better constrained, and its weighting relative to the circadian drive is increased. Within the newly constrained parameter ranges, the model predicts repayment of sleep debt consistent with experiment in both quantity and distribution, asymptoting to a maximum repayment for very long deprivations. Recovery is found to depend on circadian phase, and the model predicts that it is most efficient to recover during normal sleeping phases of the circadian cycle, in terms of the amount of recovery sleep required. The form of the homeostatic drive suggests that periods of wake during recovery from sleep deprivation are phases of relative recovery, in the sense that the homeostatic drive continues to converge toward baseline levels. This undermines the concept of sleep debt, and is in agreement with experimentally restricted recovery protocols. Finally, we compare our model to the two-process model, and demonstrate the power of physiologically based modeling by correctly predicting sleep latency times following deprivation from experimental data. link: http://identifiers.org/pubmed/18805427

Phillips2008_AscendingArousalSystem_SleepDeprivation: MODEL1006230115v0.0.1

This a model from the article: Sleep deprivation in a quantitative physiologically based model of the ascending arousa…

Details

A physiologically based quantitative model of the human ascending arousal system is used to study sleep deprivation after being calibrated on a small set of experimentally based criteria. The model includes the sleep-wake switch of mutual inhibition between nuclei which use monoaminergic neuromodulators, and the ventrolateral preoptic area. The system is driven by the circadian rhythm and sleep homeostasis. We use a small number of experimentally derived criteria to calibrate the model for sleep deprivation, then investigate model predictions for other experiments, demonstrating the scope of application. Calibration gives an improved parameter set, in which the form of the homeostatic drive is better constrained, and its weighting relative to the circadian drive is increased. Within the newly constrained parameter ranges, the model predicts repayment of sleep debt consistent with experiment in both quantity and distribution, asymptoting to a maximum repayment for very long deprivations. Recovery is found to depend on circadian phase, and the model predicts that it is most efficient to recover during normal sleeping phases of the circadian cycle, in terms of the amount of recovery sleep required. The form of the homeostatic drive suggests that periods of wake during recovery from sleep deprivation are phases of relative recovery, in the sense that the homeostatic drive continues to converge toward baseline levels. This undermines the concept of sleep debt, and is in agreement with experimentally restricted recovery protocols. Finally, we compare our model to the two-process model, and demonstrate the power of physiologically based modeling by correctly predicting sleep latency times following deprivation from experimental data. link: http://identifiers.org/pubmed/18805427

Phosphatase activities on PI(3,4,5)P3 and PI(3,4)P2: MODEL1704190000v0.0.1

Phosphatase activities on PI(3,4,5)P3 and PI(3,4)P2This model describes the action of various phosphatases on PI(3,4,5)P…

Details

The PI3K signaling pathway regulates cell growth and movement and is heavily mutated in cancer. Class I PI3Ks synthesize the lipid messenger PI(3,4,5)P3. PI(3,4,5)P3 can be dephosphorylated by 3- or 5-phosphatases, the latter producing PI(3,4)P2. The PTEN tumor suppressor is thought to function primarily as a PI(3,4,5)P3 3-phosphatase, limiting activation of this pathway. Here we show that PTEN also functions as a PI(3,4)P2 3-phosphatase, both in vitro and in vivo. PTEN is a major PI(3,4)P2 phosphatase in Mcf10a cytosol, and loss of PTEN and INPP4B, a known PI(3,4)P2 4-phosphatase, leads to synergistic accumulation of PI(3,4)P2, which correlated with increased invadopodia in epidermal growth factor (EGF)-stimulated cells. PTEN deletion increased PI(3,4)P2 levels in a mouse model of prostate cancer, and it inversely correlated with PI(3,4)P2 levels across several EGF-stimulated prostate and breast cancer lines. These results point to a role for PI(3,4)P2 in the phenotype caused by loss-of-function mutations or deletions in PTEN. link: http://identifiers.org/doi/10.1016/j.molcel.2017.09.024

Piedrafita2010_MR_System: BIOMD0000000257v0.0.1

This is the self maintaining metabolism model described in the article: A Simple Self-Maintaining Metabolic System:…

Details

A living organism must not only organize itself from within; it must also maintain its organization in the face of changes in its environment and degradation of its components. We show here that a simple (M,R)-system consisting of three interlocking catalytic cycles, with every catalyst produced by the system itself, can both establish a non-trivial steady state and maintain this despite continuous loss of the catalysts by irreversible degradation. As long as at least one catalyst is present at a sufficient concentration in the initial state, the others can be produced and maintained. The system shows bistability, because if the amount of catalyst in the initial state is insufficient to reach the non-trivial steady state the system collapses to a trivial steady state in which all fluxes are zero. It is also robust, because if one catalyst is catastrophically lost when the system is in steady state it can recreate the same state. There are three elementary flux modes, but none of them is an enzyme-maintaining mode, the entire network being necessary to maintain the two catalysts. link: http://identifiers.org/pubmed/20700491

Parameters:

NameDescription
k10r = 0.05 per_time_per_M; k10 = 0.05 per_timeReaction: STUSU => STU + SU, Rate Law: env*(k10*STUSU-k10r*STU*SU)
k2 = 10.0 per_time_per_M; k2r = 10.0 per_timeReaction: T + STUS => STUST, Rate Law: env*(k2*T*STUS-k2r*STUST)
k6r = 1.0 per_time; k6 = 1.0 per_time_per_MReaction: U + SUST => SUSTU, Rate Law: env*(k6*U*SUST-k6r*SUSTU)
k1r = 10.0 per_time; k1 = 10.0 per_time_per_MReaction: S + STU => STUS, Rate Law: env*(k1*S*STU-k1r*STUS)
k5 = 1.0 per_time_per_M; k5r = 1.0 per_timeReaction: SU + ST => SUST, Rate Law: env*(k5*ST*SU-k5r*SUST)
k4 = 0.3 per_timeReaction: STU =>, Rate Law: env*k4*STU
k3 = 2.0 per_time; k3r = 1.0 per_time_per_MReaction: STUST => ST + STU, Rate Law: env*(k3*STUST-k3r*ST*STU)
k9 = 0.1 per_time_per_M; k9r = 0.05 per_timeReaction: U + STUS => STUSU, Rate Law: env*(k9*U*STUS-k9r*STUSU)
k11 = NaN per_timeReaction: ST =>, Rate Law: env*k11*ST
k8 = NaN per_timeReaction: SU =>, Rate Law: env*k8*SU
k7 = 0.1 per_time; k7r = 0.1 per_time_per_MReaction: SUSTU => STU + SU, Rate Law: env*(k7*SUSTU-k7r*STU*SU)

States:

NameDescription
STUSTSTUST
TT
SUSTSUST
SUSU
STST
SS
UU
STUSUSTUSU
SUSTUSUSTU
STUSSTUS
STUSTU

Pinchuck2010 - Genome-scale metabolic network of Shewanella oneidensis (iSO783): MODEL1507180036v0.0.1

Pinchuck2010 - Genome-scale metabolic network of Shewanella oneidensis (iSO783)This model is described in the article:…

Details

Shewanellae are gram-negative facultatively anaerobic metal-reducing bacteria commonly found in chemically (i.e., redox) stratified environments. Occupying such niches requires the ability to rapidly acclimate to changes in electron donor/acceptor type and availability; hence, the ability to compete and thrive in such environments must ultimately be reflected in the organization and utilization of electron transfer networks, as well as central and peripheral carbon metabolism. To understand how Shewanella oneidensis MR-1 utilizes its resources, the metabolic network was reconstructed. The resulting network consists of 774 reactions, 783 genes, and 634 unique metabolites and contains biosynthesis pathways for all cell constituents. Using constraint-based modeling, we investigated aerobic growth of S. oneidensis MR-1 on numerous carbon sources. To achieve this, we (i) used experimental data to formulate a biomass equation and estimate cellular ATP requirements, (ii) developed an approach to identify cycles (such as futile cycles and circulations), (iii) classified how reaction usage affects cellular growth, (iv) predicted cellular biomass yields on different carbon sources and compared model predictions to experimental measurements, and (v) used experimental results to refine metabolic fluxes for growth on lactate. The results revealed that aerobic lactate-grown cells of S. oneidensis MR-1 used less efficient enzymes to couple electron transport to proton motive force generation, and possibly operated at least one futile cycle involving malic enzymes. Several examples are provided whereby model predictions were validated by experimental data, in particular the role of serine hydroxymethyltransferase and glycine cleavage system in the metabolism of one-carbon units, and growth on different sources of carbon and energy. This work illustrates how integration of computational and experimental efforts facilitates the understanding of microbial metabolism at a systems level. link: http://identifiers.org/pubmed/20589080

Pitkanen2014 - Metabolic reconstruction of Ashbya gossypii using CoReCo: MODEL1302010038v0.0.1

Pitkanen2014 - Metabolic reconstruction of Ashbya gossypii using CoReCoThis model was reconstructed with the CoReCo meth…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Aspergillus clavatus using CoReCo: MODEL1302010012v0.0.1

Pitkanen2014 - Metabolic reconstruction of Aspergillus clavatus using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Aspergillus fumigatus using CoReCo: MODEL1302010024v0.0.1

Pitkanen2014 - Metabolic reconstruction of Aspergillus fumigatus using CoReCoThis model was reconstructed with the CoReC…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Aspergillus nidulans using CoReCo: MODEL1302010005v0.0.1

Pitkanen2014 - Metabolic reconstruction of Aspergillus nidulans using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375