SBMLBioModels: L - M

L


Lee2017 - Paracetamol first-pass metabolism PK model: BIOMD0000000947v0.0.1

Authors developed a microfluidic gut-liver co-culture chip that aims to reproduce the first-pass metabolism of oral drug…

Details

Accurate prediction of first-pass metabolism is essential for improving the time and cost efficiency of drug development process. Here, we have developed a microfluidic gut-liver co-culture chip that aims to reproduce the first-pass metabolism of oral drugs. This chip consists of two separate layers for gut (Caco-2) and liver (HepG2) cell lines, where cells can be co-cultured in both 2D and 3D forms. Both cell lines were maintained well in the chip, verified by confocal microscopy and measurement of hepatic enzyme activity. We investigated the PK profile of paracetamol in the chip, and corresponding PK model was constructed, which was used to predict PK profiles for different chip design parameters. Simulation results implied that a larger absorption surface area and a higher metabolic capacity are required to reproduce the in vivo PK profile of paracetamol more accurately. Our study suggests the possibility of reproducing the human PK profile on a chip, contributing to accurate prediction of pharmacological effect of drugs. link: http://identifiers.org/pubmed/29116458

Parameters:

NameDescription
V_basol = 380.0; Mp_g_HepG2 = 0.59; Mp_s_HepG2 = 0.35; P_para = 103.8; Ai = 0.33Reaction: C_para__Basolateral___HepG2_ = ((P_para*Ai*(C_para_Caco_2-C_para__Basolateral___HepG2_)-Mp_s_HepG2*C_para__Basolateral___HepG2_*V_basol)-Mp_g_HepG2*C_para__Basolateral___HepG2_*V_basol)/V_basol, Rate Law: ((P_para*Ai*(C_para_Caco_2-C_para__Basolateral___HepG2_)-Mp_s_HepG2*C_para__Basolateral___HepG2_*V_basol)-Mp_g_HepG2*C_para__Basolateral___HepG2_*V_basol)/V_basol
P_sulf = 49.9; V_basol = 380.0; Mp_s_HepG2 = 0.35; Ai = 0.33Reaction: C_sulf__Basolateral___HepG2_ = (P_sulf*Ai*(C_sulf_Caco_2-C_sulf__Basolateral___HepG2_)+Mp_s_HepG2*C_para__Basolateral___HepG2_*V_basol)/V_basol, Rate Law: (P_sulf*Ai*(C_sulf_Caco_2-C_sulf__Basolateral___HepG2_)+Mp_s_HepG2*C_para__Basolateral___HepG2_*V_basol)/V_basol
P_glu = 58.9; V_basol = 380.0; Mp_g_HepG2 = 0.59; Ai = 0.33Reaction: C_glu__Basolateral___HepG2_ = (P_glu*Ai*(C_glu_Caco_2-C_glu__Basolateral___HepG2_)+Mp_g_HepG2*C_para__Basolateral___HepG2_*V_basol)/V_basol, Rate Law: (P_glu*Ai*(C_glu_Caco_2-C_glu__Basolateral___HepG2_)+Mp_g_HepG2*C_para__Basolateral___HepG2_*V_basol)/V_basol
P_para = 103.8; V_api = 500.0; Ai = 0.33Reaction: C_para_Apical = (-1)*P_para*Ai*(C_para_Apical-C_para_Caco_2)/V_api, Rate Law: (-1)*P_para*Ai*(C_para_Apical-C_para_Caco_2)/V_api
P_sulf = 49.9; Mp_s_caco = 14.9; V_caco = 0.33; Ai = 0.33Reaction: C_sulf_Caco_2 = ((P_sulf*Ai*(C_sulf_Apical-C_sulf_Caco_2)-P_sulf*Ai*(C_sulf_Caco_2-C_sulf__Basolateral___HepG2_))+Mp_s_caco*C_para_Caco_2*V_caco)/V_caco, Rate Law: ((P_sulf*Ai*(C_sulf_Apical-C_sulf_Caco_2)-P_sulf*Ai*(C_sulf_Caco_2-C_sulf__Basolateral___HepG2_))+Mp_s_caco*C_para_Caco_2*V_caco)/V_caco
P_glu = 58.9; Mp_g_caco = 17.6; V_caco = 0.33; Ai = 0.33Reaction: C_glu_Caco_2 = ((P_glu*Ai*(C_glu_Apical-C_glu_Caco_2)-P_glu*Ai*(C_glu_Caco_2-C_glu__Basolateral___HepG2_))+Mp_g_caco*C_para_Caco_2*V_caco)/V_caco, Rate Law: ((P_glu*Ai*(C_glu_Apical-C_glu_Caco_2)-P_glu*Ai*(C_glu_Caco_2-C_glu__Basolateral___HepG2_))+Mp_g_caco*C_para_Caco_2*V_caco)/V_caco
P_glu = 58.9; V_api = 500.0; Ai = 0.33Reaction: C_glu_Apical = (-1)*P_glu*Ai*(C_glu_Apical-C_glu_Caco_2)/V_api, Rate Law: (-1)*P_glu*Ai*(C_glu_Apical-C_glu_Caco_2)/V_api
P_sulf = 49.9; V_api = 500.0; Ai = 0.33Reaction: C_sulf_Apical = (-1)*P_sulf*Ai*(C_sulf_Apical-C_sulf_Caco_2)/V_api, Rate Law: (-1)*P_sulf*Ai*(C_sulf_Apical-C_sulf_Caco_2)/V_api
Mp_s_caco = 14.9; Mp_g_caco = 17.6; V_caco = 0.33; P_para = 103.8; Ai = 0.33Reaction: C_para_Caco_2 = (((P_para*Ai*(C_para_Apical-C_para_Caco_2)-P_para*Ai*(C_para_Caco_2-C_para__Basolateral___HepG2_))-Mp_s_caco*C_para_Caco_2*V_caco)-Mp_g_caco*C_para_Caco_2*V_caco)/V_caco, Rate Law: (((P_para*Ai*(C_para_Apical-C_para_Caco_2)-P_para*Ai*(C_para_Caco_2-C_para__Basolateral___HepG2_))-Mp_s_caco*C_para_Caco_2*V_caco)-Mp_g_caco*C_para_Caco_2*V_caco)/V_caco

States:

NameDescription
C para Caco 2[paracetamol; D00217]
C glu Basolateral HepG2[D00217; paracetamol; Glucuronide]
C sulf Apical[paracetamol sulfate]
C sulf Caco 2[paracetamol sulfate]
C glu Apical[paracetamol; D00217; Glucuronide]
C sulf Basolateral HepG2[paracetamol sulfate]
C para Basolateral HepG2[paracetamol; D00217]
C para Apical[D00217; paracetamol]
C glu Caco 2[Glucuronide; paracetamol; D00217]

Leeuwen2007 - Elucidating the interactions between the adhesive and transcriptional functions of beta-catenin in normal and cancerous cells: MODEL2001090001v0.0.1

Elucidating the interactions between the adhesive and transcriptional functions of beta-catenin in normal and cancerous…

Details

Wnt signalling is involved in a wide range of physiological and pathological processes. The presence of an extracellular Wnt stimulus induces cytoplasmic stabilisation and nuclear translocation of beta-catenin, a protein that also plays an essential role in cadherin-mediated adhesion. Two main hypotheses have been proposed concerning the balance between beta-catenin's adhesive and transcriptional functions: either beta-catenin's fate is determined by competition between its binding partners, or Wnt induces folding of beta-catenin into a conformation allocated preferentially to transcription. The experimental data supporting each hypotheses remain inconclusive. In this paper we present a new mathematical model of the Wnt pathway that incorporates beta-catenin's dual function. We use this model to carry out a series of in silico experiments and compare the behaviour of systems governed by each hypothesis. Our analytical results and model simulations provide further insight into the current understanding of Wnt signalling and, in particular, reveal differences in the response of the two modes of interaction between adhesion and signalling in certain in silico settings. We also exploit our model to investigate the impact of the mutations most commonly observed in human colorectal cancer. Simulations show that the amount of functional APC required to maintain a normal phenotype increases with increasing strength of the Wnt signal, a result which illustrates that the environment can substantially influence both tumour initiation and phenotype. link: http://identifiers.org/pubmed/17382967

Legewie2006_apoptosis_NC: BIOMD0000000103v0.0.1

This model represents the non-competitive binding of XIAP to Casapase-3 and Caspase-9. In other words, XIAP mediated fee…

Details

The intrinsic, or mitochondrial, pathway of caspase activation is essential for apoptosis induction by various stimuli including cytotoxic stress. It depends on the cellular context, whether cytochrome c released from mitochondria induces caspase activation gradually or in an all-or-none fashion, and whether caspase activation irreversibly commits cells to apoptosis. By analyzing a quantitative kinetic model, we show that inhibition of caspase-3 (Casp3) and Casp9 by inhibitors of apoptosis (IAPs) results in an implicit positive feedback, since cleaved Casp3 augments its own activation by sequestering IAPs away from Casp9. We demonstrate that this positive feedback brings about bistability (i.e., all-or-none behaviour), and that it cooperates with Casp3-mediated feedback cleavage of Casp9 to generate irreversibility in caspase activation. Our calculations also unravel how cell-specific protein expression brings about the observed qualitative differences in caspase activation (gradual versus all-or-none and reversible versus irreversible). Finally, known regulators of the pathway are shown to efficiently shift the apoptotic threshold stimulus, suggesting that the bistable caspase cascade computes multiple inputs into an all-or-none caspase output. As cellular inhibitory proteins (e.g., IAPs) frequently inhibit consecutive intermediates in cellular signaling cascades (e.g., Casp3 and Casp9), the feedback mechanism described in this paper is likely to be a widespread principle on how cells achieve ultrasensitivity, bistability, and irreversibility. link: http://identifiers.org/pubmed/16978046

Parameters:

NameDescription
k6 = 5.0E-5Reaction: C3 + C9_star => C3_star + C9_star, Rate Law: cytosol*k6*C3*C9_star
d = 1.0; a = 1.0; k1 = 0.002; kb1 = 0.1Reaction: C9X_C3_star + A => AC9X_C3_star, Rate Law: cytosol*(a*k1*C9X_C3_star*A-d*kb1*AC9X_C3_star)
d = 1.0; a = 1.0; k15 = 0.003; k15b = 0.001Reaction: C3_star + C9_starX => C9_starX_C3_star, Rate Law: cytosol*(a*k15*C3_star*C9_starX-d*k15b*C9_starX_C3_star)
k17 = 0.001; k17prod = 0.02Reaction: => C9, Rate Law: cytosol*(k17prod-k17*C9)
k20 = 0.001Reaction: AC9X =>, Rate Law: cytosol*k20*AC9X
k1 = 0.002; kb1 = 0.1Reaction: A + C9 => AC9, Rate Law: cytosol*(k1*A*C9-kb1*AC9)
k3 = 3.5E-4Reaction: C3 + AC9 => C3_star + AC9, Rate Law: cytosol*k3*C3*AC9
k28 = 0.001Reaction: AC9_starX =>, Rate Law: cytosol*k28*AC9_starX
k31 = 0.001Reaction: C9_starX_C3_star =>, Rate Law: cytosol*k31*C9_starX_C3_star
k9 = 0.001; k9b = 0.001Reaction: C9 + X => C9X, Rate Law: cytosol*(k9*C9*X-k9b*C9X)
d = 1.0; a = 1.0; k9 = 0.001; k9b = 0.001Reaction: C9 + C3_starX => C9X_C3_star, Rate Law: cytosol*(a*k9*C9*C3_starX-d*k9b*C9X_C3_star)
k25 = 0.001Reaction: C9_starX =>, Rate Law: cytosol*k25*C9_starX
k22prod = 0.2; k22 = 0.001Reaction: => C3, Rate Law: cytosol*(k22prod-k22*C3)
k14b = 0.1; k14 = 0.002Reaction: C9_starX + A => AC9_starX, Rate Law: cytosol*(k14*C9_starX*A-k14b*AC9_starX)
k29 = 0.001Reaction: C9X_C3_star =>, Rate Law: cytosol*k29*C9X_C3_star
k8b = 0.1; k8 = 0.002Reaction: C9_star + A => AC9_star, Rate Law: cytosol*(k8*C9_star*A-k8b*AC9_star)
k21 = 0.001Reaction: AC9 =>, Rate Law: cytosol*k21*AC9
k16prod = 0.02; k16 = 0.001Reaction: => A, Rate Law: cytosol*(k16prod-k16*A)
k13 = 0.002; k13b = 0.1Reaction: C9X + A => AC9X, Rate Law: cytosol*(k13*C9X*A-k13b*AC9X)
k5 = 2.0E-4Reaction: AC9 + C3_star => AC9_star + C3_star, Rate Law: cytosol*k5*AC9*C3_star
k2 = 5.0E-6Reaction: C3 + C9 => C3_star + C9, Rate Law: cytosol*k2*C3*C9
k4 = 2.0E-4Reaction: C9 + C3_star => C9_star + C3_star, Rate Law: cytosol*k4*C9*C3_star
k23 = 0.001Reaction: C3_star =>, Rate Law: cytosol*k23*C3_star
k19 = 0.001Reaction: C9X =>, Rate Law: cytosol*k19*C9X
k12b = 0.001; k12 = 0.001Reaction: AC9_star + X => AC9_starX, Rate Law: cytosol*(k12*AC9_star*X-k12b*AC9_starX)
k18 = 0.001; k18prod = 0.04Reaction: => X, Rate Law: cytosol*(k18prod-k18*X)
k10 = 0.001; k10b = 0.001Reaction: AC9 + X => AC9X, Rate Law: cytosol*(k10*AC9*X-k10b*AC9X)
k26 = 0.001Reaction: C9_star =>, Rate Law: cytosol*k26*C9_star
k30 = 0.001Reaction: AC9_starX_C3_star =>, Rate Law: cytosol*k30*AC9_starX_C3_star
k11b = 0.001; k11 = 0.001Reaction: C9_star + X => C9_starX, Rate Law: cytosol*(k11*C9_star*X-k11b*C9_starX)
k32 = 0.001Reaction: AC9_starX_C3_star =>, Rate Law: cytosol*k32*AC9_starX_C3_star
k27 = 0.001Reaction: AC9_star =>, Rate Law: cytosol*k27*AC9_star
k15 = 0.003; k15b = 0.001Reaction: C3_star + X => C3_starX, Rate Law: cytosol*(k15*C3_star*X-k15b*C3_starX)
k24 = 0.001Reaction: C3_starX =>, Rate Law: cytosol*k24*C3_starX
k7 = 0.0035Reaction: C3 + AC9_star => C3_star + AC9_star, Rate Law: cytosol*k7*C3*AC9_star

States:

NameDescription
C3 star[Caspase-3]
A[Apoptotic protease-activating factor 1]
C3[Caspase-3]
X[E3 ubiquitin-protein ligase XIAP]
AC9 star[Caspase-9; Apoptotic protease-activating factor 1]
C9 starX[E3 ubiquitin-protein ligase XIAP; Caspase-9]
C9X[E3 ubiquitin-protein ligase XIAP; Caspase-9]
AC9X C3 star[E3 ubiquitin-protein ligase XIAP; Caspase-9; Apoptotic protease-activating factor 1]
AC9X[E3 ubiquitin-protein ligase XIAP; Caspase-9; Apoptotic protease-activating factor 1]
C9 starX C3 star[Caspase-3; E3 ubiquitin-protein ligase XIAP; Caspase-9]
C9[Caspase-9]
C9X C3 star[E3 ubiquitin-protein ligase XIAP; Caspase-9]
C9 star[Caspase-9]
AC9 starX[E3 ubiquitin-protein ligase XIAP; Caspase-9; Apoptotic protease-activating factor 1]
C3 starX[E3 ubiquitin-protein ligase XIAP; Caspase-3]
AC9 starX C3 star[Caspase-3; Caspase-9; E3 ubiquitin-protein ligase XIAP; Apoptotic protease-activating factor 1]
AC9[Caspase-9; Apoptotic protease-activating factor 1]

Legewie2006_apoptosis_WT: BIOMD0000000102v0.0.1

The model reproduces active Caspase-3 time profile corresponding to the total Apaf-1 value of 20 nM as depicted in Fig 2…

Details

The intrinsic, or mitochondrial, pathway of caspase activation is essential for apoptosis induction by various stimuli including cytotoxic stress. It depends on the cellular context, whether cytochrome c released from mitochondria induces caspase activation gradually or in an all-or-none fashion, and whether caspase activation irreversibly commits cells to apoptosis. By analyzing a quantitative kinetic model, we show that inhibition of caspase-3 (Casp3) and Casp9 by inhibitors of apoptosis (IAPs) results in an implicit positive feedback, since cleaved Casp3 augments its own activation by sequestering IAPs away from Casp9. We demonstrate that this positive feedback brings about bistability (i.e., all-or-none behaviour), and that it cooperates with Casp3-mediated feedback cleavage of Casp9 to generate irreversibility in caspase activation. Our calculations also unravel how cell-specific protein expression brings about the observed qualitative differences in caspase activation (gradual versus all-or-none and reversible versus irreversible). Finally, known regulators of the pathway are shown to efficiently shift the apoptotic threshold stimulus, suggesting that the bistable caspase cascade computes multiple inputs into an all-or-none caspase output. As cellular inhibitory proteins (e.g., IAPs) frequently inhibit consecutive intermediates in cellular signaling cascades (e.g., Casp3 and Casp9), the feedback mechanism described in this paper is likely to be a widespread principle on how cells achieve ultrasensitivity, bistability, and irreversibility. link: http://identifiers.org/pubmed/16978046

Parameters:

NameDescription
k17 = 0.001 sec_inverse; k17prod = 0.02 nM_per_secReaction: => C9, Rate Law: cytosol*(k17prod-k17*C9)
k14 = 0.002 per_nM_per_sec; k14b = 0.1 sec_inverseReaction: C9_starX + A => AC9_starX, Rate Law: cytosol*(k14*C9_starX*A-k14b*AC9_starX)
k26 = 0.001 sec_inverseReaction: C9_star =>, Rate Law: cytosol*k26*C9_star
k19 = 0.001 sec_inverseReaction: C9X =>, Rate Law: cytosol*k19*C9X
k8b = 0.1 sec_inverse; k8 = 0.002 per_nM_per_secReaction: C9_star + A => AC9_star, Rate Law: cytosol*(k8*C9_star*A-k8b*AC9_star)
k1 = 0.002 per_nM_per_sec; kb1 = 0.1 sec_inverseReaction: A + C9 => AC9, Rate Law: cytosol*(k1*A*C9-kb1*AC9)
k12 = 0.001 per_nM_per_sec; k12b = 0.001 sec_inverseReaction: AC9_star + X => AC9_starX, Rate Law: cytosol*(k12*AC9_star*X-k12b*AC9_starX)
k2 = 5.0E-6 per_nM_per_secReaction: C3 + C9 => C3_star + C9, Rate Law: cytosol*k2*C3*C9
k13 = 0.002 per_nM_per_sec; k13b = 0.1 sec_inverseReaction: C9X + A => AC9X, Rate Law: cytosol*(k13*C9X*A-k13b*AC9X)
k6 = 5.0E-5 per_nM_per_secReaction: C3 + C9_star => C3_star + C9_star, Rate Law: cytosol*k6*C3*C9_star
k18prod = 0.04 nM_per_sec; k18 = 0.001 sec_inverseReaction: => X, Rate Law: cytosol*(k18prod-k18*X)
k7 = 0.0035 per_nM_per_secReaction: C3 + AC9_star => C3_star + AC9_star, Rate Law: cytosol*k7*C3*AC9_star
k4 = 2.0E-4 per_nM_per_secReaction: C9 + C3_star => C9_star + C3_star, Rate Law: cytosol*k4*C9*C3_star
k16prod = 0.02 nM_per_sec; k16 = 0.001 sec_inverseReaction: => A, Rate Law: cytosol*(k16prod-k16*A)
k5 = 2.0E-4 per_nM_per_secReaction: AC9 + C3_star => AC9_star + C3_star, Rate Law: cytosol*k5*AC9*C3_star
k24 = 0.001 sec_inverseReaction: C3_starX =>, Rate Law: cytosol*k24*C3_starX
k21 = 0.001 sec_inverseReaction: AC9 =>, Rate Law: cytosol*k21*AC9
k9b = 0.001 sec_inverse; k9 = 0.001 per_nM_per_secReaction: C9 + X => C9X, Rate Law: cytosol*(k9*C9*X-k9b*C9X)
k27 = 0.001 sec_inverseReaction: AC9_star =>, Rate Law: cytosol*k27*AC9_star
k20 = 0.001 sec_inverseReaction: AC9X =>, Rate Law: cytosol*k20*AC9X
k3 = 3.5E-4 per_nM_per_secReaction: C3 + AC9 => C3_star + AC9, Rate Law: cytosol*k3*C3*AC9
k28 = 0.001 sec_inverseReaction: AC9_starX =>, Rate Law: cytosol*k28*AC9_starX
k23 = 0.001 sec_inverseReaction: C3_star =>, Rate Law: cytosol*k23*C3_star
k22prod = 0.2 nM_per_sec; k22 = 0.001 sec_inverseReaction: => C3, Rate Law: cytosol*(k22prod-k22*C3)
k15b = 0.001 sec_inverse; k15 = 0.003 per_nM_per_secReaction: C3_star + X => C3_starX, Rate Law: cytosol*(k15*C3_star*X-k15b*C3_starX)
k10 = 0.001 per_nM_per_sec; k10b = 0.001 sec_inverseReaction: AC9 + X => AC9X, Rate Law: cytosol*(k10*AC9*X-k10b*AC9X)
k25 = 0.001 sec_inverseReaction: C9_starX =>, Rate Law: cytosol*k25*C9_starX
k11b = 0.001 sec_inverse; k11 = 0.001 per_nM_per_secReaction: C9_star + X => C9_starX, Rate Law: cytosol*(k11*C9_star*X-k11b*C9_starX)

States:

NameDescription
C3 star[Caspase-3]
A[Apoptotic protease-activating factor 1]
C3[Caspase-3]
X[E3 ubiquitin-protein ligase XIAP]
AC9 star[Apoptotic protease-activating factor 1; Caspase-9]
C9 starX[Caspase-9; E3 ubiquitin-protein ligase XIAP]
C9X[Caspase-9; E3 ubiquitin-protein ligase XIAP]
AC9X[Apoptotic protease-activating factor 1; Caspase-9; E3 ubiquitin-protein ligase XIAP]
C9[Caspase-9]
C9 star[Caspase-9]
AC9 starX[Apoptotic protease-activating factor 1; Caspase-9; E3 ubiquitin-protein ligase XIAP]
C3 starX[Caspase-3; E3 ubiquitin-protein ligase XIAP]
AC9[Apoptotic protease-activating factor 1; Caspase-9]

Lei2001_Yeast_Aerobic_Metabolism: BIOMD0000000245v0.0.1

This the model from the article: A biochemically structured model for Saccharomyces cerevisiae. Lei F, Rotbøll M, Jø…

Details

A biochemically structured model for the aerobic growth of Saccharomyces cerevisiae on glucose and ethanol is presented. The model focuses on the pyruvate and acetaldehyde branch points where overflow metabolism occurs when the growth changes from oxidative to oxido-reductive. The model is designed to describe the onset of aerobic alcoholic fermentation during steady-state as well as under dynamical conditions, by triggering an increase in the glycolytic flux using a key signalling component which is assumed to be closely related to acetaldehyde. An investigation of the modelled process dynamics in a continuous cultivation revealed multiple steady states in a region of dilution rates around the transition between oxidative and oxido-reductive growth. A bifurcation analysis using the two external variables, the dilution rate, D, and the inlet concentration of glucose, S(f), as parameters, showed that a fold bifurcation occurs close to the critical dilution rate resulting in multiple steady-states. The region of dilution rates within which multiple steady states may occur depends strongly on the substrate feed concentration. Consequently a single steady state may prevail at low feed concentrations, whereas multiple steady states may occur over a relatively wide range of dilution rates at higher feed concentrations. link: http://identifiers.org/pubmed/11434967

Parameters:

NameDescription
X_AcDH = 0.0075 dimensionless; k_11 = 0.02 gram per liter per hourReaction: AcDH => ; x, Rate Law: k_11*X_AcDH*x*env
X_AcDH = 0.0075 dimensionlessReaction: AcDH = x*X_AcDH, Rate Law: missing
D = 0.1 per hourReaction: s_pyr =>, Rate Law: s_pyr*D*env
K_9e = 13.0 gram per liter; k_9c = 0.00399 gram per liter per hour; k_9 = 0.008 gram per liter per hour; k_9e = 0.0751 gram per liter per hour; K_9 = 1.0E-6 gram per liter; K_9i = 25.0 liter per gram; X_a = 0.1 dimensionlessReaction: a => AcDH; x, s_glu, s_EtOH, Rate Law: ((k_9*s_glu/(s_glu+K_9)+k_9e*s_EtOH/(s_EtOH+K_9e))/(K_9i*s_glu+1)+k_9c*s_glu/(s_glu+K_9))*X_a*x*env
k_1e = 47.1 gram per liter per hour; K_1e = 0.12 gram per liter; K_1l = 0.94 gram per liter; k_1h = 0.584 gram per liter per hour; X_a = 0.1 dimensionless; k_1l = 1.43 gram per liter per hour; K_1h = 0.0116 gram per liter; K_1i = 14.2 liter per gramReaction: s_glu => s_pyr + Red; s_acetald, x, Rate Law: (k_1l*s_glu/(s_glu+K_1l)+k_1h*s_glu/(s_glu+K_1h)+k_1e*s_acetald*s_glu/(s_glu*(K_1i*s_acetald+1)+K_1e))*x*X_a*env
K_5e = 0.1 gram per liter; K_5i = 440.0 liter per gram; k_8 = 0.589 gram per liter per hour; X_a = 0.1 dimensionlessReaction: s_acetate => x + CO2 + Red; x, s_glu, Rate Law: k_8*s_acetate/((s_acetate+K_5e)*(1+K_5i*s_glu))*x*X_a*env
K_5e = 0.1 gram per liter; K_5 = 0.0102 gram per liter; K_5i = 440.0 liter per gram; k_5e = 0.775 gram per liter per hour; k_5 = 0.0104 gram per liter per hour; X_a = 0.1 dimensionlessReaction: s_acetate => CO2 + Red; x, s_glu, Rate Law: (k_5*s_acetate/(s_acetate+K_5)+k_5e*s_acetate/((s_acetate+K_5e)*(1+K_5i*s_glu)))*x*X_a*env
K_10 = 0.0023 gram per liter; k_10 = 0.392 gram per liter per hour; K_10e = 0.0018 gram per liter; k_10e = 0.00339 gram per liter per hour; X_a = 0.1 dimensionlessReaction: a => ; x, s_glu, s_EtOH, Rate Law: (k_10*s_glu/(s_glu+K_10)+k_10e*s_EtOH/(s_EtOH+K_10e))*X_a*x*env
K_6e = 0.057 gram per liter; k_6r = 0.0125 dimensionless; k_6 = 2.82 gram per liter per hour; K_6 = 0.034 gram per liter; X_a = 0.1 dimensionlessReaction: s_acetald + Red => s_EtOH; x, Rate Law: k_6*(s_acetald-k_6r*s_EtOH)/(s_acetald+K_6+K_6e*s_EtOH)*x*X_a*env
X_a = 0.1 dimensionlessReaction: a = x*X_a, Rate Law: missing
K_3 = 5.0E-7 gram per liter; k_3 = 5.81 gram per liter per hour; X_a = 0.1 dimensionlessReaction: s_pyr => s_acetald + CO2; x, Rate Law: k_3*s_pyr^4/(s_pyr^4+K_3)*x*X_a*env
k_2 = 0.501 gram per liter per hour; K_2i = 0.101 liter per gram; K_2 = 2.0E-5 gram per liter; X_a = 0.1 dimensionlessReaction: s_pyr => CO2 + Red; x, s_glu, Rate Law: k_2*s_pyr/((s_pyr+K_2)*(K_2i*s_glu+1))*x*X_a*env
k_7 = 1.203 gram per liter per hour; K_7 = 0.0101 gram per liter; X_a = 0.1 dimensionlessReaction: s_glu => x + CO2 + Red; x, Rate Law: k_7*s_glu/(s_glu+K_7)*x*X_a*env
X_AcDH = 0.0075 dimensionless; K_4 = 2.64E-4 gram per liter; k_4 = 4.8 gram per liter per hour; X_a = 0.1 dimensionlessReaction: s_acetald => s_acetate + Red; x, s_EtOH, Rate Law: k_4*s_acetald/(s_acetald+K_4)*x*X_a*X_AcDH*env

States:

NameDescription
s acetald[acetaldehyde; Acetaldehyde]
Red[NADH; NADH]
xBM
CO2[carbon dioxide; CO2]
aBM(active)
S f[glucose; C00293]
s EtOH[ethanol; Ethanol]
s glu[glucose; C00293]
s pyr[pyruvate; Pyruvate]
AcDHBM(AcDH)
s acetate[CHEBI_40480; Acetate]

Leipold1995_ThrombinFormation+inhibitors: MODEL1109150002v0.0.1

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

Details

A mathematical model has been developed to simulate the generation of thrombin by the tissue factor pathway. The model gives reasonable predictions of published experimental results without the adjustment of any parameter values. The model also accounts explicitly for the effects of serine protease inhibitors on thrombin generation. Simulations to define the optimum affinity profile of an inhibitor in this system indicate that for an inhibitor simultaneously potent against VIIa, IXa, and Xa, inhibition of thrombin generation decreases dramatically as the affinity for thrombin increases. Additional simulations show that the reason for this behavior is the sequestration of the inhibitor by small amounts of thrombin generated early in the reaction. This model is also useful for predicting the potency of compounds that inhibit thrombosis in rats. We believe that this is the first mathematical model of blood coagulation that considers the effects of exogenous inhibitors. Such a model, or extensions thereof, should be useful for evaluating targets for therapeutic intervention in the processes of blood coagulation. link: http://identifiers.org/pubmed/7592704

Leipold1995_ThrombinFormation-inhibitors: MODEL1109150001v0.0.1

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

Details

A mathematical model has been developed to simulate the generation of thrombin by the tissue factor pathway. The model gives reasonable predictions of published experimental results without the adjustment of any parameter values. The model also accounts explicitly for the effects of serine protease inhibitors on thrombin generation. Simulations to define the optimum affinity profile of an inhibitor in this system indicate that for an inhibitor simultaneously potent against VIIa, IXa, and Xa, inhibition of thrombin generation decreases dramatically as the affinity for thrombin increases. Additional simulations show that the reason for this behavior is the sequestration of the inhibitor by small amounts of thrombin generated early in the reaction. This model is also useful for predicting the potency of compounds that inhibit thrombosis in rats. We believe that this is the first mathematical model of blood coagulation that considers the effects of exogenous inhibitors. Such a model, or extensions thereof, should be useful for evaluating targets for therapeutic intervention in the processes of blood coagulation. link: http://identifiers.org/pubmed/7592704

Leloup1998_CircClock_LD: BIOMD0000000171v0.0.1

# Leloup and Goldbeter, 1998 This model was created after the article by Leloup and Goldbeter, *J Biol Rhythms* 1998,…

Details

The authors present a model for circadian oscillations of the Period (PER) and Timeless (TIM) proteins in Drosophila. The model for the circadian clock is based on multiple phosphorylation of PER and TIM and on the negative feedback exerted by a nuclear PER-TIM complex on the transcription of the per and tim genes. Periodic behavior occurs in a large domain of parameter space in the form of limit cycle oscillations. These sustained oscillations occur in conditions corresponding to continuous darkness or to entrainment by light-dark cycles and are in good agreement with experimental observations on the temporal variations of PER and TIM and of per and tim mRNAs. Birhythmicity (coexistence of two periodic regimes) and aperiodic oscillations (chaos) occur in a restricted range of parameter values. The results are compared to the predictions of a model based on the sole regulation by PER. Both the formation of a complex between PER and TIM and protein phosphorylation are found to favor oscillatory behavior. Determining how the period depends on several key parameters allows us to test possible molecular explanations proposed for the altered period in the per(l) and per(s) mutants. The extended model further allows the construction of phase-response curves based on the light-induced triggering of TIM degradation. These curves, established as a function of both the duration and magnitude of the effect of a light pulse, match the phase-response curves obtained experimentally in the wild type and per(s) mutant of Drosophila. link: http://identifiers.org/pubmed/9486845

Parameters:

NameDescription
K_2T=2.0 nanomoleperlitre; V_2T=1.0 nanoMperHourReaction: T1 => T0, Rate Law: V_2T*T1/(K_2T+T1)*cytoplasm
k_sP=0.9 perhourReaction: => P0; M_P, Rate Law: k_sP*M_P*cytoplasm
K_4P=2.0 nanomoleperlitre; V_4P=1.0 nanoMperHourReaction: P2 => P1, Rate Law: V_4P*P2/(K_4P+P2)*cytoplasm
kd_C=0.01 perhourReaction: C =>, Rate Law: kd_C*C*cytoplasm
K_4T=2.0 nanomoleperlitre; V_4T=1.0 nanoMperHourReaction: T2 => T1, Rate Law: V_4T*T2/(K_4T+T2)*cytoplasm
v_mT=0.7 nanoMperHour; K_mT=0.2 nanomoleperlitre; kd = 0.01 perhourReaction: M_T =>, Rate Law: (v_mT/(K_mT+M_T)+kd)*M_T*cytoplasm
v_dP=2.0 nanoMperHour; K_dP=0.2 nanomoleperlitreReaction: P2 =>, Rate Law: v_dP*P2/(K_dP+P2)*cytoplasm
K_mP=0.2 nanomoleperlitre; v_mP=0.8 nanoMperHour; kd = 0.01 perhourReaction: M_P =>, Rate Law: (v_mP/(K_mP+M_P)+kd)*M_P*cytoplasm
K_3P=2.0 nanomoleperlitre; V_3P=8.0 nanoMperHourReaction: P1 => P2, Rate Law: V_3P*P1/(K_3P+P1)*cytoplasm
kd = 0.01 perhourReaction: P1 =>, Rate Law: kd*P1*cytoplasm
k_sT=0.9 perhourReaction: => T0; M_T, Rate Law: k_sT*M_T*cytoplasm
V_1P=8.0 nanoMperHour; K_1P=2.0 nanomoleperlitreReaction: P0 => P1, Rate Law: V_1P*P0/(K_1P+P0)*cytoplasm
K_dT=0.2 nanomoleperlitre; v_dT = 2.0 nanoMperHourReaction: T2 =>, Rate Law: v_dT*T2/(K_dT+T2)*cytoplasm
Ki_P=1.0 nanomoleperlitre; v_sP=0.8 nanomolperhour; n = 4.0 dimensionlessReaction: => M_P; CN, Rate Law: v_sP*Ki_P^n/(Ki_P^n+CN^n)
K_2P=2.0 nanomoleperlitre; V_2P=1.0 nanoMperHourReaction: P1 => P0, Rate Law: V_2P*P1/(K_2P+P1)*cytoplasm
kd_CN=0.01 perhourReaction: CN =>, Rate Law: kd_CN*CN*nucleus
v_sT=1.0 nanomolperhour; n = 4.0 dimensionless; Ki_T=1.0 nanomoleperlitreReaction: => M_T; CN, Rate Law: v_sT*Ki_T^n/(Ki_T^n+CN^n)
k3=1.2 pernMperHour; k4=0.6 perhourReaction: P2 + T2 => C, Rate Law: (k3*T2*P2-k4*C)*cytoplasm
K_1T=2.0 nanomoleperlitre; V_1T=8.0 nanoMperHourReaction: T0 => T1, Rate Law: V_1T*T0/(K_1T+T0)*cytoplasm
k1=1.2 perhour; k2=0.2 perhourReaction: C => CN, Rate Law: k1*C*cytoplasm-k2*CN*nucleus
V_3T=8.0 nanoMperHour; K_3T=2.0 nanomoleperlitreReaction: T1 => T2, Rate Law: V_3T*T1/(K_3T+T1)*cytoplasm

States:

NameDescription
M P[messenger RNA; (5')ppPur-mRNA]
T0[Protein timeless]
C[Period circadian protein; Protein timeless; protein complex]
Pt[Period circadian protein]
P2[Period circadian protein; Phosphoprotein]
P1[Period circadian protein; Phosphoprotein]
M T[messenger RNA; (5')ppPur-mRNA]
P0[Period circadian protein]
CN[Period circadian protein; Protein timeless; protein complex]
Tt[Protein timeless]
T1[Protein timeless; Phosphoprotein]
T2[Protein timeless]

Leloup1999_CircadianRhythms_Drosophila: BIOMD0000000298v0.0.1

This a model from the article: Limit cycle models for circadian rhythms based on transcriptional regulation in Droso…

Details

We examine theoretical models for circadian oscillations based on transcriptional regulation in Drosophila and Neurospora. For Drosophila, the molecular model is based on the negative feedback exerted on the expression of the per and tim genes by the complex formed between the PER and TIM proteins. For Neurospora, similarly, the model relies on the feedback exerted on the expression of the frq gene by its protein product FRQ. In both models, sustained rhythmic variations in protein and mRNA levels occur in continuous darkness, in the form of limit cycle oscillations. The effect of light on circadian rhythms is taken into account in the models by considering that it triggers degradation of the TIM protein in Drosophila, and frq transcription in Neurospora. When incorporating the control exerted by light at the molecular level, we show that the models can account for the entrainment of circadian rhythms by light-dark cycles and for the damping of the oscillations in constant light, though such damping occurs more readily in the Drosophila model. The models account for the phase shifts induced by light pulses and allow the construction of phase response curves. These compare well with experimental results obtained in Drosophila. The model for Drosophila shows that when applied at the appropriate phase, light pulses of appropriate duration and magnitude can permanently or transiently suppress circadian rhythmicity. We investigate the effects of the magnitude of light-induced changes on oscillatory behavior. Finally, we discuss the common and distinctive features of circadian oscillations in the two organisms. link: http://identifiers.org/pubmed/10643740

Parameters:

NameDescription
V2T = 1.0; kd = 0.01; V3T = 8.0; K3T = 2.0; K2T = 2.0; K4T = 2.0; V1T = 8.0; K1T = 2.0; V4T = 1.0Reaction: T1 = (V1T*T0/(K1T+T0)+V4T*T2/(K4T+T2))-(V2T*T1/(K2T+T1)+V3T*T1/(K3T+T1)+kd*T1), Rate Law: (V1T*T0/(K1T+T0)+V4T*T2/(K4T+T2))-(V2T*T1/(K2T+T1)+V3T*T1/(K3T+T1)+kd*T1)
kd = 0.01; V3T = 8.0; K3T = 2.0; K4T = 2.0; k4 = 0.6; vdT = 3.0; k3 = 1.2; V4T = 1.0; KdT = 0.2Reaction: T2 = (V3T*T1/(K3T+T1)+k4*C)-(V4T*T2/(K4T+T2)+k3*P2*T2+vdT*T2/(KdT+T2)+kd*T2), Rate Law: (V3T*T1/(K3T+T1)+k4*C)-(V4T*T2/(K4T+T2)+k3*P2*T2+vdT*T2/(KdT+T2)+kd*T2)
V2T = 1.0; kd = 0.01; ksT = 0.9; K2T = 2.0; V1T = 8.0; K1T = 2.0Reaction: T0 = (ksT*MT+V2T*T1/(K2T+T1))-(V1T*T0/(K1T+T0)+kd*T0), Rate Law: (ksT*MT+V2T*T1/(K2T+T1))-(V1T*T0/(K1T+T0)+kd*T0)
kd = 0.01; n = 4.0; KIP = 1.0; vmP = 1.0; KmP = 0.2; vsP = 1.1Reaction: MP = vsP*KIP^n/(KIP^n+CN^n)-(vmP*MP/(KmP+MP)+kd*MP), Rate Law: vsP*KIP^n/(KIP^n+CN^n)-(vmP*MP/(KmP+MP)+kd*MP)
k2 = 0.2; k1 = 0.8; kdN = 0.01Reaction: CN = k1*C-(k2*CN+kdN*CN), Rate Law: k1*C-(k2*CN+kdN*CN)
kd = 0.01; n = 4.0; vsT = 1.0; KIT = 1.0; KmT = 0.2; vmT = 0.7Reaction: MT = vsT*KIT^n/(KIT^n+CN^n)-(vmT*MT/(KmT+MT)+kd*MT), Rate Law: vsT*KIT^n/(KIT^n+CN^n)-(vmT*MT/(KmT+MT)+kd*MT)
kd = 0.01; V1P = 8.0; V2P = 1.0; K2P = 2.0; K1P = 2.0; ksP = 0.9Reaction: P0 = (ksP*MP+V2P*P1/(K2P+P1))-(V1P*P0/(K1P+P0)+kd*P0), Rate Law: (ksP*MP+V2P*P1/(K2P+P1))-(V1P*P0/(K1P+P0)+kd*P0)
V4P = 1.0; kd = 0.01; V1P = 8.0; V2P = 1.0; K1P = 2.0; K2P = 2.0; K3P = 2.0; K4P = 2.0; V3P = 8.0Reaction: P1 = (V1P*P0/(K1P+P0)+V4P*P2/(K4P+P2))-(V2P*P1/(K2P+P1)+V3P*P1/(K3P+P1)+kd*P1), Rate Law: (V1P*P0/(K1P+P0)+V4P*P2/(K4P+P2))-(V2P*P1/(K2P+P1)+V3P*P1/(K3P+P1)+kd*P1)
k2 = 0.2; k1 = 0.8; k4 = 0.6; k3 = 1.2; kdC = 0.01Reaction: C = (k3*P2*T2+k2*CN)-(k4*C+k1*C+kdC*C), Rate Law: (k3*P2*T2+k2*CN)-(k4*C+k1*C+kdC*C)
V4P = 1.0; kd = 0.01; KdP = 0.2; K3P = 2.0; k4 = 0.6; K4P = 2.0; V3P = 8.0; k3 = 1.2; vdP = 2.2Reaction: P2 = (V3P*P1/(K3P+P1)+k4*C)-(V4P*P2/(K4P+P2)+k3*P2*T2+vdP*P2/(KdP+P2)+kd*P2), Rate Law: (V3P*P1/(K3P+P1)+k4*C)-(V4P*P2/(K4P+P2)+k3*P2*T2+vdP*P2/(KdP+P2)+kd*P2)

States:

NameDescription
CN[Period circadian protein; Protein timeless]
MP[Period circadian protein; messenger RNA; (5')ppPur-mRNA]
T0[Protein timeless]
C[Protein timeless; Period circadian protein]
T1[Protein timeless; Phosphoprotein]
T2[Protein timeless; Phosphoprotein]
P2[Period circadian protein; Phosphoprotein]
P1[Period circadian protein; Phosphoprotein]
P0[Period circadian protein]
MT[Protein timeless; messenger RNA; (5')ppPur-mRNA]

Leloup1999_CircadianRhythms_Neurospora: BIOMD0000000299v0.0.1

This a model from the article: Limit cycle models for circadian rhythms based on transcriptional regulation in Drosoph…

Details

We examine theoretical models for circadian oscillations based on transcriptional regulation in Drosophila and Neurospora. For Drosophila, the molecular model is based on the negative feedback exerted on the expression of the per and tim genes by the complex formed between the PER and TIM proteins. For Neurospora, similarly, the model relies on the feedback exerted on the expression of the frq gene by its protein product FRQ. In both models, sustained rhythmic variations in protein and mRNA levels occur in continuous darkness, in the form of limit cycle oscillations. The effect of light on circadian rhythms is taken into account in the models by considering that it triggers degradation of the TIM protein in Drosophila, and frq transcription in Neurospora. When incorporating the control exerted by light at the molecular level, we show that the models can account for the entrainment of circadian rhythms by light-dark cycles and for the damping of the oscillations in constant light, though such damping occurs more readily in the Drosophila model. The models account for the phase shifts induced by light pulses and allow the construction of phase response curves. These compare well with experimental results obtained in Drosophila. The model for Drosophila shows that when applied at the appropriate phase, light pulses of appropriate duration and magnitude can permanently or transiently suppress circadian rhythmicity. We investigate the effects of the magnitude of light-induced changes on oscillatory behavior. Finally, we discuss the common and distinctive features of circadian oscillations in the two organisms. link: http://identifiers.org/pubmed/10643740

Parameters:

NameDescription
n = 4.0; Km = 0.5; vm = 0.505; KI = 1.0; vs = 1.6Reaction: M = vs*KI^n/(KI^n+FN^n)-vm*M/(Km+M), Rate Law: vs*KI^n/(KI^n+FN^n)-vm*M/(Km+M)
Kd = 0.13; k2 = 0.6; ks = 0.5; vd = 1.4; k1 = 0.5Reaction: FC = (ks*M+k2*FN)-(vd*FC/(Kd+FC)+k1*FC), Rate Law: (ks*M+k2*FN)-(vd*FC/(Kd+FC)+k1*FC)
k2 = 0.6; k1 = 0.5Reaction: FN = k1*FC-k2*FN, Rate Law: k1*FC-k2*FN

States:

NameDescription
FN[Frequency clock protein]
M[Frequency clock protein; messenger RNA; (5')ppPur-mRNA]
FC[Frequency clock protein]

Leloup1999_CircClock: BIOMD0000000021v0.0.1

This model originates from BioModels Database: A Database of Annotated Published Models. It is copyright (c) 2005-2009 T…

Details

In Drosophila, circadian oscillations in the levels of two proteins, PER and TIM, result from the negative feedback exerted by a PER-TIM complex on the expression of the per and tim genes which code for these two proteins. On the basis of these experimental observations, we have recently proposed a theoretical model for circadian oscillations of the PER and TIM proteins in Drosophila. Here we show that for constant environmental conditions this model is capable of generating autonomous chaotic oscillations. For other parameter values, the model can also display birhythmicity, i.e. the coexistence between two stable regimes of limit cycle oscillations. We analyse the occurrence of chaos and birhythmicity by means of bifurcation diagrams and locate the different domains of complex oscillatory behavior in parameter space. The relative smallness of these domains raises doubts as to the possible physiological significance of chaos and birhythmicity in regard to circadian rhythm generation. Beyond the particular context of circadian rhythms we discuss the results in the light of other mechanisms underlying chaos and birhythmicity in regulated biological systems. Copyright 1999 Academic Press. link: http://identifiers.org/pubmed/10366496

Parameters:

NameDescription
k3=1.2; k4=0.6Reaction: P2 + T2 => CC, Rate Law: Cell*k3*P2*T2-Cell*k4*CC
k_dC=0.01Reaction: CC =>, Rate Law: Cell*k_dC*CC
k_d=0.01Reaction: P1 =>, Rate Law: Cell*k_d*P1
k_dN=0.01Reaction: Cn =>, Rate Law: compartment_0000002*k_dN*Cn
V_dT = 2.0; K_dT=0.2; k_d=0.01Reaction: T2 =>, Rate Law: Cell*k_d*T2+Cell*V_dT*T2/(K_dT+T2)
V_2T=1.0; K_2T=2.0Reaction: T1 => T0, Rate Law: Cell*V_2T*T1/(K_2T+T1)
V_3P=8.0; K_3P=2.0Reaction: P1 => P2, Rate Law: Cell*V_3P*P1/(K_3P+P1)
V_1T=8.0; K_1T=2.0Reaction: T0 => T1, Rate Law: Cell*V_1T*T0/(K_1T+T0)
k1=0.6; k2=0.2Reaction: CC => Cn, Rate Law: Cell*k1*CC-compartment_0000002*k2*Cn
V_2P=1.0; K_2P=2.0Reaction: P1 => P0, Rate Law: Cell*V_2P*P1/(K_2P+P1)
V_mP=0.7; K_mP=0.2; k_d=0.01Reaction: Mp =>, Rate Law: Cell*k_d*Mp+Cell*V_mP*Mp/(K_mP+Mp)
K_IT=1.0; n=4.0; V_sT=1.0Reaction: => Mt; Cn, Rate Law: Cell*V_sT*K_IT^n/(K_IT^n+Cn^n)
v_sP=1.0; K_IP=1.0; n=4.0Reaction: => Mp; Cn, Rate Law: Cell*v_sP*K_IP^n/(K_IP^n+Cn^n)
k_sP=0.9Reaction: => P0; Mp, Rate Law: Cell*k_sP*Mp
V_1P=8.0; K1_P=2.0Reaction: P0 => P1, Rate Law: Cell*V_1P*P0/(K1_P+P0)
V_dP=2.0; K_dP=0.2; k_d=0.01Reaction: P2 =>, Rate Law: Cell*k_d*P2+Cell*V_dP*P2/(K_dP+P2)
V_mT = 0.7; K_mT=0.2; k_d=0.01Reaction: Mt =>, Rate Law: Cell*k_d*Mt+Cell*V_mT*Mt/(K_mT+Mt)
k_sT=0.9Reaction: => T0; Mt, Rate Law: Cell*k_sT*Mt
K_4P=2.0; V_4P=1.0Reaction: P2 => P1, Rate Law: Cell*V_4P*P2/(K_4P+P2)
K_3T=2.0; V_3T=8.0Reaction: T1 => T2, Rate Law: Cell*V_3T*T1/(K_3T+T1)
V_4T=1.0; K_4T=2.0Reaction: T2 => T1, Rate Law: Cell*V_4T*T2/(K_4T+T2)

States:

NameDescription
Cn[Protein timeless; Period circadian protein]
T1[Protein timeless]
Mp[messenger RNA; RNA]
T2[Protein timeless]
T0[Protein timeless]
CC[Protein timeless; Period circadian protein]
P2[Period circadian protein]
P1[Period circadian protein]
Mt[messenger RNA; RNA]
P0[Period circadian protein]

Leloup2003_CircClock_DD: BIOMD0000000073v0.0.1

This model is according to the paper *Toward a detailed computational model for the mammalian circadian clock* . In thi…

Details

We present a computational model for the mammalian circadian clock based on the intertwined positive and negative regulatory loops involving the Per, Cry, Bmal1, Clock, and Rev-Erb alpha genes. In agreement with experimental observations, the model can give rise to sustained circadian oscillations in continuous darkness, characterized by an antiphase relationship between Per/Cry/Rev-Erbalpha and Bmal1 mRNAs. Sustained oscillations correspond to the rhythms autonomously generated by suprachiasmatic nuclei. For other parameter values, damped oscillations can also be obtained in the model. These oscillations, which transform into sustained oscillations when coupled to a periodic signal, correspond to rhythms produced by peripheral tissues. When incorporating the light-induced expression of the Per gene, the model accounts for entrainment of the oscillations by light-dark cycles. Simulations show that the phase of the oscillations can then vary by several hours with relatively minor changes in parameter values. Such a lability of the phase could account for physiological disorders related to circadian rhythms in humans, such as advanced or delayed sleep phase syndrome, whereas the lack of entrainment by light-dark cycles can be related to the non-24h sleep-wake syndrome. The model uncovers the possible existence of multiple sources of oscillatory behavior. Thus, in conditions where the indirect negative autoregulation of Per and Cry expression is inoperative, the model indicates the possibility that sustained oscillations might still arise from the negative autoregulation of Bmal1 expression. link: http://identifiers.org/pubmed/12775757

Parameters:

NameDescription
Vs=1.5; K=0.7; n=4.0Reaction: => species_7; species_3, Rate Law: cell*Vs*species_3^n/(K^n+species_3^n)
V=0.5; Km=0.3Reaction: species_2 =>, Rate Law: cell*V*species_2/(Km+species_2)
V=0.2; Km=0.1Reaction: species_13 => species_3, Rate Law: cell*V*species_13/(Km+species_13)
k1=0.4; k2=0.2Reaction: species_1 => species_3, Rate Law: cell*(k1*species_1-k2*species_3)
Vs=1.1; K=0.6; n=4.0Reaction: => species_5; species_3, Rate Law: cell*Vs*species_3^n/(K^n+species_3^n)
k=0.12Reaction: => species_1; species_0, Rate Law: cell*k*species_0
V=0.1; Km=0.1Reaction: species_14 => species_12, Rate Law: cell*V*species_14/(Km+species_14)
vsb=1.0; m=2.0; K=2.2Reaction: => species_0; species_3, Rate Law: cell*vsb*K^m/(K^m+species_3^m)
k=1.6Reaction: => species_4; species_5, Rate Law: cell*k*species_5
k1=0.12Reaction: species_4 =>, Rate Law: cell*k1*species_4
k1=0.01Reaction: species_11 =>, Rate Law: cell*k1*species_11
k=0.6Reaction: => species_8; species_7, Rate Law: cell*k*species_7
V=0.3; Km=0.1Reaction: species_9 => species_8, Rate Law: cell*V*species_9/(Km+species_9)
V=0.7; Km=0.3Reaction: species_11 =>, Rate Law: cell*V*species_11/(Km+species_11)
V=0.5; Km=0.1Reaction: species_3 => species_13, Rate Law: cell*V*species_3/(Km+species_3)
Km=0.31; V=1.1Reaction: species_7 =>, Rate Law: cell*V*species_7/(Km+species_7)
V=1.0; Km=0.4Reaction: species_5 =>, Rate Law: cell*V*species_5/(Km+species_5)
V=0.4; Km=0.1Reaction: species_12 => species_14, Rate Law: cell*V*species_12/(Km+species_12)
V=0.8; Km=0.4Reaction: species_0 =>, Rate Law: cell*V*species_0/(Km+species_0)
k1=0.5; k2=0.1Reaction: species_12 + species_3 => species_15, Rate Law: cell*(k1*species_12*species_3-k2*species_15)
V=0.6; Km=0.1Reaction: species_4 => species_6, Rate Law: cell*V*species_4/(Km+species_4)
V=0.8; Km=0.3Reaction: species_15 =>, Rate Law: cell*V*species_15/(Km+species_15)
V=0.6; Km=0.3Reaction: species_13 =>, Rate Law: cell*V*species_13/(Km+species_13)

States:

NameDescription
species 9[Period circadian protein homolog 1; Period circadian protein homolog 2; Period circadian protein homolog 3]
species 2[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
species 6[Cryptochrome-1; Cryptochrome-2]
species 10[Period circadian protein homolog 1; Cryptochrome-1; Period circadian protein homolog 2; Cryptochrome-1; Period circadian protein homolog 3; Cryptochrome-1]
species 11[Period circadian protein homolog 1; Cryptochrome-1; Period circadian protein homolog 2; Cryptochrome-2; Period circadian protein homolog 3; Cryptochrome-2]
species 1[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
species 4[Cryptochrome-1; Cryptochrome-2]
species 14[Period circadian protein homolog 1; Cryptochrome-1; Period circadian protein homolog 1; Cryptochrome-2; Period circadian protein homolog 3; Cryptochrome-2]
species 3[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
species 0[messenger RNA]
species 8[Period circadian protein homolog 1; Period circadian protein homolog 2; Period circadian protein homolog 3]
species 12[Period circadian protein homolog 1; Cryptochrome-1; Period circadian protein homolog 3; Cryptochrome-1; Period circadian protein homolog 3; Cryptochrome-2]
species 7[messenger RNA]
species 5[messenger RNA]
species 15In
species 13[Aryl hydrocarbon receptor nuclear translocator-like protein 1]

Leloup2003_CircClock_DD_REV-ERBalpha: BIOMD0000000074v0.0.1

This is model in continous darkness (DD) described in the article *Toward a detailed computational model for the mammali…

Details

We present a computational model for the mammalian circadian clock based on the intertwined positive and negative regulatory loops involving the Per, Cry, Bmal1, Clock, and Rev-Erb alpha genes. In agreement with experimental observations, the model can give rise to sustained circadian oscillations in continuous darkness, characterized by an antiphase relationship between Per/Cry/Rev-Erbalpha and Bmal1 mRNAs. Sustained oscillations correspond to the rhythms autonomously generated by suprachiasmatic nuclei. For other parameter values, damped oscillations can also be obtained in the model. These oscillations, which transform into sustained oscillations when coupled to a periodic signal, correspond to rhythms produced by peripheral tissues. When incorporating the light-induced expression of the Per gene, the model accounts for entrainment of the oscillations by light-dark cycles. Simulations show that the phase of the oscillations can then vary by several hours with relatively minor changes in parameter values. Such a lability of the phase could account for physiological disorders related to circadian rhythms in humans, such as advanced or delayed sleep phase syndrome, whereas the lack of entrainment by light-dark cycles can be related to the non-24h sleep-wake syndrome. The model uncovers the possible existence of multiple sources of oscillatory behavior. Thus, in conditions where the indirect negative autoregulation of Per and Cry expression is inoperative, the model indicates the possibility that sustained oscillations might still arise from the negative autoregulation of Bmal1 expression. link: http://identifiers.org/pubmed/12775757

Parameters:

NameDescription
V=0.2; Km=0.1Reaction: species_2 => species_1, Rate Law: compartment_0*V*species_2/(Km+species_2)
m=2.0; K=1.0; vsb=1.8Reaction: => species_0; species_18, Rate Law: compartment_0*vsb*K^m/(K^m+species_18^m)
k=1.2Reaction: => species_8; species_7, Rate Law: compartment_0*k*species_7
k1=1.0; k2=0.2Reaction: species_12 + species_3 => species_15, Rate Law: compartment_0*(k1*species_12*species_3-k2*species_15)
V=3.4; Km=0.3Reaction: species_9 =>, Rate Law: compartment_0*V*species_9/(Km+species_9)
Vs=2.4; K=0.6; n=2.0Reaction: => species_7; species_3, Rate Law: compartment_0*Vs*species_3^n/(K^n+species_3^n)
V=4.4; Km=0.3Reaction: species_17 =>, Rate Law: compartment_0*V*species_17/(Km+species_17)
V=2.2; Km=0.3Reaction: species_7 =>, Rate Law: compartment_0*V*species_7/(Km+species_7)
Km=1.006; V=9.6Reaction: species_8 => species_9, Rate Law: compartment_0*V*species_8/(Km+species_8)
Vs=1.6; K=0.6; n=2.0Reaction: => species_16; species_3, Rate Law: compartment_0*Vs*species_3^n/(K^n+species_3^n)
Km=1.006; V=2.4Reaction: species_10 => species_11, Rate Law: compartment_0*V*species_10/(Km+species_10)
Km=1.006; V=1.2Reaction: species_4 => species_6, Rate Law: compartment_0*V*species_4/(Km+species_4)
V=1.6; Km=0.4Reaction: species_16 =>, Rate Law: compartment_0*V*species_16/(Km+species_16)
Km=1.006; V=1.4Reaction: species_1 => species_2, Rate Law: compartment_0*V*species_1/(Km+species_1)
k2=0.4; k1=0.8Reaction: species_1 => species_3, Rate Law: compartment_0*(k1*species_1-k2*species_3)
k=1.7Reaction: => species_17; species_16, Rate Law: compartment_0*k*species_16
V=1.3; Km=0.4Reaction: species_0 =>, Rate Law: compartment_0*V*species_0/(Km+species_0)
Vs=2.2; K=0.6; n=2.0Reaction: => species_5; species_3, Rate Law: compartment_0*Vs*species_3^n/(K^n+species_3^n)
V=0.4; Km=0.1Reaction: species_13 => species_3, Rate Law: compartment_0*V*species_13/(Km+species_13)
V=2.0; Km=0.4Reaction: species_5 =>, Rate Law: compartment_0*V*species_5/(Km+species_5)
V=3.0; Km=0.3Reaction: species_2 =>, Rate Law: compartment_0*V*species_2/(Km+species_2)
V=0.6; Km=0.1Reaction: species_9 => species_8, Rate Law: compartment_0*V*species_9/(Km+species_9)
V=0.8; Km=0.3Reaction: species_18 =>, Rate Law: compartment_0*V*species_18/(Km+species_18)
k=3.2Reaction: => species_4; species_5, Rate Law: compartment_0*k*species_5
k=0.32Reaction: => species_1; species_0, Rate Law: compartment_0*k*species_0
k1=0.02Reaction: species_2 =>, Rate Law: compartment_0*k1*species_2
V=1.6; Km=0.3Reaction: species_15 =>, Rate Law: compartment_0*V*species_15/(Km+species_15)
V=1.4; Km=0.3Reaction: species_6 =>, Rate Law: compartment_0*V*species_6/(Km+species_6)

States:

NameDescription
species 9[Period circadian protein homolog 1; Period circadian protein homolog 2; Period circadian protein homolog 3]
species 2[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
species 6[Cryptochrome-1; Cryptochrome-2]
species 10[Period circadian protein homolog 1; Cryptochrome-1; Period circadian protein homolog 3; Cryptochrome-2]
species 11[Period circadian protein homolog 1; Cryptochrome-1; Period circadian protein homolog 3; Cryptochrome-2]
species 1[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
species 18[Rev-erb alpha]
species 4[Cryptochrome-1; Cryptochrome-2]
species 16[messenger RNA]
species 14[Period circadian protein homolog 1; Cryptochrome-1; Period circadian protein homolog 2; Cryptochrome-1; Period circadian protein homolog 3; Cryptochrome-2]
species 3[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
species 0[messenger RNA]
species 8[Period circadian protein homolog 1; Period circadian protein homolog 2; Period circadian protein homolog 3]
species 17[Rev-erb alpha]
species 12[Period circadian protein homolog 1; Cryptochrome-1; Period circadian protein homolog 3; Cryptochrome-2]
species 7[messenger RNA]
species 5[messenger RNA]
species 15In
species 13[Aryl hydrocarbon receptor nuclear translocator-like protein 1]

Leloup2003_CircClock_LD: BIOMD0000000078v0.0.1

This model is described in the paper *Toward a detailed computational model for the mammalian circadian clock* . In thi…

Details

We present a computational model for the mammalian circadian clock based on the intertwined positive and negative regulatory loops involving the Per, Cry, Bmal1, Clock, and Rev-Erb alpha genes. In agreement with experimental observations, the model can give rise to sustained circadian oscillations in continuous darkness, characterized by an antiphase relationship between Per/Cry/Rev-Erbalpha and Bmal1 mRNAs. Sustained oscillations correspond to the rhythms autonomously generated by suprachiasmatic nuclei. For other parameter values, damped oscillations can also be obtained in the model. These oscillations, which transform into sustained oscillations when coupled to a periodic signal, correspond to rhythms produced by peripheral tissues. When incorporating the light-induced expression of the Per gene, the model accounts for entrainment of the oscillations by light-dark cycles. Simulations show that the phase of the oscillations can then vary by several hours with relatively minor changes in parameter values. Such a lability of the phase could account for physiological disorders related to circadian rhythms in humans, such as advanced or delayed sleep phase syndrome, whereas the lack of entrainment by light-dark cycles can be related to the non-24h sleep-wake syndrome. The model uncovers the possible existence of multiple sources of oscillatory behavior. Thus, in conditions where the indirect negative autoregulation of Per and Cry expression is inoperative, the model indicates the possibility that sustained oscillations might still arise from the negative autoregulation of Bmal1 expression. link: http://identifiers.org/pubmed/12775757

Parameters:

NameDescription
V=0.5; Km=0.3Reaction: species_2 =>, Rate Law: cell*V*species_2/(Km+species_2)
V=0.2; Km=0.1Reaction: species_13 => species_3, Rate Law: cell*V*species_13/(Km+species_13)
k1=0.4; k2=0.2Reaction: species_4 + species_8 => species_10, Rate Law: cell*(k1*species_4*species_8-k2*species_10)
Vs=1.1; K=0.6; n=4.0Reaction: => species_5; species_3, Rate Law: cell*Vs*species_3^n/(K^n+species_3^n)
k=0.12Reaction: => species_1; species_0, Rate Law: cell*k*species_0
V=0.1; Km=0.1Reaction: species_6 => species_4, Rate Law: cell*V*species_6/(Km+species_6)
vsb=1.0; m=2.0; K=2.2Reaction: => species_0; species_3, Rate Law: cell*vsb*K^m/(K^m+species_3^m)
k=1.6Reaction: => species_4; species_5, Rate Law: cell*k*species_5
k1=0.12Reaction: species_4 =>, Rate Law: cell*k1*species_4
k1=0.01Reaction: species_7 =>, Rate Law: cell*k1*species_7
k=0.6Reaction: => species_8; species_7, Rate Law: cell*k*species_7
V=0.3; Km=0.1Reaction: species_9 => species_8, Rate Law: cell*V*species_9/(Km+species_9)
V=0.7; Km=0.3Reaction: species_14 =>, Rate Law: cell*V*species_14/(Km+species_14)
parameter_0000082 = NaN; K=0.7; n=4.0Reaction: => species_7; species_3, Rate Law: cell*parameter_0000082*species_3^n/(K^n+species_3^n)
V=0.5; Km=0.1Reaction: species_3 => species_13, Rate Law: cell*V*species_3/(Km+species_3)
Km=0.31; V=1.1Reaction: species_7 =>, Rate Law: cell*V*species_7/(Km+species_7)
V=1.0; Km=0.4Reaction: species_5 =>, Rate Law: cell*V*species_5/(Km+species_5)
V=0.4; Km=0.1Reaction: species_8 => species_9, Rate Law: cell*V*species_8/(Km+species_8)
k1=0.5; k2=0.1Reaction: species_12 + species_3 => species_15, Rate Law: cell*(k1*species_12*species_3-k2*species_15)
V=0.8; Km=0.4Reaction: species_0 =>, Rate Law: cell*V*species_0/(Km+species_0)
V=0.6; Km=0.1Reaction: species_4 => species_6, Rate Law: cell*V*species_4/(Km+species_4)
V=0.8; Km=0.3Reaction: species_15 =>, Rate Law: cell*V*species_15/(Km+species_15)
V=0.6; Km=0.3Reaction: species_13 =>, Rate Law: cell*V*species_13/(Km+species_13)

States:

NameDescription
species 9[Period circadian protein homolog 3; Period circadian protein homolog 2; Period circadian protein homolog 1]
species 2[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
species 6[Cryptochrome-2; Cryptochrome-1]
species 10[Period circadian protein homolog 3; Cryptochrome-2; Period circadian protein homolog 1; Cryptochrome-2; Period circadian protein homolog 1; Cryptochrome-1]
species 11[Period circadian protein homolog 3; Cryptochrome-2; Period circadian protein homolog 1; Cryptochrome-1]
species 1[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
species 4[Cryptochrome-2; Cryptochrome-1]
species 14[Period circadian protein homolog 3; Cryptochrome-2; Period circadian protein homolog 2; Cryptochrome-2; Period circadian protein homolog 1; Cryptochrome-1]
species 3[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
species 0[messenger RNA]
species 8[Period circadian protein homolog 3; Period circadian protein homolog 2; Period circadian protein homolog 1]
species 12[Period circadian protein homolog 3; Cryptochrome-2; Period circadian protein homolog 1; Cryptochrome-2; Period circadian protein homolog 1; Cryptochrome-1]
species 7[messenger RNA]
species 5[messenger RNA]
species 15In
species 13[Aryl hydrocarbon receptor nuclear translocator-like protein 1]

Leloup2003_CircClock_LD_REV-ERBalpha: BIOMD0000000083v0.0.1

This is model is according to the paper*Toward a detailed computational model for the mammalian circadian clock* In thi…

Details

We present a computational model for the mammalian circadian clock based on the intertwined positive and negative regulatory loops involving the Per, Cry, Bmal1, Clock, and Rev-Erb alpha genes. In agreement with experimental observations, the model can give rise to sustained circadian oscillations in continuous darkness, characterized by an antiphase relationship between Per/Cry/Rev-Erbalpha and Bmal1 mRNAs. Sustained oscillations correspond to the rhythms autonomously generated by suprachiasmatic nuclei. For other parameter values, damped oscillations can also be obtained in the model. These oscillations, which transform into sustained oscillations when coupled to a periodic signal, correspond to rhythms produced by peripheral tissues. When incorporating the light-induced expression of the Per gene, the model accounts for entrainment of the oscillations by light-dark cycles. Simulations show that the phase of the oscillations can then vary by several hours with relatively minor changes in parameter values. Such a lability of the phase could account for physiological disorders related to circadian rhythms in humans, such as advanced or delayed sleep phase syndrome, whereas the lack of entrainment by light-dark cycles can be related to the non-24h sleep-wake syndrome. The model uncovers the possible existence of multiple sources of oscillatory behavior. Thus, in conditions where the indirect negative autoregulation of Per and Cry expression is inoperative, the model indicates the possibility that sustained oscillations might still arise from the negative autoregulation of Bmal1 expression. link: http://identifiers.org/pubmed/12775757

Parameters:

NameDescription
k6 = 0.4; k5 = 0.8Reaction: Bc => Bn, Rate Law: cell*(k5*Bc-k6*Bn)
Kdp = 0.1; V4B = 0.4Reaction: Bnp => Bn, Rate Law: cell*V4B*Bnp/(Kdp+Bnp)
Kp = 1.006; V3PC = 2.4Reaction: PCn => PCnp, Rate Law: cell*V3PC*PCn/(Kp+PCn)
Kdp = 0.1; V2C = 0.2Reaction: Ccp => Cc, Rate Law: cell*V2C*Ccp/(Kdp+Ccp)
Kd = 0.3; vdCC = 1.4Reaction: Ccp =>, Rate Law: cell*vdCC*Ccp/(Kd+Ccp)
vmB = 1.3; KmB = 0.4Reaction: Mb =>, Rate Law: cell*vmB*Mb/(KmB+Mb)
Kdp = 0.1; V2B = 0.2Reaction: Bcp => Bc, Rate Law: cell*V2B*Bcp/(Kdp+Bcp)
k4 = 0.4; k3 = 0.8Reaction: Cc + Pc => PCc, Rate Law: cell*(k3*Cc*Pc-k4*PCc)
KmC = 0.4; vmC = 2.0Reaction: Mc =>, Rate Law: cell*vmC*Mc/(KmC+Mc)
kdnC = 0.02Reaction: Cc =>, Rate Law: cell*kdnC*Cc
vdBN = 3.0; Kd = 0.3Reaction: Bnp =>, Rate Law: cell*vdBN*Bnp/(Kd+Bnp)
Kdp = 0.1; V4PC = 0.2Reaction: PCnp => PCn, Rate Law: cell*V4PC*PCnp/(Kdp+PCnp)
Kp = 1.006; V1P = 9.6Reaction: Pc => Pcp, Rate Law: cell*V1P*Pc/(Kp+Pc)
Kp = 1.006; V1PC = 2.4Reaction: PCc => PCcp, Rate Law: cell*V1PC*PCc/(Kp+PCc)
Kd = 0.3; vdRC = 4.4Reaction: Rc =>, Rate Law: cell*vdRC*Rc/(Kd+Rc)
Kd = 0.3; vdBC = 3.0Reaction: Bcp =>, Rate Law: cell*vdBC*Bcp/(Kd+Bcp)
Kp = 1.006; V1C = 1.2Reaction: Cc => Ccp, Rate Law: cell*V1C*Cc/(Kp+Cc)
kdmb = 0.02Reaction: Mb =>, Rate Law: cell*kdmb*Mb
KmP = 0.3; vmP = 2.2Reaction: Mp =>, Rate Law: cell*vmP*Mp/(KmP+Mp)
Kd = 0.3; vdIN = 1.6Reaction: In =>, Rate Law: cell*vdIN*In/(Kd+In)
m = 2.0; K=1.0; vsB = 1.8Reaction: => Mb; Rn, Rate Law: cell*vsB*K^m/(K^m+Rn^m)
k2 = 0.4; k1 = 0.8Reaction: PCc => PCn, Rate Law: cell*(k1*PCc-k2*PCn)
kdmc = 0.02Reaction: Mc =>, Rate Law: cell*kdmc*Mc
kdn = 0.02Reaction: Bn =>, Rate Law: cell*kdn*Bn
Kdp = 0.1; V2P = 0.6Reaction: Pcp => Pc, Rate Law: cell*V2P*Pcp/(Kdp+Pcp)
vdRN = 0.8; Kd = 0.3Reaction: Rn =>, Rate Law: cell*vdRN*Rn/(Kd+Rn)
k9 = 0.8; k10 = 0.4Reaction: Rc => Rn, Rate Law: cell*(k9*Rc-k10*Rn)
Kp = 1.006; V1B = 1.4Reaction: Bc => Bcp, Rate Law: cell*V1B*Bc/(Kp+Bc)
parameter_0000097 = 3.0; n = 2.0; KAP = 0.6Reaction: => Mp; Bn, Rate Law: cell*parameter_0000097*Bn^n/(KAP^n+Bn^n)
vmR = 1.6; kmR = 0.4Reaction: Mr =>, Rate Law: cell*vmR*Mr/(kmR+Mr)
kdmr = 0.02Reaction: Mr =>, Rate Law: cell*kdmr*Mr
Kp = 1.006; V3B = 1.4Reaction: Bn => Bnp, Rate Law: cell*V3B*Bn/(Kp+Bn)
ksB = 0.32Reaction: => Bc; Mb, Rate Law: cell*ksB*Mb
vdPCC = 1.4; Kd = 0.3Reaction: PCcp =>, Rate Law: cell*vdPCC*PCcp/(Kd+PCcp)
k7 = 1.0; k8 = 0.2Reaction: PCn + Bn => In, Rate Law: cell*(k7*PCn*Bn-k8*In)
Kd = 0.3; VdPC = 3.4Reaction: Pcp =>, Rate Law: cell*VdPC*Pcp/(Kd+Pcp)
Kdp = 0.1; V2PC = 0.2Reaction: PCcp => PCc, Rate Law: cell*V2PC*PCcp/(Kdp+PCcp)
ksP = 1.2Reaction: => Pc; Mp, Rate Law: cell*ksP*Mp
vsC = 2.2; KAC = 0.6; n = 2.0Reaction: => Mc; Bn, Rate Law: cell*vsC*Bn^n/(KAC^n+Bn^n)
h = 2.0; vsR = 1.6; KAR = 0.6Reaction: => Mr; Bn, Rate Law: cell*vsR*Bn^h/(KAR^h+Bn^h)
Kd = 0.3; vdPCN = 1.4Reaction: PCnp =>, Rate Law: cell*vdPCN*PCnp/(Kd+PCnp)
ksR = 1.7Reaction: => Rc; Mr, Rate Law: cell*ksR*Mr
ksC = 3.2Reaction: => Cc; Mc, Rate Law: cell*ksC*Mc
kdmp = 0.02Reaction: Mp =>, Rate Law: cell*kdmp*Mp

States:

NameDescription
Ccp[Cryptochrome-2; Cryptochrome-1]
PCc[Cryptochrome-1; Period circadian protein homolog 1; Cryptochrome-2; Period circadian protein homolog 3]
Bnp[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
PCcp[Period circadian protein homolog 3; Period circadian protein homolog 1; Cryptochrome-1; Cryptochrome-2]
Mb[Aryl hydrocarbon receptor nuclear translocator-like protein 1; messenger RNA]
Mr[Rev-erb alpha; messenger RNA]
Pc[Period circadian protein homolog 2; Period circadian protein homolog 1; Period circadian protein homolog 3]
Bcp[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
Rc[Rev-erb alpha]
Bn[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
Cc[Cryptochrome-2; Cryptochrome-1]
Pcp[Period circadian protein homolog 1; Period circadian protein homolog 2; Period circadian protein homolog 3]
Mp[Period circadian protein homolog 3; Period circadian protein homolog 2; Period circadian protein homolog 1; messenger RNA]
Bc[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
PCnp[Period circadian protein homolog 3; Cryptochrome-2; Cryptochrome-1; Period circadian protein homolog 1]
PCn[Cryptochrome-2; Cryptochrome-1; Period circadian protein homolog 3; Period circadian protein homolog 1]
Rn[Rev-erb alpha]
InIn
Mc[Cryptochrome-1; Cryptochrome-2; messenger RNA]

Leloup2004 - Mammalian Circadian Rhythm models for 23.8 and 24.2 hours timeperiod: BIOMD0000000975v0.0.1

We extend the study of a computational model recently proposed for the mammalian circadian clock (Proc. Natl Acad. Sci.…

Details

We extend the study of a computational model recently proposed for the mammalian circadian clock (Proc. Natl Acad. Sci. USA 100 (2003) 7051). The model, based on the intertwined positive and negative regulatory loops involving the Per, Cry, Bmal1, and Clock genes, can give rise to sustained circadian oscillations in conditions of continuous darkness. These limit cycle oscillations correspond to circadian rhythms autonomously generated by suprachiasmatic nuclei and by some peripheral tissues. By using different sets of parameter values producing circadian oscillations, we compare the effect of the various parameters and show that both the occurrence and the period of the oscillations are generally most sensitive to parameters related to synthesis or degradation of Bmal1 mRNA and BMAL1 protein. The mechanism of circadian oscillations relies on the formation of an inactive complex between PER and CRY and the activators CLOCK and BMAL1 that enhance Per and Cry expression. Bifurcation diagrams and computer simulations nevertheless indicate the possible existence of a second source of oscillatory behavior. Thus, sustained oscillations might arise from the sole negative autoregulation of Bmal1 expression. This second oscillatory mechanism may not be functional in physiological conditions, and its period need not necessarily be circadian. When incorporating the light-induced expression of the Per gene, the model accounts for entrainment of the oscillations by light-dark (LD) cycles. Long-term suppression of circadian oscillations by a single light pulse can occur in the model when a stable steady state coexists with a stable limit cycle. The phase of the oscillations upon entrainment in LD critically depends on the parameters that govern the level of CRY protein. Small changes in the parameters governing CRY levels can shift the peak in Per mRNA from the L to the D phase, or can prevent entrainment. The results are discussed in relation to physiological disorders of the sleep-wake cycle linked to perturbations of the human circadian clock, such as the familial advanced sleep phase syndrome or the non-24h sleep-wake syndrome. link: http://identifiers.org/pubmed/15363675

Lemaire2004 - Role of RANK/RANKL/OPG pathway in bone remodelling process: BIOMD0000000278v0.0.1

This a model from the article: Modeling the interactions between osteoblast and osteoclast activities in bone remode…

Details

We propose a mathematical model explaining the interactions between osteoblasts and osteoclasts, two cell types specialized in the maintenance of the bone integrity. Bone is a dynamic, living tissue whose structure and shape continuously evolves during life. It has the ability to change architecture by removal of old bone and replacement with newly formed bone in a localized process called remodeling. The model described here is based on the idea that the relative proportions of immature and mature osteoblasts control the degree of osteoclastic activity. In addition, osteoclasts control osteoblasts differentially depending on their stage of differentiation. Despite the tremendous complexity of the bone regulatory system and its fragmentary understanding, we obtain surprisingly good correlations between the model simulations and the experimental observations extracted from the literature. The model results corroborate all behaviors of the bone remodeling system that we have simulated, including the tight coupling between osteoblasts and osteoclasts, the catabolic effect induced by continuous administration of PTH, the catabolic action of RANKL, as well as its reversal by soluble antagonist OPG. The model is also able to simulate metabolic bone diseases such as estrogen deficiency, vitamin D deficiency, senescence and glucocorticoid excess. Conversely, possible routes for therapeutic interventions are tested and evaluated. Our model confirms that anti-resorptive therapies are unable to partially restore bone loss, whereas bone formation therapies yield better results. The model enables us to determine and evaluate potential therapies based on their efficacy. In particular, the model predicts that combinations of anti-resorptive and anabolic therapies provide significant benefits compared with monotherapy, especially for certain type of skeletal disease. Finally, the model clearly indicates that increasing the size of the pool of preosteoblasts is an essential ingredient for the therapeutic manipulation of bone formation. This model was conceived as the first step in a bone turnover modeling platform. These initial modeling results are extremely encouraging and lead us to proceed with additional explorations into bone turnover and skeletal remodeling. link: http://identifiers.org/pubmed/15234198

Parameters:

NameDescription
D_C = 0.0021; D_A = 0.7; Phi_L = NaN; Phi_C = NaNReaction: C = D_C*Phi_L-D_A*Phi_C*C, Rate Law: D_C*Phi_L-D_A*Phi_C*C
D_B = NaN; D_R = 7.0E-4; Phi_C = NaNReaction: R = D_R*Phi_C-D_B*R/Phi_C, Rate Law: D_R*Phi_C-D_B*R/Phi_C
D_B = NaN; Phi_C = NaN; k_B = 0.189Reaction: B = D_B*R/Phi_C-k_B*B, Rate Law: D_B*R/Phi_C-k_B*B

States:

NameDescription
B[osteoblast]
C[osteoclast]
R[osteoblast]

Lemon2003_Ca2Dynamics: MODEL1006230039v0.0.1

This a model from the article: Metabotropic receptor activation, desensitization and sequestration-I: modelling calciu…

Details

A mathematical account is given of the processes governing the time courses of calcium ions (Ca2+), inositol 1,4,5-trisphosphate (IP(3)) and phosphatidylinositol 4,5-bisphosphate (PIP(2)) in single cells following the application of external agonist to metabotropic receptors. A model is constructed that incorporates the regulation of metabotropic receptor activity, the G-protein cascade and the Ca2+ dynamics in the cytosol. It is subsequently used to reproduce observations on the extent of desensitization and sequestration of the P(2)Y(2) receptor following its activation by uridine triphosphate (UTP). The theory predicts the dependence on agonist concentration of the change in the number of receptors in the membrane as well as the time course of disappearance of receptors from the plasmalemma, upon exposure to agonist. In addition, the extent of activation and desensitization of the receptor, using the calcium transients in cells initiated by exposure to agonist, is also predicted. Model predictions show the significance of membrane PIP(2) depletion and resupply on the time course of IP(3) and Ca2+ levels. Results of the modelling also reveal the importance of receptor recycling and PIP(2) resupply for maintaining Ca2+ and IP(3) levels during sustained application of agonist. link: http://identifiers.org/pubmed/12782119

Lenbury1991_CortisolSecretionSystem: MODEL0479926177v0.0.1

This a model from the article: Modelling fluctuation phenomena in the plasma cortisol secretion system in normal man.…

Details

A system of three non-linear differential equations with exponential feedback terms is proposed to model the self-regulating cortisol secretion system and explain the fluctuation patterns observed in clinical data. It is shown that the model exhibits bifurcation and chaos patterns for a certain range of parametric values. This helps us to explain clinical observations and characterize different dynamic behaviors of the self-regulative system. link: http://identifiers.org/pubmed/1668715

Lenbury2001_InsulinKineticsModel_A: BIOMD0000000878v0.0.1

This a model from the article: Modeling insulin kinetics: responses to a single oral glucose administration or ambulat…

Details

This paper presents a nonlinear mathematical model of the glucose-insulin feedback system, which has been extended to incorporate the beta-cells' function on maintaining and regulating plasma insulin level in man. Initially, a gastrointestinal absorption term for glucose is utilized to effect the glucose absorption by the intestine and the subsequent release of glucose into the bloodstream, taking place at a given initial rate and falling off exponentially with time. An analysis of the model is carried out by the singular perturbation technique in order to derive boundary conditions on the system parameters which identify, in particular, the existence of limit cycles in our model system consistent with the oscillatory patterns often observed in clinical data. We then utilize a sinusoidal term to incorporate the temporal absorption of glucose in order to study the responses in the patients under ambulatory-fed conditions. A numerical investigation is carried out in this case to construct a bifurcation diagram to identify the ranges of parametric values for which chaotic behavior can be expected, leading to interesting biological interpretations. link: http://identifiers.org/pubmed/11226623

Parameters:

NameDescription
r_5 = 0.1; r_6 = 0.1; y_hat = 1.24; z_hat = 2.57039578276886Reaction: => z; y, z, Rate Law: COMpartment*(r_5*(y-y_hat)*(z_hat-z)+r_6*z*(z_hat-z))
epsilon = 0.1; r_4 = 0.1Reaction: y => ; x, Rate Law: COMpartment*epsilon*r_4*x
r_7 = 0.05Reaction: z =>, Rate Law: COMpartment*r_7*z
r_1 = 0.2; c_1 = 0.1Reaction: => x; y, z, Rate Law: COMpartment*z*(r_1*y+c_1)
r_2 = 0.1Reaction: x => ; z, Rate Law: COMpartment*z*r_2*x
epsilon = 0.1; c_2 = 0.1; r_3 = 0.1Reaction: => y; z, Rate Law: COMpartment*(epsilon*r_3/z+epsilon*c_2)

States:

NameDescription
x[Insulin]
z[pancreatic beta cell]
y[C2831]

Lenbury2001_InsulinKineticsModel_B: MODEL1201140003v0.0.1

This a model from the article: Modeling insulin kinetics: responses to a single oral glucose administration or ambulat…

Details

This paper presents a nonlinear mathematical model of the glucose-insulin feedback system, which has been extended to incorporate the beta-cells' function on maintaining and regulating plasma insulin level in man. Initially, a gastrointestinal absorption term for glucose is utilized to effect the glucose absorption by the intestine and the subsequent release of glucose into the bloodstream, taking place at a given initial rate and falling off exponentially with time. An analysis of the model is carried out by the singular perturbation technique in order to derive boundary conditions on the system parameters which identify, in particular, the existence of limit cycles in our model system consistent with the oscillatory patterns often observed in clinical data. We then utilize a sinusoidal term to incorporate the temporal absorption of glucose in order to study the responses in the patients under ambulatory-fed conditions. A numerical investigation is carried out in this case to construct a bifurcation diagram to identify the ranges of parametric values for which chaotic behavior can be expected, leading to interesting biological interpretations. link: http://identifiers.org/pubmed/11226623

Leon-Triana2020 - CAR T-cell therapy in B-cell acute lymphoblastic leukaemia: BIOMD0000001011v0.0.1

This model is based on the publication: "CAR T cell therapy in B-cell acute lymphoblastic leukaemia: Insights from mathe…

Details

Immunotherapies use components of the patient immune system to selectively target can- cer cells. The use of chimeric antigenic receptor (CAR) T cells to treat B-cell malignancies –leukaemias and lymphomas–is one of the most successful examples, with many patients experiencing long-lasting full responses to this therapy. This treatment works by extract- ing the patient’s T cells and transducing them with the CAR, enabling them to recognize and target cells carrying the antigen CD19 + , which is expressed in these haematological cancers. Here we put forward a mathematical model describing the time response of leukaemias to the injection of CAR T cells. The model accounts for mature and progenitor B-cells, leukaemic cells, CAR T cells and side effects by including the main biological processes involved. The model explains the early post-injection dynamics of the different compart- ments and the fact that the number of CAR T cells injected does not critically affect the treatment outcome. An explicit formula is found that gives the maximum CAR T cell ex- pansion in vivo and the severity of side effects. Our mathematical model captures other known features of the response to this immunotherapy. It also predicts that CD19 + cancer relapses could be the result of competition between leukaemic and CAR T cells, analogous to predator-prey dynamics. We discuss this in the light of the available evidence and the possibility of controlling relapses by early re-challenging of the leukaemia cells with stored CAR T cells. link: http://identifiers.org/doi/10.1016/j.cnsns.2020.105570

Leon-Triana2020 - CAR T-cell therapy in B-cell acute lymphoblastic leukaemia with contribution from immature B cells: BIOMD0000001012v0.0.1

This model is based on the publication: "CAR T cell therapy in B-cell acute lymphoblastic leukaemia: Insights from mathe…

Details

Immunotherapies use components of the patient immune system to selectively target can- cer cells. The use of chimeric antigenic receptor (CAR) T cells to treat B-cell malignancies –leukaemias and lymphomas–is one of the most successful examples, with many patients experiencing long-lasting full responses to this therapy. This treatment works by extract- ing the patient’s T cells and transducing them with the CAR, enabling them to recognize and target cells carrying the antigen CD19 + , which is expressed in these haematological cancers. Here we put forward a mathematical model describing the time response of leukaemias to the injection of CAR T cells. The model accounts for mature and progenitor B-cells, leukaemic cells, CAR T cells and side effects by including the main biological processes involved. The model explains the early post-injection dynamics of the different compart- ments and the fact that the number of CAR T cells injected does not critically affect the treatment outcome. An explicit formula is found that gives the maximum CAR T cell ex- pansion in vivo and the severity of side effects. Our mathematical model captures other known features of the response to this immunotherapy. It also predicts that CD19 + cancer relapses could be the result of competition between leukaemic and CAR T cells, analogous to predator-prey dynamics. We discuss this in the light of the available evidence and the possibility of controlling relapses by early re-challenging of the leukaemia cells with stored CAR T cells. link: http://identifiers.org/doi/10.1016/j.cnsns.2020.105570

Leon-Triana2021 - Competition between tumour cells and dual-target CAR T-cells: BIOMD0000001014v0.0.1

This model of the use of chimeric antigen receptor (CAR)-T cell therapy in the treatment of solid tumours is described i…

Details

Chimeric antigen receptor (CAR)-T cell-based therapies have achieved substantial success against B-cell malignancies, which has led to a growing scientific and clinical interest in extending their use to solid cancers. However, results for solid tumours have been limited up to now, in part due to the immunosuppressive tumour microenvironment, which is able to inactivate CAR-T cell clones. In this paper we put forward a mathematical model describing the competition of CAR-T and tumour cells, taking into account their immunosuppressive capacity. Using the mathematical model, we show that the use of large numbers of CAR-T cells targetting the solid tumour antigens could overcome the immunosuppressive potential of cancer. To achieve such high levels of CAR-T cells we propose, and study computationally, the manufacture and injection of CAR-T cells targetting two antigens: CD19 and a tumour-associated antigen. We study in silico the resulting dynamics of the disease after the injection of this product and find that the expansion of the CAR-T cell population in the blood and lymphopoietic organs could lead to the massive production of an army of CAR-T cells targetting the solid tumour, and potentially overcoming its immune suppression capabilities. This strategy could benefit from the combination with PD-1 inhibitors and low tumour loads. Our computational results provide theoretical support for the treatment of different types of solid tumours using T cells engineered with combination treatments of dual CARs with on- and off-tumour activity and anti-PD-1 drugs after completion of classical cytoreductive treatments. link: http://identifiers.org/pubmed/33572301

Leon-Triana2021 - Competition between tumour cells and single-target CAR T-cells: BIOMD0000001013v0.0.1

This model of the use of chimeric antigen receptor (CAR)-T cell therapy in the treatment of solid tumours is described i…

Details

Chimeric antigen receptor (CAR)-T cell-based therapies have achieved substantial success against B-cell malignancies, which has led to a growing scientific and clinical interest in extending their use to solid cancers. However, results for solid tumours have been limited up to now, in part due to the immunosuppressive tumour microenvironment, which is able to inactivate CAR-T cell clones. In this paper we put forward a mathematical model describing the competition of CAR-T and tumour cells, taking into account their immunosuppressive capacity. Using the mathematical model, we show that the use of large numbers of CAR-T cells targetting the solid tumour antigens could overcome the immunosuppressive potential of cancer. To achieve such high levels of CAR-T cells we propose, and study computationally, the manufacture and injection of CAR-T cells targetting two antigens: CD19 and a tumour-associated antigen. We study in silico the resulting dynamics of the disease after the injection of this product and find that the expansion of the CAR-T cell population in the blood and lymphopoietic organs could lead to the massive production of an army of CAR-T cells targetting the solid tumour, and potentially overcoming its immune suppression capabilities. This strategy could benefit from the combination with PD-1 inhibitors and low tumour loads. Our computational results provide theoretical support for the treatment of different types of solid tumours using T cells engineered with combination treatments of dual CARs with on- and off-tumour activity and anti-PD-1 drugs after completion of classical cytoreductive treatments. link: http://identifiers.org/pubmed/33572301

Levchenko2000_MAPK_noScaffold: BIOMD0000000011v0.0.1

# MAPK cascade in solution (no scaffold) DescriptionThis model describes a basic 3- stage Mitogen Activated Prote…

Details

In addition to preventing crosstalk among related signaling pathways, scaffold proteins might facilitate signal transduction by preforming multimolecular complexes that can be rapidly activated by incoming signal. In many cases, such as mitogen-activated protein kinase (MAPK) cascades, scaffold proteins are necessary for full activation of a signaling pathway. To date, however, no detailed biochemical model of scaffold action has been suggested. Here we describe a quantitative computer model of MAPK cascade with a generic scaffold protein. Analysis of this model reveals that formation of scaffold-kinase complexes can be used effectively to regulate the specificity, efficiency, and amplitude of signal propagation. In particular, for any generic scaffold there exists a concentration value optimal for signal amplitude. The location of the optimum is determined by the concentrations of the kinases rather than their binding constants and in this way is scaffold independent. This effect and the alteration of threshold properties of the signal propagation at high scaffold concentrations might alter local signaling properties at different subcellular compartments. Different scaffold levels and types might then confer specialized properties to tune evolutionarily conserved signaling modules to specific cellular contexts. link: http://identifiers.org/pubmed/10823939

Parameters:

NameDescription
a3=3.3Reaction: MEK + RAFp => MEKRAFp, Rate Law: a3*MEK*RAFp
a7=20.0Reaction: MAPK + MEKpp => MAPKMEKpp, Rate Law: a7*MAPK*MEKpp
a5=3.3Reaction: MEKp + RAFp => MEKpRAFp, Rate Law: a5*MEKp*RAFp
k5=0.1Reaction: MEKpRAFp => MEKpp + RAFp, Rate Law: k5*MEKpRAFp
k3=0.1Reaction: MEKRAFp => MEKp + RAFp, Rate Law: k3*MEKRAFp
k10=0.1Reaction: MAPKppMAPKPH => MAPKp + MAPKPH, Rate Law: k10*MAPKppMAPKPH
a2=0.5Reaction: RAFp + RAFPH => RAFpRAFPH, Rate Law: a2*RAFp*RAFPH
a6=10.0Reaction: MEKPH + MEKpp => MEKppMEKPH, Rate Law: a6*MEKPH*MEKpp
a9=20.0Reaction: MAPKp + MEKpp => MAPKpMEKpp, Rate Law: a9*MAPKp*MEKpp
k4=0.1Reaction: MEKpMEKPH => MEK + MEKPH, Rate Law: k4*MEKpMEKPH
k7=0.1Reaction: MAPKMEKpp => MAPKp + MEKpp, Rate Law: k7*MAPKMEKpp
d6=0.8Reaction: MEKppMEKPH => MEKPH + MEKpp, Rate Law: d6*MEKppMEKPH
d2=0.5Reaction: RAFpRAFPH => RAFp + RAFPH, Rate Law: d2*RAFpRAFPH
a4=10.0Reaction: MEKp + MEKPH => MEKpMEKPH, Rate Law: a4*MEKp*MEKPH
k9=0.1Reaction: MAPKpMEKpp => MAPKpp + MEKpp, Rate Law: k9*MAPKpMEKpp
d10=0.4Reaction: MAPKppMAPKPH => MAPKPH + MAPKpp, Rate Law: d10*MAPKppMAPKPH
a1=1.0Reaction: RAF + RAFK => RAFRAFK, Rate Law: a1*RAF*RAFK
d3=0.42Reaction: MEKRAFp => MEK + RAFp, Rate Law: d3*MEKRAFp
d1=0.4Reaction: RAFRAFK => RAF + RAFK, Rate Law: d1*RAFRAFK
d5=0.4Reaction: MEKpRAFp => MEKp + RAFp, Rate Law: d5*MEKpRAFp
k8=0.1Reaction: MAPKpMAPKPH => MAPK + MAPKPH, Rate Law: k8*MAPKpMAPKPH
d8=0.4Reaction: MAPKpMAPKPH => MAPKp + MAPKPH, Rate Law: d8*MAPKpMAPKPH
d9=0.6Reaction: MAPKpMEKpp => MAPKp + MEKpp, Rate Law: d9*MAPKpMEKpp
a10=5.0Reaction: MAPKPH + MAPKpp => MAPKppMAPKPH, Rate Law: a10*MAPKPH*MAPKpp
d4=0.8Reaction: MEKpMEKPH => MEKp + MEKPH, Rate Law: d4*MEKpMEKPH
k2=0.1Reaction: RAFpRAFPH => RAF + RAFPH, Rate Law: k2*RAFpRAFPH
k1=0.1Reaction: RAFRAFK => RAFK + RAFp, Rate Law: k1*RAFRAFK
d7=0.6Reaction: MAPKMEKpp => MAPK + MEKpp, Rate Law: d7*MAPKMEKpp
k6=0.1Reaction: MEKppMEKPH => MEKp + MEKPH, Rate Law: k6*MEKppMEKPH
a8=5.0Reaction: MAPKp + MAPKPH => MAPKpMAPKPH, Rate Law: a8*MAPKp*MAPKPH

States:

NameDescription
MAPKPH[Dual specificity protein phosphatase 1-B]
RAFK[IPR003577]
RAFpRAFPH[RAF proto-oncogene serine/threonine-protein kinase]
MEKppMEKPH[Dual specificity mitogen-activated protein kinase kinase 1]
MEKpp[Dual specificity mitogen-activated protein kinase kinase 1]
MAPKp[Mitogen-activated protein kinase 1]
MEKpMEKPH[Dual specificity mitogen-activated protein kinase kinase 1]
MAPK[Mitogen-activated protein kinase 1]
MEKpRAFp[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
MAPKpMAPKPH[Mitogen-activated protein kinase 1; Dual specificity protein phosphatase 1-B]
MAPKMEKpp[Mitogen-activated protein kinase 1; Dual specificity mitogen-activated protein kinase kinase 1]
MAPKppMAPKPH[Mitogen-activated protein kinase 1; Dual specificity protein phosphatase 1-B]
MEKPHMEK phosphatase
MEKp[Dual specificity mitogen-activated protein kinase kinase 1]
RAFp[RAF proto-oncogene serine/threonine-protein kinase]
MEK[Dual specificity mitogen-activated protein kinase kinase 1]
MAPKpMEKpp[Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
RAF[RAF proto-oncogene serine/threonine-protein kinase]
RAFPHRAF phosphatase
RAFRAFK[RAF proto-oncogene serine/threonine-protein kinase; IPR003577]
MAPKpp[Mitogen-activated protein kinase 1]
MEKRAFp[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]

Levchenko2000_MAPK_Scaffold: BIOMD0000000014v0.0.1

# MAPK cascade on a scaffold CitationLevchenko, A., Bruck, J., Sternberg, P.W. (2000) .Scaffold proteins may biph…

Details

In addition to preventing crosstalk among related signaling pathways, scaffold proteins might facilitate signal transduction by preforming multimolecular complexes that can be rapidly activated by incoming signal. In many cases, such as mitogen-activated protein kinase (MAPK) cascades, scaffold proteins are necessary for full activation of a signaling pathway. To date, however, no detailed biochemical model of scaffold action has been suggested. Here we describe a quantitative computer model of MAPK cascade with a generic scaffold protein. Analysis of this model reveals that formation of scaffold-kinase complexes can be used effectively to regulate the specificity, efficiency, and amplitude of signal propagation. In particular, for any generic scaffold there exists a concentration value optimal for signal amplitude. The location of the optimum is determined by the concentrations of the kinases rather than their binding constants and in this way is scaffold independent. This effect and the alteration of threshold properties of the signal propagation at high scaffold concentrations might alter local signaling properties at different subcellular compartments. Different scaffold levels and types might then confer specialized properties to tune evolutionarily conserved signaling modules to specific cellular contexts. link: http://identifiers.org/pubmed/10823939

Parameters:

NameDescription
d3=0.42Reaction: K_K_2_0_3_1 => K_2_0 + K_3_1, Rate Law: d3*K_K_2_0_3_1
a3=3.3Reaction: K_2_0 + K_3_1 => K_K_2_0_3_1, Rate Law: a3*K_2_0*K_3_1
a7=20.0Reaction: K_1_0 + K_2_2 => K_K_1_0_2_2, Rate Law: a7*K_1_0*K_2_2
k3=0.1Reaction: S_m1_0_1 => S_m1_1_1, Rate Law: k3*S_m1_0_1
k1a=100.0Reaction: RAFK + S_2_1_0 => S_RAFK_2_1_0, Rate Law: k1a*RAFK*S_2_1_0
a2=0.5Reaction: RAFP + K_3_1 => K_RAFP_3_1, Rate Law: a2*RAFP*K_3_1
a9=20.0Reaction: K_1_1 + K_2_2 => K_K_1_1_2_2, Rate Law: a9*K_1_1*K_2_2
kon=10.0Reaction: K_1_0 + S_m1_1_m1 => S_0_1_m1, Rate Law: kon*K_1_0*S_m1_1_m1
k4=0.1Reaction: K_MEKP_2_1 => MEKP + K_2_0, Rate Law: k4*K_MEKP_2_1
koff=0.5Reaction: S_m1_0_1 => K_2_0 + S_m1_m1_1, Rate Law: koff*S_m1_0_1
d9=0.6Reaction: K_K_1_1_2_2 => K_1_1 + K_2_2, Rate Law: d9*K_K_1_1_2_2
d2=0.5Reaction: K_RAFP_3_1 => RAFP + K_3_1, Rate Law: d2*K_RAFP_3_1
k2=0.1Reaction: K_RAFP_3_1 => RAFP + K_3_0, Rate Law: k2*K_RAFP_3_1
k1=0.1Reaction: S_RAFK_2_2_0 => RAFK + S_2_2_1, Rate Law: k1*S_RAFK_2_2_0
d1a=0.0Reaction: S_RAFK_2_0_0 => RAFK + S_2_0_0, Rate Law: d1a*S_RAFK_2_0_0
kpoff=0.05Reaction: S_m1_0_1 => K_3_1 + S_m1_0_m1, Rate Law: kpoff*S_m1_0_1
k9a=0.1Reaction: S_1_2_m1 => S_2_2_m1, Rate Law: k9a*S_1_2_m1
d7=0.6Reaction: K_K_1_0_2_2 => K_1_0 + K_2_2, Rate Law: d7*K_K_1_0_2_2
k5a=0.1Reaction: S_1_1_1 => S_1_2_1, Rate Law: k5a*S_1_1_1
k9=0.1Reaction: K_K_1_1_2_2 => K_1_2 + K_2_2, Rate Law: k9*K_K_1_1_2_2
kpon=0.0Reaction: K_3_1 + S_m1_0_m1 => S_m1_0_1, Rate Law: kpon*K_3_1*S_m1_0_m1

States:

NameDescription
K 1 2[Mitogen-activated protein kinase 1]
S 0 m1 m1[Mitogen-activated protein kinase 1]
S m1 1 m1[Dual specificity mitogen-activated protein kinase kinase 1]
K K 1 1 2 2[Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
RAFK[IPR003577]
S 1 1 1[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
S RAFK m1 m1 0[RAF proto-oncogene serine/threonine-protein kinase]
S RAFK m1 0 0[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
S 1 2 0[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
S 2 2 0[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
K 2 0[Dual specificity mitogen-activated protein kinase kinase 1]
RAFPRAF phosphatase
S m1 0 0[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
K 2 2[Dual specificity mitogen-activated protein kinase kinase 1]
S 0 m1 0[RAF proto-oncogene serine/threonine-protein kinase; Mitogen-activated protein kinase 1]
S m1 0 1[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
K 1 0[Mitogen-activated protein kinase 1]
S 1 2 1[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
S 2 2 1[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
K K 2 0 3 1[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
S 1 2 m1[Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]

Lever2014 - Phenotypic models of T cell activation.: MODEL1907260003v0.0.1

This is a phenotypic model of a kinetic proofreading mechanism used to describe dynamics governing interactions between…

Details

T cell activation is a crucial checkpoint in adaptive immunity, and this activation depends on the binding parameters that govern the interactions between T cell receptors (TCRs) and peptide-MHC complexes (pMHC complexes). Despite extensive experimental studies, the relationship between the TCR-pMHC binding parameters and T cell activation remains controversial. To make sense of conflicting experimental data, a variety of verbal and mathematical models have been proposed. However, it is currently unclear which model or models are consistent or inconsistent with experimental data. A key problem is that a direct comparison between the models has not been carried out, in part because they have been formulated in different frameworks. For this Analysis article, we reformulated published models of T cell activation into phenotypic models, which allowed us to directly compare them. We find that a kinetic proofreading model that is modified to include limited signalling is consistent with the majority of published data. This model makes the intriguing prediction that the stimulation hierarchy of two different pMHC complexes (or two different TCRs that are specific for the same pMHC complex) may reverse at different pMHC concentrations. link: http://identifiers.org/pubmed/25145757

Lewkiewics2019 - effects of aging on naive T cell populations and diversity: BIOMD0000000824v0.0.1

This model is built by COPASI 4.24(Build197), based on paper: A mathematical model of the effects of aging on naive T-c…

Details

The human adaptive immune response is known to weaken in advanced age, resulting in increased severity of pathogen-born illness, poor vaccine efficacy, and a higher prevalence of cancer in the elderly. Age-related erosion of the T cell compartment has been implicated as a likely cause, but the underlying mechanisms driving this immunosenescence have not been quantitatively modeled and systematically analyzed. T cell receptor diversity, or the extent of pathogen-derived antigen responsiveness of the T cell pool, is known to diminish with age, but inherent experimental difficulties preclude accurate analysis on the full organismal level. In this paper, we formulate a mechanistic mathematical model of T cell population dynamics on the immunoclonal subpopulation level, which provides quantitative estimates of diversity. We define different estimates for diversity that depend on the individual number of cells in a specific immunoclone. We show that diversity decreases with age primarily due to diminished thymic output of new T cells and the resulting overall loss of small immunoclones. link: http://identifiers.org/pubmed/31201663

Parameters:

NameDescription
gamma = 1.8E10Reaction: => N, Rate Law: compartment*gamma
K = 1.0E10; myu_1 = 0.04; myu_0 = 0.18Reaction: myu = myu_0+myu_1*N^2/(N^2+K^2), Rate Law: missing
p = 0.17Reaction: => N, Rate Law: compartment*p*N

States:

NameDescription
myumyu
N[Natural Killer T-Cell]

Li2008 - Caulobacter Cell Cycle: BIOMD0000000718v0.0.1

This a model from the article: A Quantitative Study of the Division Cycle of Caulobacter crescentus Stalked Cells. S…

Details

Progression of a cell through the division cycle is tightly controlled at different steps to ensure the integrity of genome replication and partitioning to daughter cells. From published experimental evidence, we propose a molecular mechanism for control of the cell division cycle in Caulobacter crescentus. The mechanism, which is based on the synthesis and degradation of three "master regulator" proteins (CtrA, GcrA, and DnaA), is converted into a quantitative model, in order to study the temporal dynamics of these and other cell cycle proteins. The model accounts for important details of the physiology, biochemistry, and genetics of cell cycle control in stalked C. crescentus cell. It reproduces protein time courses in wild-type cells, mimics correctly the phenotypes of many mutant strains, and predicts the phenotypes of currently uncharacterized mutants. Since many of the proteins involved in regulating the cell cycle of C. crescentus are conserved among many genera of alpha-proteobacteria, the proposed mechanism may be applicable to other species of importance in agriculture and medicine. link: http://identifiers.org/pubmed/18225942

Parameters:

NameDescription
kd_DivK = 0.002 1/minReaction: DivK =>, Rate Law: Caulobacter*kd_DivK*DivK
Jd_CtrA_Divk_P = 0.55 1; kd_CtrA2 = 0.15 1/minReaction: CtrA => ; DivK_P, CtrA, Rate Law: Caulobacter*kd_CtrA2*DivK_P^2/(Jd_CtrA_Divk_P^2+DivK_P^2)*CtrA
ktrans_DivK_P = 0.0295 1/minReaction: DivK_P => DivK, Rate Law: Caulobacter*ktrans_DivK_P*DivK_P
km_fts = 0.4 1/min; Jm_fts = 0.95 1Reaction: hfts => ; CcrM, Rate Law: Caulobacter*km_fts*CcrM^4/(Jm_fts^4+CcrM^4)*hfts
kd_GcrA = 0.022 1/minReaction: GcrA => ; CtrA, DnaA, Rate Law: Caulobacter*kd_GcrA*GcrA
km_cori = 0.4 1/min; Jm_cori = 0.95 1Reaction: hcori => ; CcrM, Rate Law: Caulobacter*km_cori*CcrM^4/(Jm_cori^4+CcrM^4)*hcori
kd_CtrA1 = 0.002 1/minReaction: CtrA =>, Rate Law: Caulobacter*kd_CtrA1*CtrA
kd_CcrM = 0.07 1/minReaction: CcrM =>, Rate Law: Caulobacter*kd_CcrM*CcrM
ks_I = 0.08 1/minReaction: => I; CtrA, hccrM, Rate Law: Caulobacter*ks_I*CtrA*hccrM
JZring_Fts = 0.78 1; kzring_closed2 = 0.6 1/min; Ja_closed = 0.1 1Reaction: Zring => ; Fts, Rate Law: Caulobacter*kzring_closed2*(Fts/JZring_Fts)^4*Zring/(Ja_closed+Zring)
Ja_closed = 0.1 1; kzring_closed1 = 1.0E-4 1/minReaction: Zring =>, Rate Law: Caulobacter*kzring_closed1*Zring/(Ja_closed+Zring)
kd_Fts = 0.035 1/minReaction: Fts =>, Rate Law: Caulobacter*kd_Fts*Fts
Ji_GcrA_CtrA = 0.2 1; ks_GcrA = 0.045 1/minReaction: => GcrA; CtrA, DnaA, Rate Law: Caulobacter*ks_GcrA*Ji_GcrA_CtrA^2/(Ji_GcrA_CtrA^2+CtrA^2)*DnaA
Ja_DnaA_CtrA = 0.3 1; ks_DnaA = 0.0165 1/min; Ji_DnaA_GcrA = 0.5 1Reaction: => DnaA; GcrA, CtrA, hcori, Rate Law: Caulobacter*ks_DnaA*Ji_DnaA_GcrA^2/(Ji_DnaA_GcrA^2+GcrA^2)*CtrA^2/(Ja_DnaA_CtrA^2+CtrA^2)*(2-hcori)
ks_DivK = 0.0054 1/minReaction: => DivK; CtrA, Rate Law: Caulobacter*ks_DivK*CtrA
kd_I = 0.04 1/minReaction: I =>, Rate Law: Caulobacter*kd_I*I
thetacori = 2.0E-4 1; ka_Ini = 0.01 1/min; thetaGcrA = 0.45 1; thetaCtrA = 0.2 1; thetaDnaA = 0.6 1Reaction: => Ini; DnaA, GcrA, CtrA, hcori, Rate Law: Caulobacter*ka_Ini*(DnaA/thetaDnaA)^4*(GcrA/thetaGcrA)^4/(1+(DnaA/thetaDnaA)^4+(GcrA/thetaGcrA)^4+(CtrA/thetaCtrA)^4+hcori/thetacori)
km_ccrM = 0.4 1/min; Jm_ccrM = 0.95 1Reaction: hccrM => ; CcrM, Rate Law: Caulobacter*km_ccrM*CcrM^4/(Jm_ccrM^4+CcrM^4)*hccrM
ks_CtrA_P1 = 0.0083 1/min; Ji_CtrA_CtrA = 0.4 1Reaction: => CtrA; CtrA, hctrA, GcrA, Rate Law: Caulobacter*ks_CtrA_P1*Ji_CtrA_CtrA^2/(Ji_CtrA_CtrA^2+CtrA^2)*hctrA*GcrA
ks_Fts = 0.063 1/minReaction: => Fts; CtrA, hfts, Rate Law: Caulobacter*ks_Fts*CtrA*hfts
Jm_ctrA = 0.95 1; km_ctrA = 0.4 1/minReaction: hctrA => ; CcrM, Rate Law: Caulobacter*km_ctrA*CcrM^4/(Jm_ctrA^4+CcrM^4)*hctrA
Pelong = 0.05 1; Count = 2.0 1; kelong = 0.006 1/minReaction: => Elong; Elong, Rate Law: Caulobacter*kelong*Elong^4/(Elong^4+Pelong^4)*Count
ks_CtrA_P2 = 0.073 1/min; Ja_CtrA_CtrA = 0.45 1Reaction: => CtrA; CtrA, hctrA, Rate Law: Caulobacter*ks_CtrA_P2*CtrA^2/(Ja_CtrA_CtrA^2+CtrA^2)*hctrA
kd_Divk_P = 0.002 1/minReaction: DivK_P => ; DivK, Rate Law: Caulobacter*kd_Divk_P*DivK
kd_DnaA = 0.007 1/minReaction: DnaA =>, Rate Law: Caulobacter*kd_DnaA*DnaA
ks_CcrM = 0.072 1/minReaction: => CcrM; I, Rate Law: Caulobacter*ks_CcrM*I
Ja_open = 0.01 1; kzring_open = 0.8 1/minReaction: => Zring, Rate Law: Caulobacter*kzring_open*(1-Zring)/((Ja_open+1)-Zring)
ktrans_DivK = 0.5 1/minReaction: DivK => DivK_P; Zring, Rate Law: Caulobacter*ktrans_DivK*DivK*(1-Zring)

States:

NameDescription
Ini[DNA replication initiation]
GcrA[Cell cycle regulatory protein GcrA]
CtrA[Cell cycle transcriptional regulator CtrA]
hftshfts
hctrAhctrA
DNA[NCIT_C449]
CcrM[Modification methylase CcrMI]
hccrMhccrM
Fts[Cell division protein FtsQ; Cell division protein FtsZ]
Elong[DNA strand elongation]
hcorihcori
II
Zring[organelle]
DivK[Cell division response regulator DivK]
DivK P[Cell division response regulator DivK]
DnaA[Chromosomal replication initiator protein DnaA]

Li2009- Assymetric Caulobacter cell cycle: BIOMD0000000727v0.0.1

The asymmetric cell division cycle of Caulobacter crescentus is orchestrated by an elaborate gene-protein regulatory net…

Details

The asymmetric cell division cycle of Caulobacter crescentus is orchestrated by an elaborate gene-protein regulatory network, centered on three major control proteins, DnaA, GcrA and CtrA. The regulatory network is cast into a quantitative computational model to investigate in a systematic fashion how these three proteins control the relevant genetic, biochemical and physiological properties of proliferating bacteria. Different controls for both swarmer and stalked cell cycles are represented in the mathematical scheme. The model is validated against observed phenotypes of wild-type cells and relevant mutants, and it predicts the phenotypes of novel mutants and of known mutants under novel experimental conditions. Because the cell cycle control proteins of Caulobacter are conserved across many species of alpha-proteobacteria, the model we are proposing here may be applicable to other genera of importance to agriculture and medicine (e.g., Rhizobium, Brucella). link: http://identifiers.org/pubmed/19680425

Parameters:

NameDescription
Ja_i_CtrA_P = 0.5; ks_I = 0.09Reaction: => I; CtrA_P, hccrM, Rate Law: Caulobacter*ks_I*CtrA_P^2/(Ja_i_CtrA_P^2+CtrA_P^2)*hccrM
ktrans_CtrA = 0.095Reaction: CtrA => CtrA_P; CtrA, CckA_P, Rate Law: Caulobacter*ktrans_CtrA*CtrA*CckA_P
ktrans_ParAADP = 0.8Reaction: ParAADP => ; Count, ParAADP, Rate Law: Caulobacter*ktrans_ParAADP*(Count-1)*ParAADP
kd_FtsQ = 0.035Reaction: FtsQ =>, Rate Law: Caulobacter*kd_FtsQ*FtsQ
ktrans_CpdR_P = 0.5Reaction: => CpdR; CpdR_tot, CpdR, Rate Law: Caulobacter*ktrans_CpdR_P*(CpdR_tot-CpdR)
Ji_PodJL_CtrA_P = 0.6; ks_PodJL = 0.043Reaction: => PodJL; CtrA_P, GcrA, DnaA, Rate Law: Caulobacter*ks_PodJL*Ji_PodJL_CtrA_P^2/(Ji_PodJL_CtrA_P^2+CtrA_P^2)*GcrA*DnaA
H = 0.0; jsep_PodJL = 0.3; ksep_PodJL = 0.3Reaction: PodJL => ; Z, Rate Law: Caulobacter*ksep_PodJL*PodJL*H*(1-Z)/((jsep_PodJL+1)-Z)
ks_DivK = 0.0024; Ja_DivK = 0.06Reaction: => DivK; CtrA_P, Rate Law: Caulobacter*ks_DivK*CtrA_P^2/(Ja_DivK^2+CtrA_P^2)
H = 0.0; JDivk_P_PodJL = 0.3; ktrans_DivK_P = 0.6Reaction: DivK_P => DivK; DivK_P, PodJL, Z, Rate Law: Caulobacter*ktrans_DivK_P*DivK_P*PodJL^2/(JDivk_P_PodJL^2+PodJL^2)*(1+H*(Z-1))
ks_Zring = 0.035Reaction: => Zring; Zring, FtsZ, Rate Law: Caulobacter*ks_Zring*(1-Zring)*FtsZ
thethaCtrA_P = 0.5; thethaCori = 0.05; ka_Ini = 0.01; thethaDnaA = 0.65; thethaGcrA = 0.65Reaction: => Ini; DnaA, GcrA, CtrA_P, hcori, Count, Rate Law: Caulobacter*ka_Ini*(DnaA/thethaDnaA)^4*(GcrA/thethaGcrA)^4/(1+(DnaA/thethaDnaA)^4+(GcrA/thethaGcrA)^4+(CtrA_P/thethaCtrA_P)^4+(GcrA/thethaGcrA)^4*(CtrA_P/thethaCtrA_P)^4+(hcori/thethaCori)^4)*Count
kd_CcrM = 0.07Reaction: CcrM =>, Rate Law: Caulobacter*kd_CcrM*CcrM
ks_CtrA_P2 = 0.14; Ja_CtrA_CtrA_P = 0.45Reaction: => CtrA; CtrA_P, hctrA, Rate Law: Caulobacter*ks_CtrA_P2*CtrA_P^2/(Ja_CtrA_CtrA_P^2+CtrA_P^2)*hctrA
ks_DnaA1 = 0.0031; JiDnaA_GcrA = 0.6Reaction: => DnaA; GcrA, hcori, Rate Law: Caulobacter*ks_DnaA1*JiDnaA_GcrA^2/(JiDnaA_GcrA^2+GcrA^2)*(2-hcori)
kd_RcdA = 0.017Reaction: RcdA =>, Rate Law: Caulobacter*kd_RcdA*RcdA
kd_PodJL1 = 0.05Reaction: PodJL =>, Rate Law: Caulobacter*kd_PodJL1*PodJL
kd_DivJ = 0.002Reaction: DivJ =>, Rate Law: Caulobacter*kd_DivJ*DivJ
Ji_Ccka_DivK_P = 0.3; ktrans_CckA = 0.2Reaction: => CckA_P; CckA_tot, CckA_P, DivK_P, Rate Law: Caulobacter*ktrans_CckA*(CckA_tot-CckA_P)*Ji_Ccka_DivK_P^2/(Ji_Ccka_DivK_P^2+DivK_P^2)
ktrans_CckA_P = 0.05Reaction: CckA_P =>, Rate Law: Caulobacter*ktrans_CckA_P*CckA_P
Ji_CtrA_CtrA_P = 0.4; ks_CtrA_P1 = 0.0159Reaction: => CtrA; CtrA_P, GcrA, hctrA, Rate Law: Caulobacter*ks_CtrA_P1*Ji_CtrA_CtrA_P^2/(Ji_CtrA_CtrA_P^2+CtrA_P^2)*GcrA*hctrA
Jm_ctrA = 0.95; km_ctrA = 0.4Reaction: hctrA => ; CcrM, Rate Law: Caulobacter*km_ctrA*CcrM^4/(Jm_ctrA^4+CcrM^4)*hctrA
ks_PerP = 0.04Reaction: => PerP; CtrA_P, PodJL, Rate Law: Caulobacter*ks_PerP*CtrA_P*PodJL
km_Cori = 0.4; Jm_Cori = 0.95Reaction: hcori => ; CcrM, Rate Law: Caulobacter*km_Cori*CcrM^4/(Jm_Cori^4+CcrM^4)*hcori
Jd_PodJL_PerP = 0.45; kd_PodJL2 = 0.002Reaction: PodJL => ; PerP, PodJL, Rate Law: Caulobacter*kd_PodJL2*PerP^2/(Jd_PodJL_PerP^2+PerP^2)*PodJL
ktrans_ParAATP = 0.5Reaction: => ParAADP; ParA_tot, ParAADP, Rate Law: Caulobacter*ktrans_ParAATP*(ParA_tot-ParAADP)
kd_DnaA = 0.007Reaction: DnaA =>, Rate Law: Caulobacter*kd_DnaA*DnaA
ks_FtsZ = 0.036; JiFtsZ_CtrA_P = 0.7Reaction: => FtsZ; CtrA_P, DnaA, hftsZ, Rate Law: Caulobacter*ks_FtsZ*JiFtsZ_CtrA_P^2/(JiFtsZ_CtrA_P^2+CtrA_P^2)*DnaA*(1-hftsZ)
Pelong = 0.05; kelong = 0.0065Reaction: => Elong; Elong, Count, Rate Law: Caulobacter*kelong*Elong^4/(Pelong^4+Elong^4)*Count
kd_FtsZ2 = 0.02Reaction: FtsZ => ; Zring, FtsZ, Rate Law: Caulobacter*kd_FtsZ2*(1-Zring)*FtsZ
kd_FtsZ1 = 0.009Reaction: FtsZ =>, Rate Law: Caulobacter*kd_FtsZ1*FtsZ
ks_RcdA = 0.023; Ja_RcdA_CtrA_P = 0.4Reaction: => RcdA; CtrA_P, Rate Law: Caulobacter*ks_RcdA*CtrA_P^2/(Ja_RcdA_CtrA_P^2+CtrA_P^2)
kZ_closed1 = 1.0E-4; thethaParAADP = 0.3; thethaZring = 0.3; kZ_closed2 = 1.6; JZ_FtsQ = 0.8; Ja_closed = 0.05Reaction: Z => ; FtsQ, Zring, ParAADP, Rate Law: Caulobacter*(kZ_closed1+kZ_closed2*FtsQ^4/(JZ_FtsQ^4+FtsQ^4)*(Zring/thethaZring)^4/(1+(Zring/thethaZring)^4+(ParAADP/thethaParAADP)^4))*Z/(Ja_closed+Z)
H = 0.0; ksep_PerP = 0.011; Jsep_PerP = 0.3Reaction: PerP => ; Z, Rate Law: Caulobacter*ksep_PerP*PerP*H*(1-Z)/((Jsep_PerP+1)-Z)
kd_PerP = 0.02Reaction: PerP =>, Rate Law: Caulobacter*kd_PerP*PerP
ks_CcrM = 0.072Reaction: => CcrM; I, Rate Law: Caulobacter*ks_CcrM*I
kd_CtrA1 = 0.002Reaction: CtrA =>, Rate Law: Caulobacter*kd_CtrA1*CtrA
ktrans_DivK = 0.15; H = 0.0; JDivk_DivJ = 0.3Reaction: DivK => DivK_P; DivK, DivJ, Z, Rate Law: Caulobacter*ktrans_DivK*DivK*DivJ^2/(JDivk_DivJ^2+DivJ^2)*(Z+H*(1-Z))
km_ftsz = 0.4; Jm_ftsZ = 0.95Reaction: hftsZ => ; CcrM, Rate Law: Caulobacter*km_ftsz*CcrM^4/(Jm_ftsZ^4+CcrM^4)*hftsZ
H = 0.0; Jsep_DivJ = 0.3; ksep_divJ = 0.3Reaction: DivJ => ; DivJ, Z, Rate Law: Caulobacter*ksep_divJ*DivJ*(1-H)*(1-Z)/((Jsep_DivJ+1)-Z)
kd_FtsZ3 = 0.3Reaction: FtsZ => ; Z, FtsZ, Rate Law: Caulobacter*kd_FtsZ3*(1-Z)*FtsZ
ks_DnaA2 = 0.0022; Ja_Dna_CtrA_P = 0.3Reaction: => DnaA; CtrA_P, hcori, Rate Law: Caulobacter*ks_DnaA2*CtrA_P^2/(Ja_Dna_CtrA_P^2+CtrA_P^2)*(2-hcori)
ktrans_CpdR = 0.6; Ja_CpdR_CckA_P = 0.8Reaction: CpdR => ; CpdR, CckA_P, Rate Law: Caulobacter*ktrans_CpdR*CpdR*CckA_P^2/(Ja_CpdR_CckA_P^2+CckA_P^2)
Ja_open = 0.01; kZ_open = 0.8Reaction: => Z; Z, Rate Law: Caulobacter*kZ_open*(1-Z)/((Ja_open+1)-Z)
kd_GcrA = 0.022Reaction: GcrA =>, Rate Law: Caulobacter*kd_GcrA*GcrA
H = 0.0; Ji_DivJ_PodJL = 0.13; ks_DivJ2 = 0.025Reaction: => DivJ; PodJL, Rate Law: Caulobacter*(1-H)*ks_DivJ2*Ji_DivJ_PodJL^2/(Ji_DivJ_PodJL^2+PodJL^2)
Jd_CtrA_DivK_P = 0.55; jd_CtrA_RcdA = 0.5; kd_CtrA2 = 0.25; jd_CtrA_CpdR = 0.6Reaction: CtrA => ; DivK_P, CpdR, RcdA, CtrA, Rate Law: Caulobacter*kd_CtrA2*DivK_P^2/(Jd_CtrA_DivK_P^2+DivK_P^2)*CpdR^4/(jd_CtrA_CpdR^4+CpdR^4)*RcdA^4/(jd_CtrA_RcdA^4+RcdA^4)*CtrA
Ja_FtsQ_DNA = 0.05; ks_FtsQ = 0.06; Ja_FtsQ_CtrA_P = 0.5Reaction: => FtsQ; CtrA_P, hcori, Rate Law: Caulobacter*ks_FtsQ*CtrA_P^2/(Ja_FtsQ_CtrA_P^2+CtrA_P^2)*hcori^4/(Ja_FtsQ_DNA^4+hcori^4)
ktrans_CtrA_P = 0.025Reaction: CtrA_P => CtrA, Rate Law: Caulobacter*ktrans_CtrA_P*CtrA_P
Ji_GcrA_CtrA = 0.4; ks_GcrA = 0.055Reaction: => GcrA; CtrA_P, DnaA, Rate Law: Caulobacter*ks_GcrA*Ji_GcrA_CtrA^2/(Ji_GcrA_CtrA^2+CtrA_P^2)*DnaA
kd_DivK = 0.002Reaction: DivK =>, Rate Law: Caulobacter*kd_DivK*DivK
km_ccrM = 0.4; jm_ccrM = 0.95Reaction: hccrM => ; CcrM, Rate Law: Caulobacter*km_ccrM*CcrM^4/(jm_ccrM^4+CcrM^4)*hccrM
ks_DivJ1 = 0.002Reaction: => DivJ, Rate Law: Caulobacter*ks_DivJ1
kd_I = 0.04Reaction: I =>, Rate Law: Caulobacter*kd_I*I

States:

NameDescription
ZZ
CckA P[Sensory transduction histidine kinase/receiver protein CckA2.7.13.3]
CtrA[Cell cycle transcriptional regulator CtrA]
hftsZ[Ratio]
hctrA[Ratio]
CcrM[Modification methylase CcrMI]
RcdA[Regulator of CtrA degradation]
hccrM[Ratio]
FtsQ[Cell division protein FtsQ]
Elong[DNA strand elongation]
Zring[organelle]
DivK[Cell division response regulator DivK]
CpdR[CpdR]
ParAADP[Chromosome partitioning protein ParA]
CtrA P[Cell cycle transcriptional regulator CtrA]
DnaA[Chromosomal replication initiator protein DnaA]
Ini[DNA replication initiation]
GcrA[Cell cycle regulatory protein GcrA]
DivJ[Histidine protein kinase DivJ2.7.13.3]
FtsZ[Cell division protein FtsZ]
DNA[DNA]
hcori[Ratio]
II
PodJL[Localization factor PodJL]
PerP[Aspartyl protease perP]
DivK P[Phosphorylated Peptide; Cell division response regulator DivK]

Li2010_YeastGlycolysis: MODEL1012110001v0.0.1

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

Details

The behaviour of biological systems can be deduced from their mathematical models. However, multiple sources of data in diverse forms are required in the construction of a model in order to define its components and their biochemical reactions, and corresponding parameters. Automating the assembly and use of systems biology models is dependent upon data integration processes involving the interoperation of data and analytical resources.Taverna workflows have been developed for the automated assembly of quantitative parameterised metabolic networks in the Systems Biology Markup Language (SBML). A SBML model is built in a systematic fashion by the workflows which starts with the construction of a qualitative network using data from a MIRIAM-compliant genome-scale model of yeast metabolism. This is followed by parameterisation of the SBML model with experimental data from two repositories, the SABIO-RK enzyme kinetics database and a database of quantitative experimental results. The models are then calibrated and simulated in workflows that call out to COPASIWS, the web service interface to the COPASI software application for analysing biochemical networks. These systems biology workflows were evaluated for their ability to construct a parameterised model of yeast glycolysis.Distributed information about metabolic reactions that have been described to MIRIAM standards enables the automated assembly of quantitative systems biology models of metabolic networks based on user-defined criteria. Such data integration processes can be implemented as Taverna workflows to provide a rapid overview of the components and their relationships within a biochemical system. link: http://identifiers.org/pubmed/21114840

Li2012 Calcium mediated synaptic plasticity: BIOMD0000000628v0.0.1

Li2012 Calcium mediated synaptic plasticityThis model is an extension of  [BIOMD0000000183](http://www.ebi.ac.uk/biomod…

Details

NMDA receptor dependent long-term potentiation (LTP) and long-term depression (LTD) are two prominent forms of synaptic plasticity, both of which are triggered by post-synaptic calcium elevation. To understand how calcium selectively stimulates two opposing processes, we developed a detailed computational model and performed simulations with different calcium input frequencies, amplitudes, and durations. We show that with a total amount of calcium ions kept constant, high frequencies of calcium pulses stimulate calmodulin more efficiently. Calcium input activates both calcineurin and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) at all frequencies, but increased frequencies shift the relative activation from calcineurin to CaMKII. Irrespective of amplitude and duration of the inputs, the total amount of calcium ions injected adjusts the sensitivity of the system to calcium input frequencies. At a given frequency, the quantity of CaMKII activated is proportional to the total amount of calcium. Thus, an input of a small amount of calcium at high frequencies can induce the same activation of CaMKII as a larger amount, at lower frequencies. Finally, the extent of activation of CaMKII signals with high calcium frequency is further controlled by other factors, including the availability of calmodulin, and by the potency of phosphatase inhibitors. link: http://identifiers.org/pubmed/22962589

Parameters:

NameDescription
K_CamR_to_T_Ca1 = 62928.5313294502Reaction: CamR_Ca1_D => CamT_Ca1_D, Rate Law: Spine*K_CamR_to_T_Ca1*CamR_Ca1_D
K_D_PKA_on = 5600000.0Reaction: D + PKA => D_PKA, Rate Law: Spine*K_D_PKA_on*D*PKA
K_CamT_Ca_D_off = 5.12626262626263Reaction: CamT_Ca1_D => CamT + Ca, Rate Law: Spine*K_CamT_Ca_D_off*CamT_Ca1_D
K_CamT_Ca_A_off = 2941.41414141414Reaction: CamT_Ca2_AD => CamT_Ca1_D + Ca, Rate Law: Spine*K_CamT_Ca_A_off*CamT_Ca2_AD
K_CamR_Ca_C_off = 24.36Reaction: CamR_Ca3_ACD_CaMKII => CamR_Ca2_AD_CaMKII + Ca, Rate Law: Spine*K_CamR_Ca_C_off*CamR_Ca3_ACD_CaMKII
K_CamR_PP2B_on = 4.6E7Reaction: CamR_Ca2_BD + PP2B => CamR_Ca2_BD_PP2B, Rate Law: Spine*K_CamR_PP2B_on*CamR_Ca2_BD*PP2B
K_CamR_Ca_D_off = 0.0203Reaction: CamR_Ca2_BD => CamR_Ca1_B + Ca, Rate Law: Spine*K_CamR_Ca_D_off*CamR_Ca2_BD
K_CaMKII_autoPhosphorylation = 0.0Reaction: CamR_Ca4_ABCD_CaMKII => CamR_Ca4_ABCD_CaMKIIp, Rate Law: Spine*K_CaMKII_autoPhosphorylation*CamR_Ca4_ABCD_CaMKII
K_CamT_to_R_Ca2 = 12216.9934343434Reaction: CamT_Ca2_BD => CamR_Ca2_BD, Rate Law: Spine*K_CamT_to_R_Ca2*CamT_Ca2_BD
K_Cam_Ca_on = 1400000.0Reaction: CamR_Ca1_B + Ca => CamR_Ca2_BD, Rate Law: Spine*K_Cam_Ca_on*CamR_Ca1_B*Ca
K_CamR_PP2B_off = 0.4Reaction: CamR_PP2B => CamR + PP2B, Rate Law: Spine*K_CamR_PP2B_off*CamR_PP2B
K_CamR_Ca_B_off = 0.02324Reaction: CamR_Ca1_B_PP2B => CamR_PP2B + Ca, Rate Law: Spine*K_CamR_Ca_B_off*CamR_Ca1_B_PP2B
K_CamR_PP2B_D_off = 0.2Reaction: Dp_CamR_Ca2_AC_PP2B => D + CamR_Ca2_AC_PP2B, Rate Law: Spine*K_CamR_PP2B_D_off*Dp_CamR_Ca2_AC_PP2B
K_CamMKIIp_PP1a_on = 3000000.0Reaction: CamR_Ca2_CD_CaMKIIp + PP1a => CamR_Ca2_CD_CaMKIIp_PP1a, Rate Law: Spine*K_CamMKIIp_PP1a_on*CamR_Ca2_CD_CaMKIIp*PP1a
K_CamT_to_R_Ca1 = 768.797448718562Reaction: CamT_Ca1_D => CamR_Ca1_D, Rate Law: Spine*K_CamT_to_R_Ca1*CamT_Ca1_D
K_CamT_Ca_C_off = 6151.51515151515Reaction: CamT_Ca3_ABC => CamT_Ca2_AB + Ca, Rate Law: Spine*K_CamT_Ca_C_off*CamT_Ca3_ABC
K_CamT_Ca_B_off = 5.86868686868687Reaction: CamT_Ca3_ABD => CamT_Ca2_AD + Ca, Rate Law: Spine*K_CamT_Ca_B_off*CamT_Ca3_ABD
K_CamMKIIp_PP1a_p_off = 2.0Reaction: CamR_Ca1_B_CaMKIIp_PP1a => CamR_Ca1_B_CaMKII + PP1a, Rate Law: Spine*K_CamMKIIp_PP1a_p_off*CamR_Ca1_B_CaMKIIp_PP1a
K_CamR_PP2B_Dp_on = 4100000.0Reaction: Dp + CamR_PP2B => Dp_CamR_PP2B, Rate Law: Spine*K_CamR_PP2B_Dp_on*Dp*CamR_PP2B
K_CamR_PP2B_Dp_off = 6.4Reaction: Dp_CamR_PP2B => Dp + CamR_PP2B, Rate Law: Spine*K_CamR_PP2B_Dp_off*Dp_CamR_PP2B
K_CamMKIIp_PP1a_off = 0.5Reaction: CamR_Ca2_CD_CaMKIIp_PP1a => CamR_Ca2_CD_CaMKIIp + PP1a, Rate Law: Spine*K_CamMKIIp_PP1a_off*CamR_Ca2_CD_CaMKIIp_PP1a
K_CamR_CaMKIIp_off = 0.001Reaction: CamR_Ca3_ABD_CaMKIIp => CamR_Ca3_ABD + CaMKIIp, Rate Law: Spine*K_CamR_CaMKIIp_off*CamR_Ca3_ABD_CaMKIIp
K_CamR_CaMKII_p_on = 3200000.0Reaction: CamR_Ca2_BD + CaMKII => CamR_Ca2_BD_CaMKII, Rate Law: Spine*K_CamR_CaMKII_p_on*CamR_Ca2_BD*CaMKII
K_CamR_Ca_A_off = 11.648Reaction: CamR_Ca1_A_PP2B => CamR_PP2B + Ca, Rate Law: Spine*K_CamR_Ca_A_off*CamR_Ca1_A_PP2B
K_CamR_CaMKII_off = 0.343Reaction: CamR_Ca2_BD_CaMKII => CamR_Ca2_BD + CaMKII, Rate Law: Spine*K_CamR_CaMKII_off*CamR_Ca2_BD_CaMKII

States:

NameDescription
CamR Ca2 BD CaMKIICamR_Ca2_BD_CaMKII
CamT Ca1 DCamT_Ca1_D
CamR Ca4 ABCD CaMKII[calcium(2+); Calcium/calmodulin-dependent protein kinase type II subunit alpha; Calmodulin-3Calmodulin-1Calmodulin-2]
CamR Ca1 B CaMKII[calcium(2+); Calcium/calmodulin-dependent protein kinase type II subunit alpha; Calmodulin-3Calmodulin-1Calmodulin-2]
CamR Ca2 BC CaMKIICamR_Ca2_BC_CaMKII
CamT Ca2 AD[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2]
CamR CaMKII[calcium(2+); Calcium/calmodulin-dependent protein kinase type II subunit alpha]
CamT Ca2 ACCamT_Ca2_AC
PP1a[Protein phosphatase 1F]
CaMKII[Calcium/calmodulin-dependent protein kinase type II subunit alpha]
CamT Ca2 AB[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2]
PKA[cAMP-dependent protein kinase catalytic subunit alpha]
CamR Ca2 CD CaMKIICamR_Ca2_CD_CaMKII
D[Protein phosphatase 1 regulatory subunit 1B]
CamR Ca3 ABDCamR_Ca3_ABD
CamR PP2B[Calmodulin-3Calmodulin-1Calmodulin-2; urn:miriam:reactome:R-HSA-140202]
CamR Ca3 ACDCamR_Ca3_ACD
CamR Ca2 BDCamR_Ca2_BD
CamR CaMKIIp[urn:miriam:kegg:C00562; Calcium/calmodulin-dependent protein kinase type II subunit alpha; Calmodulin-3Calmodulin-1Calmodulin-2]
CamR Ca1 C CaMKIICamR_Ca1_C_CaMKII
CamR Ca1 A CaMKII[calcium(2+); Calcium/calmodulin-dependent protein kinase type II subunit alpha; Calmodulin-3Calmodulin-1Calmodulin-2]
Ca[calcium(2+)]
CamR Ca1 A CaMKIIp[urn:miriam:kegg:C00562; calcium(2+); Calcium/calmodulin-dependent protein kinase type II subunit alpha; Calmodulin-3Calmodulin-1Calmodulin-2]
CamR Ca3 ACD CaMKIICamR_Ca3_ACD_CaMKII

Li2016 - Model for pancreatic cancer patients receiving immunotherapy: BIOMD0000000929v0.0.1

immunotherapy offers a better prognosis for pancreatic cancer patients. As a direct extension of this work, various new…

Details

Pancreatic cancer is one of the most deadly types of cancer since it typically spreads rapidly and can seldom be detected in its early stage. Pancreatic cancer therapy is thus a challenging task, and appropriate prognosis or assessment for pancreatic cancer therapy is of critical importance. In this work, based on available clinical data in Niu et al. (2013) we develop a mathematical prognosis model that can predict the overall survival of pancreatic cancer patients who receive immunotherapy. The mathematical model incorporates pancreatic cancer cells, pancreatic stellate cells, three major classes of immune effector cells CD8+ T cells, natural killer cells, helper T cells, and two major classes of cytokines interleukin-2 (IL-2) and interferon-γ (IFN-γ). The proposed model describes the dynamic interaction between tumor and immune cells. In order for the model to be able to generate appropriate prognostic results for disease progression, the distribution and stability properties of equilibria in the mathematical model are computed and analysed in absence of treatments. In addition, numerical simulations for disease progression with or without treatments are performed. It turns out that the median overall survival associated with CIK immunotherapy is prolonged from 7 to 13months compared with the survival without treatment, this is consistent with the clinical data observed in Niu et al. (2013). The validity of the proposed mathematical prognosis model is thus verified. Our study confirms that immunotherapy offers a better prognosis for pancreatic cancer patients. As a direct extension of this work, various new therapy methods that are under exploration and clinical trials could be assessed or evaluated using the newly developed mathematical prognosis model. link: http://identifiers.org/pubmed/27338302

Parameters:

NameDescription
a_p = 0.00195; b_p = 1.7857E-9Reaction: => Pancreatic_stellate_cells__P, Rate Law: Pancreas*a_p*Pancreatic_stellate_cells__P*(1-b_p*Pancreatic_stellate_cells__P)
b_t = 0.02Reaction: CD8__T_cells__T =>, Rate Law: Pancreas*b_t*CD8__T_cells__T
a_h = 9600.0Reaction: => helper_T_cells__H, Rate Law: Pancreas*a_h
g_h = 0.3; tau1_alpha1 = 2.2483E11; p_h = 0.125Reaction: => helper_T_cells__H; helper_T_cells__H, Rate Law: Pancreas*p_h*helper_T_cells__H*helper_T_cells__H/(g_h*tau1_alpha1+helper_T_cells__H)
a_n = 130000.0Reaction: => NK_cells__N, Rate Law: Pancreas*a_n
a_c = 0.0195; b_c = 1.02E-11Reaction: => Pancreatic_cancer_cells__C, Rate Law: Pancreas*a_c*Pancreatic_cancer_cells__C*(1-b_c*Pancreatic_cancer_cells__C)
a_t = 3500.0Reaction: => CD8__T_cells__T, Rate Law: Pancreas*a_t
beta2_tau2 = 4.4691E-13; gamma2_tau2 = 4.4691E-13; f_h = 0.125; h_h = 0.3; alpha2_tau2 = 4.4691E-13Reaction: => helper_T_cells__H; CD8__T_cells__T, NK_cells__N, helper_T_cells__H, Rate Law: Pancreas*f_h*(alpha2_tau2*CD8__T_cells__T+beta2_tau2*NK_cells__N+gamma2_tau2*helper_T_cells__H)*helper_T_cells__H/(h_h+alpha2_tau2*CD8__T_cells__T+beta2_tau2*NK_cells__N+gamma2_tau2*helper_T_cells__H)
b_h = 0.0012Reaction: helper_T_cells__H =>, Rate Law: Pancreas*b_h*helper_T_cells__H
p_t = 0.125; tau1_alpha1 = 2.2483E11; g_t = 0.3Reaction: => CD8__T_cells__T; helper_T_cells__H, Rate Law: Pancreas*p_t*helper_T_cells__H*CD8__T_cells__T/(g_t*tau1_alpha1+helper_T_cells__H)
c_n = 1.0E-13Reaction: NK_cells__N => ; Pancreatic_cancer_cells__C, Rate Law: Pancreas*c_n*Pancreatic_cancer_cells__C*NK_cells__N
c_c = 1.5E-11Reaction: Pancreatic_cancer_cells__C => ; NK_cells__N, Rate Law: Pancreas*c_c*NK_cells__N*Pancreatic_cancer_cells__C
beta2_tau2 = 4.4691E-13; gamma2_tau2 = 4.4691E-13; f_t = 0.125; h_t = 0.3; alpha2_tau2 = 4.4691E-13Reaction: => CD8__T_cells__T; CD8__T_cells__T, NK_cells__N, helper_T_cells__H, Rate Law: Pancreas*f_t*(alpha2_tau2*CD8__T_cells__T+beta2_tau2*NK_cells__N+gamma2_tau2*helper_T_cells__H)*CD8__T_cells__T/(h_t+alpha2_tau2*CD8__T_cells__T+beta2_tau2*NK_cells__N+gamma2_tau2*helper_T_cells__H)
lambda_p = 0.015Reaction: Pancreatic_stellate_cells__P =>, Rate Law: Pancreas*lambda_p*Pancreatic_stellate_cells__P
e_t = 5.0E-12Reaction: => CD8__T_cells__T; NK_cells__N, Pancreatic_cancer_cells__C, Rate Law: Pancreas*e_t*NK_cells__N*Pancreatic_cancer_cells__C
b_p = 1.7857E-9; f_p = 0.125; mu_p = 5.6E7Reaction: => Pancreatic_stellate_cells__P; Pancreatic_cancer_cells__C, Rate Law: Pancreas*f_p*Pancreatic_cancer_cells__C/(mu_p+Pancreatic_cancer_cells__C)*Pancreatic_stellate_cells__P*(1-b_p*Pancreatic_stellate_cells__P)
d_t = 3.0E-10Reaction: CD8__T_cells__T =>, Rate Law: Pancreas*d_t*CD8__T_cells__T^2
mu_c = 3.482115E-12; b_c = 1.02E-11Reaction: => Pancreatic_cancer_cells__C; Pancreatic_stellate_cells__P, Rate Law: Pancreas*mu_c*Pancreatic_stellate_cells__P*Pancreatic_cancer_cells__C*(1-b_c*Pancreatic_cancer_cells__C)
g_n = 0.3; tau1_alpha1 = 2.2483E11; p_n = 0.125Reaction: => NK_cells__N; helper_T_cells__H, Rate Law: Pancreas*p_n*helper_T_cells__H*NK_cells__N/(g_n*tau1_alpha1+helper_T_cells__H)
c_t = 3.42E-12Reaction: CD8__T_cells__T => ; Pancreatic_cancer_cells__C, Rate Law: Pancreas*c_t*CD8__T_cells__T*Pancreatic_cancer_cells__C
l = 0.666666666666667; s = 0.3; d_c = 0.005Reaction: Pancreatic_cancer_cells__C => ; CD8__T_cells__T, Rate Law: Pancreas*d_c*(CD8__T_cells__T/Pancreatic_cancer_cells__C)^l/(s+(CD8__T_cells__T/Pancreatic_cancer_cells__C)^l)*Pancreatic_cancer_cells__C
b_n = 0.015Reaction: NK_cells__N =>, Rate Law: Pancreas*b_n*NK_cells__N
c_h = 5.0E-11Reaction: helper_T_cells__H =>, Rate Law: Pancreas*c_h*helper_T_cells__H^2
beta2_tau2 = 4.4691E-13; gamma2_tau2 = 4.4691E-13; f_n = 0.125; h_n = 0.3; alpha2_tau2 = 4.4691E-13Reaction: => NK_cells__N; CD8__T_cells__T, NK_cells__N, helper_T_cells__H, Rate Law: Pancreas*f_n*(alpha2_tau2*CD8__T_cells__T+beta2_tau2*NK_cells__N+gamma2_tau2*helper_T_cells__H)*NK_cells__N/(h_n+alpha2_tau2*CD8__T_cells__T+beta2_tau2*NK_cells__N+gamma2_tau2*helper_T_cells__H)

States:

NameDescription
Pancreatic stellate cells P[Pancreatic Stellate Cell]
NK cells N[Natural Killer Cell]
Pancreatic cancer cells C[pancreatic cancer cell]
CD8 T cells T[0004219]
helper T cells Hhelper T cells (H)

Li2019 - Neurogranin stimulates Ca/calmodulin-dependent kinase II by inhibiting Calcineurin: MODEL1903010001v0.0.1

Calmodulin sits at the center of molecular mechanisms underlying learning and memory. Its complex, and sometimes opposit…

Details

Calmodulin sits at the centre of molecular mechanisms underlying learning and memory. Its complex, and sometimes opposite influences, via the binding to various proteins, are yet to be fully understood. Calcium/calmodulin-dependent protein kinase II (CaMKII) and calcineurin (CaN) both bind open calmodulin, favouring Long-term potentiation (LTP) or depression (LTD) respectively. Neurogranin binds to the closed conformation of calmodulin and its impact on synaptic plasticity is less clear. We set up a mechanistic computational model based on allosteric principles to simulate calmodulin state transitions and its interaction with calcium ions and the three binding partners mentioned above. We simulated calcium spikes at various frequencies and show that neurogranin regulates synaptic plasticity along three modalities. At low spike frequencies, neurogranin inhibits the onset of LTD by limiting CaN activation. At intermediate frequencies, neurogranin limits LTP by precluding binding of CaMKII with calmodulin. Finally, at high spike frequencies, neurogranin promotes LTP by enhancing CaMKII autophosphorylation. While neurogranin might act as a calmodulin buffer, it does not significantly preclude the calmodulin opening by calcium. On the contrary, neurogranin synchronizes the opening of calmodulin’s two lobes and promotes their activation at specific frequencies, increasing the chance of CaMKII trans-autophosphorylation. Taken together, our study reveals dynamic regulatory roles played by neurogranin on synaptic plasticity, which provide mechanistic explanations to opposing experimental findings. link: http://identifiers.org/doi/10.1101/597278

Liao2011 - Genome-scale metabolic reconstruction of Klebsiella pneumoniae (iYL1228): MODEL1507180054v0.0.1

Liao2011 - Genome-scale metabolic reconstruction of Klebsiella pneumoniae (iYL1228)This model is described in the articl…

Details

Klebsiella pneumoniae is a Gram-negative bacterium of the family Enterobacteriaceae that possesses diverse metabolic capabilities: many strains are leading causes of hospital-acquired infections that are often refractory to multiple antibiotics, yet other strains are metabolically engineered and used for production of commercially valuable chemicals. To study its metabolism, we constructed a genome-scale metabolic model (iYL1228) for strain MGH 78578, experimentally determined its biomass composition, experimentally determined its ability to grow on a broad range of carbon, nitrogen, phosphorus and sulfur sources, and assessed the ability of the model to accurately simulate growth versus no growth on these substrates. The model contains 1,228 genes encoding 1,188 enzymes that catalyze 1,970 reactions and accurately simulates growth on 84% of the substrates tested. Furthermore, quantitative comparison of growth rates between the model and experimental data for nine of the substrates also showed good agreement. The genome-scale metabolic reconstruction for K. pneumoniae presented here thus provides an experimentally validated in silico platform for further studies of this important industrial and biomedical organism. link: http://identifiers.org/pubmed/21296962

Liebal2012 - B.subtilis post-transcriptional instability model: BIOMD0000000459v0.0.1

Liebal2012 - B.subtilis post-transcription instability modelAn important transcription factor of B.subsilis is sigma B…

Details

In Bacillus subtilis the σ(B) mediated general stress response provides protection against various environmental and energy related stress conditions. To better understand the general stress response, we need to explore the mechanism by which the components interact. Here, we performed experiments in B. subtilis wild type and mutant strains to test and validate a mathematical model of the dynamics of σ(B) activity. In the mutant strain BSA115, σ(B) transcription is inducible by the addition of IPTG and negative control of σ(B) activity by the anti-sigma factor RsbW is absent. In contrast to our expectations of a continuous β-galactosidase activity from a ctc::lacZ fusion, we observed a transient activity in the mutant. To explain this experimental finding, we constructed mathematical models reflecting different hypotheses regarding the regulation of σ(B) and β-galactosidase dynamics. Only the model assuming instability of either ctc::lacZ mRNA or β-galactosidase protein is able to reproduce the experiments in silico. Subsequent Northern blot experiments revealed stable high-level ctc::lacZ mRNA concentrations after the induction of the σ(B) response. Therefore, we conclude that protein instability following σ(B) activation is the most likely explanation for the experimental observations. Our results thus support the idea that B. subtilis increases the cytoplasmic proteolytic degradation to adapt the proteome in face of environmental challenges following activation of the general stress response. The findings also have practical implications for the analysis of stress response dynamics using lacZ reporter gene fusions, a frequently used strategy for the σ(B) response. link: http://identifiers.org/pubmed/22511268

Parameters:

NameDescription
kxs = 9.32517E-8; kxd = 1.08559E-9Reaction: sigb => x; sigb, x, Rate Law: kxs*sigb-kxd*x
kbd = 0.0164812; kbs = 100.0Reaction: IPTG => sigb; IPTG, sigb, Rate Law: IPTG*kbs-kbd*sigb
kzd = 1.34615E-7; kzx = 0.00317772; kzs = 9.03538E-7Reaction: sigb => lacz; x, sigb, lacz, x, Rate Law: kzs*sigb-lacz*(kzd+kzx*x)

States:

NameDescription
sigb[IPR006288]
lacz[galactosidase activity]
x[inhibitor]
IPTG[isopropyl beta-D-thiogalactopyranoside]

Liebal2012 - B.subtilis sigB proteolysis model: BIOMD0000000460v0.0.1

Liebal2012 - B.subtilis sigB proteolysis modelAn important transcription factor of B.subsilis is sigma B . Liebal et al…

Details

In Bacillus subtilis the σ(B) mediated general stress response provides protection against various environmental and energy related stress conditions. To better understand the general stress response, we need to explore the mechanism by which the components interact. Here, we performed experiments in B. subtilis wild type and mutant strains to test and validate a mathematical model of the dynamics of σ(B) activity. In the mutant strain BSA115, σ(B) transcription is inducible by the addition of IPTG and negative control of σ(B) activity by the anti-sigma factor RsbW is absent. In contrast to our expectations of a continuous β-galactosidase activity from a ctc::lacZ fusion, we observed a transient activity in the mutant. To explain this experimental finding, we constructed mathematical models reflecting different hypotheses regarding the regulation of σ(B) and β-galactosidase dynamics. Only the model assuming instability of either ctc::lacZ mRNA or β-galactosidase protein is able to reproduce the experiments in silico. Subsequent Northern blot experiments revealed stable high-level ctc::lacZ mRNA concentrations after the induction of the σ(B) response. Therefore, we conclude that protein instability following σ(B) activation is the most likely explanation for the experimental observations. Our results thus support the idea that B. subtilis increases the cytoplasmic proteolytic degradation to adapt the proteome in face of environmental challenges following activation of the general stress response. The findings also have practical implications for the analysis of stress response dynamics using lacZ reporter gene fusions, a frequently used strategy for the σ(B) response. link: http://identifiers.org/pubmed/22511268

Parameters:

NameDescription
kbd = 5.8E-9; kbx = 8.4E-5; kbs = 100.0Reaction: IPTG => sigb; x, IPTG, sigb, x, Rate Law: IPTG*kbs-sigb*(kbd+kbx*x)
kzs = 1.7E-6; kzd = 0.052Reaction: sigb => lacz; lacz, sigb, Rate Law: (-kzd*lacz)+kzs*sigb
kxd = 1.2E-13; kxs = 2.0E-6Reaction: sigb => x; sigb, x, Rate Law: kxs*sigb-kxd*x

States:

NameDescription
sigb[IPR006288]
lacz[galactosidase activity]
IPTG[isopropyl beta-D-thiogalactopyranoside]
x[inhibitor]

Liebal2012 - B.subtilis transcription inhibition model: BIOMD0000000461v0.0.1

Liebal2012 - B.subtilis transcription inhibition modelAn important transcription factor of B.subsilis is sigma B . Lieb…

Details

In Bacillus subtilis the σ(B) mediated general stress response provides protection against various environmental and energy related stress conditions. To better understand the general stress response, we need to explore the mechanism by which the components interact. Here, we performed experiments in B. subtilis wild type and mutant strains to test and validate a mathematical model of the dynamics of σ(B) activity. In the mutant strain BSA115, σ(B) transcription is inducible by the addition of IPTG and negative control of σ(B) activity by the anti-sigma factor RsbW is absent. In contrast to our expectations of a continuous β-galactosidase activity from a ctc::lacZ fusion, we observed a transient activity in the mutant. To explain this experimental finding, we constructed mathematical models reflecting different hypotheses regarding the regulation of σ(B) and β-galactosidase dynamics. Only the model assuming instability of either ctc::lacZ mRNA or β-galactosidase protein is able to reproduce the experiments in silico. Subsequent Northern blot experiments revealed stable high-level ctc::lacZ mRNA concentrations after the induction of the σ(B) response. Therefore, we conclude that protein instability following σ(B) activation is the most likely explanation for the experimental observations. Our results thus support the idea that B. subtilis increases the cytoplasmic proteolytic degradation to adapt the proteome in face of environmental challenges following activation of the general stress response. The findings also have practical implications for the analysis of stress response dynamics using lacZ reporter gene fusions, a frequently used strategy for the σ(B) response. link: http://identifiers.org/pubmed/22511268

Parameters:

NameDescription
kxd = 9.0; kxs = 0.76Reaction: sigb => x; x, sigb, Rate Law: (-kxd*x)+kxs*sigb/(1+x)
kbd = 0.044; kbs = 100.0Reaction: IPTG => sigb; IPTG, sigb, Rate Law: IPTG*kbs-kbd*sigb
kzs = 4.0E-4; kzd = 0.041Reaction: sigb => lacz; x, lacz, sigb, x, Rate Law: (-kzd*lacz)+kzs*sigb/(1+x)

States:

NameDescription
sigb[IPR006288]
lacz[Beta-galactosidase 12; beta-galactosidase activity]
IPTG[isopropyl beta-D-thiogalactopyranoside]
x[inhibitor]

Lim2014 - HTLV-I infection A dynamic struggle between viral persistence and host immunity: BIOMD0000000887v0.0.1

This is a four-dimensional, non-linear system of ordinary differential equations that describes the dynamic interactions…

Details

Human T-lymphotropic virus type I (HTLV-I) causes chronic infection for which there is no cure or neutralising vaccine. HTLV-I has been clinically linked to the development of adult T-cell leukaemia/lymphoma (ATL), an aggressive blood cancer, and HAM/TSP, a progressive neurological and inflammatory disease. Infected individuals typically mount a large, persistently activated CD8(+) cytotoxic T-lymphocyte (CTL) response against HTLV-I-infected cells, but ultimately fail to effectively eliminate the virus. Moreover, the identification of determinants to disease manifestation has thus far been elusive. A key issue in current HTLV-I research is to better understand the dynamic interaction between persistent infection by HTLV-I and virus-specific host immunity. Recent experimental hypotheses for the persistence of HTLV-I in vivo have led to the development of mathematical models illuminating the balance between proviral latency and activation in the target cell population. We investigate the role of a constantly changing anti-viral immune environment acting in response to the effects of infected T-cell activation and subsequent viral expression. The resulting model is a four-dimensional, non-linear system of ordinary differential equations that describes the dynamic interactions among viral expression, infected target cell activation, and the HTLV-I-specific CTL response. The global dynamics of the model is established through the construction of appropriate Lyapunov functions. Examining the particular roles of viral expression and host immunity during the chronic phase of HTLV-I infection offers important insights regarding the evolution of viral persistence and proposes a hypothesis for pathogenesis. link: http://identifiers.org/pubmed/24583256

Parameters:

NameDescription
tau = 0.003Reaction: u => y, Rate Law: compartment*tau*u
mu_3 = 0.03Reaction: y =>, Rate Law: compartment*mu_3*y
mu_1 = 0.012Reaction: x =>, Rate Law: compartment*mu_1*x
mu_4 = 0.03Reaction: z =>, Rate Law: compartment*mu_4*z
lambda = 10.0Reaction: => x, Rate Law: compartment*lambda
beta = 0.001Reaction: x => u; y, Rate Law: compartment*beta*x*y
r = 0.011Reaction: => u; y, Rate Law: compartment*r*y
gamma = 0.029Reaction: y => ; z, Rate Law: compartment*gamma*y*z
nu = 0.036Reaction: => z; y, Rate Law: compartment*nu*y
mu_2 = 0.03Reaction: u =>, Rate Law: compartment*mu_2*u

States:

NameDescription
x[CD4-positive helper T cell; infected cell]
u[CD4-positive helper T cell]
z[cytotoxic T cell]
y[CD4-positive helper T cell; infected cell]

Lindblad1996_ActionPotential_AtrialMyocyte: MODEL1006230015v0.0.1

This a model from the article: A model of the action potential and underlying membrane currents in a rabbit atrial cel…

Details

We have developed a mathematical model of the rabbit atrial myocyte and have used it in an examination of the ionic basis of the atrial action potential. Available biophysical data have been incorporated into the model to quantify the specific ultrastructural morphology, intracellular ion buffering, and time- and voltage-dependent currents and transport mechanisms of the rabbit atrial cell. When possible, mathematical expressions describing ionic currents identified in rabbit atrium are based on whole cell voltage-clamp data from enzymatically isolated rabbit atrial myocytes. This membrane model is coupled to equations describing Na+, K+, and Ca2+ homeostasis, including the uptake and release of Ca2+ by the sarcoplasmic reticulum and Ca2+ buffering. The resulting formulation can accurately simulate the whole cell voltage-clamp data on which it is based and provides fits to a family of rabbit atrial cell action potentials obtained at 35 degrees C over a range of stimulus rates (0.2-3.0 Hz). The model is utilized to provide a qualitative prediction of the intracellular Ca2+ concentration transient during the action potential and to illustrate the interactions between membrane currents that underlie repolarization in the rabbit atrial myocyte. link: http://identifiers.org/pubmed/8897964

Linke2017 - Synchronization of Cyclins' expression by the Fkh2 transcription factor in the budding yeast cell cycle: BIOMD0000000934v0.0.1

Modeling of the expression of cyclin B2 and B3 in the budding yeast cell cycle

Details

Precise timing of cell division is achieved by coupling waves of cyclin-dependent kinase (Cdk) activity with a transcriptional oscillator throughout cell cycle progression. Although details of transcription of cyclin genes are known, it is unclear which is the transcriptional cascade that modulates their expression in a timely fashion. Here, we demonstrate that a Clb/Cdk1-mediated regulation of the Fkh2 transcription factor synchronizes the temporal mitotic CLB expression in budding yeast. A simplified kinetic model of the cyclin/Cdk network predicts a linear cascade where a Clb/Cdk1-mediated regulation of an activator molecule drives CLB3 and CLB2 expression. Experimental validation highlights Fkh2 as modulator of CLB3 transcript levels, besides its role in regulating CLB2 expression. A Boolean model based on the minimal number of interactions needed to capture the information flow of the Clb/Cdk1 network supports the role of an activator molecule in the sequential activation, and oscillatory behavior, of mitotic Clb cyclins. This work illustrates how transcription and phosphorylation networks can be coupled by a Clb/Cdk1-mediated regulation that synchronizes them. link: http://identifiers.org/pubmed/28649434

Parameters:

NameDescription
k6 = 0.7Reaction: Cdk1_Clb5or6 => Clb5or6_degraded, Rate Law: cell*k6*Cdk1_Clb5or6
k2 = 5.0Reaction: Sic1 + Cdk1_Clb5or6 => Cdk1_Clb5or6_Sic1, Rate Law: cell*k2*Sic1*Cdk1_Clb5or6
k13 = 0.01Reaction: Cdk1_Clb1or2_Sic1 => Clb1or2_degraded, Rate Law: cell*k13*Cdk1_Clb1or2_Sic1
k4 = 0.01Reaction: Cdk1_Clb5or6_Sic1 => Clb5or6_degraded, Rate Law: cell*k4*Cdk1_Clb5or6_Sic1
k3 = 0.5Reaction: Cdk1_Clb5or6_Sic1 => Sic1 + Cdk1_Clb5or6, Rate Law: cell*k3*Cdk1_Clb5or6_Sic1
k11 = 5.0Reaction: Sic1 + Cdk1_Clb1or2 => Cdk1_Clb1or2_Sic1, Rate Law: cell*k11*Sic1*Cdk1_Clb1or2
k7 = 0.01; kA = 100.0Reaction: => Cdk1_Clb3or4; Cdk1_Clb5or6, Rate Law: cell*k7*(1+kA*Cdk1_Clb5or6)
kB = 1000.0; kC = 100.0; k9 = 0.001; kD = 100.0Reaction: => Cdk1_Clb1or2; Cdk1_Clb1or2, Cdk1_Clb3or4, Cdk1_Clb5or6, Rate Law: cell*k9*(1+kD*Cdk1_Clb1or2+kB*Cdk1_Clb3or4+kC*Cdk1_Clb5or6)
k10 = 0.7Reaction: Cdk1_Clb1or2 => Clb1or2_degraded, Rate Law: cell*k10*Cdk1_Clb1or2
k26 = 0.001Reaction: Sic1 =>, Rate Law: cell*k26*Sic1
k15 = 5.0Reaction: Sic1 + Cdk1_Clb3or4 => Cdk1_Clb3or4_Sic1, Rate Law: cell*k15*Sic1*Cdk1_Clb3or4
k5 = 0.05Reaction: Cdk1_Clb5or6_Sic1 => Cdk1_Clb5or6 + Sic1_degraded_re5; Cdk1_Clb5or6, Cdk1_Clb3or4, Cdk1_Clb1or2, Rate Law: cell*k5*Cdk1_Clb5or6_Sic1*(1+Cdk1_Clb5or6+Cdk1_Clb3or4+Cdk1_Clb1or2)
k18 = 0.05Reaction: Cdk1_Clb3or4_Sic1 => Sic1_degraded_re18; Cdk1_Clb5or6, Cdk1_Clb3or4, Cdk1_Clb1or2, Rate Law: cell*k18*Cdk1_Clb3or4_Sic1*(1+Cdk1_Clb5or6+Cdk1_Clb3or4+Cdk1_Clb1or2)
k8 = 0.7Reaction: Cdk1_Clb3or4 => Clb3or4_degraded, Rate Law: cell*k8*Cdk1_Clb3or4
k12 = 0.5Reaction: Cdk1_Clb1or2_Sic1 => Sic1 + Cdk1_Clb1or2, Rate Law: cell*k12*Cdk1_Clb1or2_Sic1
k16 = 0.5Reaction: Cdk1_Clb3or4_Sic1 => Sic1 + Cdk1_Clb3or4, Rate Law: cell*k16*Cdk1_Clb3or4_Sic1
k17 = 0.01Reaction: Cdk1_Clb3or4_Sic1 => Clb3or4_degraded, Rate Law: cell*k17*Cdk1_Clb3or4_Sic1
k1 = 0.1Reaction: => Cdk1_Clb5or6, Rate Law: cell*k1
k14 = 0.05Reaction: Cdk1_Clb1or2_Sic1 => Sic1_degraded_re14; Cdk1_Clb5or6, Cdk1_Clb3or4, Cdk1_Clb1or2, Rate Law: cell*k14*Cdk1_Clb1or2_Sic1*(1+Cdk1_Clb5or6+Cdk1_Clb3or4+Cdk1_Clb1or2)

States:

NameDescription
Sic1 degraded re5[Protein SIC1; Removed]
Clb1or2 degraded[G2/mitotic-specific cyclin-2; Removed]
Clb12 total[G2/mitotic-specific cyclin-2]
Clb34 total[G2/mitotic-specific cyclin-3]
Cdk1 Clb1or2[G2/mitotic-specific cyclin-2; Cyclin-dependent kinase 1; protein-containing complex]
Clb5or6 degraded[S-phase entry cyclin-5; Removed]
Sic1[Protein SIC1]
Clb56 total[S-phase entry cyclin-5]
Sic1 degraded re18[Protein SIC1; Removed]
Cdk1 Clb3or4[G2/mitotic-specific cyclin-3; Cyclin-dependent kinase 1; protein-containing complex]
Cdk1 Clb3or4 Sic1[G2/mitotic-specific cyclin-3; Cyclin-dependent kinase 1; Protein SIC1; protein-containing complex]
Cdk1 Clb5or6 Sic1[Protein SIC1; Cyclin-dependent kinase 1; S-phase entry cyclin-5; protein-containing complex]
Sic1 degraded re14[Protein SIC1; Removed]
Clb3or4 degraded[G2/mitotic-specific cyclin-3; Removed]
Cdk1 Clb1or2 Sic1[Cyclin-dependent kinase 1; Protein SIC1; G2/mitotic-specific cyclin-2; protein-containing complex]
Cdk1 Clb5or6[Cyclin-dependent kinase 1; S-phase entry cyclin-5; protein-containing complex]

Lipniacki2004 - Mathematical model of NFKB regulatory module: BIOMD0000000786v0.0.1

its a mathematical model studying impact of TNF on NFKB nuclear dynamics. This model is derived from Hoffmann2002 (PMID:…

Details

The two-feedback-loop regulatory module of nuclear factor kappaB (NF-kappaB) signaling pathway is modeled by means of ordinary differential equations. The constructed model involves two-compartment kinetics of the activators IkappaB (IKK) and NF-kappaB, the inhibitors A20 and IkappaBalpha, and their complexes. In resting cells, the unphosphorylated IkappaBalpha binds to NF-kappaB and sequesters it in an inactive form in the cytoplasm. In response to extracellular signals such as tumor necrosis factor or interleukin-1, IKK is transformed from its neutral form (IKKn) into its active form (IKKa), a form capable of phosphorylating IkappaBalpha, leading to IkappaBalpha degradation. Degradation of IkappaBalpha releases the main activator NF-kappaB, which then enters the nucleus and triggers transcription of the inhibitors and numerous other genes. The newly synthesized IkappaBalpha leads NF-kappaB out of the nucleus and sequesters it in the cytoplasm, while A20 inhibits IKK converting IKKa into the inactive form (IKKi), a form different from IKKn, no longer capable of phosphorylating IkappaBalpha. After parameter fitting, the proposed model is able to properly reproduce time behavior of all variables for which the data are available: NF-kappaB, cytoplasmic IkappaBalpha, A20 and IkappaBalpha mRNA transcripts, IKK and IKK catalytic activity in both wild-type and A20-deficient cells. The model allows detailed analysis of kinetics of the involved proteins and their complexes and gives the predictions of the possible responses of whole kinetics to the change in the level of a given activator or inhibitor. link: http://identifiers.org/pubmed/15094015

Parameters:

NameDescription
Kv = 5.0; i1 = 0.0025Reaction: => NFKB_nuc; NFKB, Rate Law: function_for_indirect_transport(i1, Kv, NFKB)
a1 = 0.5Reaction: NFKB_nuc + IkB_nuc => IkB_NFKB_nuc, Rate Law: Nucleus*a1*NFKB_nuc*IkB_nuc
a2 = 0.2Reaction: IKK_active + IkB => IKKactive_IkB, Rate Law: Cytosol*a2*IKK_active*IkB
Kv = 5.0; e2a = 0.01Reaction: IkB_NFKB_nuc =>, Rate Law: Nucleus*function_for_transport(e2a, Kv, IkB_NFKB_nuc)
t2 = 0.1Reaction: IKKactive_IkB_NFKB => IKK_active + NFKB, Rate Law: Cytosol*t2*IKKactive_IkB_NFKB
c5 = 3.0E-4Reaction: A20 =>, Rate Law: Cytosol*c5*A20
i1 = 0.0025Reaction: NFKB =>, Rate Law: Cytosol*i1*NFKB
a3 = 1.0Reaction: IKK_active + IkB_NFKB => IKKactive_IkB_NFKB, Rate Law: Cytosol*a3*IKK_active*IkB_NFKB
i1a = 0.001Reaction: IkB =>, Rate Law: Cytosol*i1a*IkB
c3a = 4.0E-4Reaction: IkB_mRNA =>, Rate Law: Nucleus*c3a*IkB_mRNA
Kprod = 2.5E-5Reaction: => IKK_neutral, Rate Law: Cytosol*Constant_flux__irreversible(Kprod)
Kdeg = 1.25E-4Reaction: IKK_inact =>, Rate Law: Cytosol*Kdeg*IKK_inact
c4 = 0.5Reaction: => A20; A20_mRNA, Rate Law: function_for_substrateless_production(c4, A20_mRNA)
c1c = 5.0E-7Reaction: => cgen_mRNA; NFKB_nuc, Rate Law: Nucleus*function_for_substrateless_production(c1c, NFKB_nuc)
c5a = 1.0E-4Reaction: IkB =>, Rate Law: Cytosol*c5a*IkB
Kv = 5.0; e1a = 5.0E-4Reaction: IkB_nuc =>, Rate Law: Nucleus*function_for_transport(e1a, Kv, IkB_nuc)
c1a = 5.0E-7Reaction: => IkB_mRNA; NFKB_nuc, Rate Law: Nucleus*function_for_substrateless_production(c1a, NFKB_nuc)
e1a = 5.0E-4Reaction: => IkB; IkB_nuc, Rate Law: function_for_indirect_production(e1a, IkB_nuc)
c3c = 4.0E-4Reaction: cgen_mRNA =>, Rate Law: Nucleus*c3c*cgen_mRNA
TNF_R = 0.0Reaction: TNF = TNF_R, Rate Law: missing
c6a = 2.0E-5Reaction: IkB_NFKB => NFKB, Rate Law: Cytosol*c6a*IkB_NFKB
k2 = 0.1Reaction: IKK_active => IKK_inact; TNF, A20, Rate Law: Cytosol*function_for_R26(k2, TNF, A20, IKK_active)
t1 = 0.1Reaction: IKKactive_IkB => IKK_active, Rate Law: Cytosol*t1*IKKactive_IkB
c1 = 5.0E-7Reaction: => A20_mRNA; NFKB_nuc, Rate Law: Nucleus*function_for_substrateless_production(c1, NFKB_nuc)
Kv = 5.0; i1a = 0.001Reaction: => IkB_nuc; IkB, Rate Law: function_for_indirect_transport(i1a, Kv, IkB)
c4a = 0.5Reaction: => IkB; IkB_mRNA, Rate Law: function_for_substrateless_production(c4a, IkB_mRNA)
k1 = 0.0025Reaction: IKK_neutral => IKK_active; TNF, Rate Law: Cytosol*function_for_R3(k1, TNF, IKK_neutral)
k3 = 0.0015Reaction: IKK_active => IKK_inact, Rate Law: Cytosol*k3*IKK_active
e2a = 0.01Reaction: => IkB_NFKB; IkB_NFKB_nuc, Rate Law: function_for_indirect_production(e2a, IkB_NFKB_nuc)
c3 = 4.0E-4Reaction: A20_mRNA =>, Rate Law: Nucleus*c3*A20_mRNA

States:

NameDescription
IKKactive IkB NFKB[Nuclear factor NF-kappa-B p105 subunit; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor alpha]
IKK inact[Inhibitor of nuclear factor kappa-B kinase subunit alpha]
TNF[Tumor necrosis factor]
A20[Tumor necrosis factor alpha-induced protein 3]
IkB mRNA[NF-kappa-B inhibitor alpha]
IkB NFKB nuc[NF-kappa-B inhibitor alpha; Nuclear factor NF-kappa-B p105 subunit]
IKK active[Inhibitor of nuclear factor kappa-B kinase subunit alpha]
IkB[NF-kappa-B inhibitor alpha]
IKKactive IkB[NF-kappa-B inhibitor alpha; Inhibitor of nuclear factor kappa-B kinase subunit alpha]
NFKB nuc[Nuclear factor NF-kappa-B p105 subunit]
IkB NFKB[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha]
cgen mRNAcgen_mRNA
IKK neutral[Inhibitor of nuclear factor kappa-B kinase subunit alpha]
IkB nuc[NF-kappa-B inhibitor alpha]
NFKB[Nuclear factor NF-kappa-B p105 subunit]
A20 mRNA[Tumor necrosis factor alpha-induced protein 3]

Liu1999_PulsatileSecretion: MODEL1006230060v0.0.1

This a model from the article: A dynamical model for the pubsatile secretion of the hypothalamo-pituitary-adrenal axis…

Details

We propose a dynamical model for the hypothalamo-pituitary-thyroid axis. This model takes into account both the interactions of hormones in this axis and the binding of thyroid hormones with proteins in plasma and tissues. It can account for the pulsatile character of hormone secretion in this axis as well as many experimental results. link: http://identifiers.org/pubmed/7772725

Liu2009_GlucoseMobilization_UptakeModel: MODEL1112050001v0.0.1

This a model from the article: A molecular mathematical model of glucose mobilization and uptake. Liu W, Hsin C, Tan…

Details

A new molecular mathematical model is developed by considering the kinetics of GLUT2, GLUT3, and GLUT4, the process of glucose mobilization by glycogen phosphorylase and glycogen synthase in liver, and the dynamics of the insulin signaling pathway. The new model can qualitatively reproduce the experimental glucose and insulin data. It also enables us to use the Bendixson criterion about the existence of periodic orbits of a two-dimensional dynamical system to mathematically predict that the oscillations of glucose and insulin are not caused by liver, instead they would be caused by the mechanism of insulin secretion from pancreatic beta cells. Furthermore it enables us to conduct a parametric sensitivity analysis. The analysis shows that both glucose and insulin are most sensitive to the rate constant for conversion of PI(3,4,5)P(3) to PI(4,5)P(2), the multiplicative factor modulating the rate constant for conversion of PI(3,4,5)P(3) to PI(4,5)P(2), the multiplicative factor that modulates insulin receptor dephosphorylation rate, and the maximum velocity of GLUT4. Moreover, the sensitivity analysis predicts that an increase of the apparent velocity of GLUT4, a combination of elevated mobilization rate of GLUT4 to the plasma membrane and an extended duration of GLUT4 on the plasma membrane, will result in a decrease in the needs of plasma insulin. On the other hand, an increase of the GLUT4 internalization rate results in an elevated demand of insulin to stimulate the mobilization of GLUT4 from the intracellular store to the plasma membrane. link: http://identifiers.org/pubmed/19651146

Liu2010_Hormonal_Crosstalk_Arabidopsis: BIOMD0000000269v0.0.1

This is the single cell model for analysis of hormonal crosstalk in Arabidopsis described in the article: Modelling an…

Details

An important question in plant biology is how genes influence the crosstalk between hormones to regulate growth. In this study, we model POLARIS (PLS) gene function and crosstalk between auxin, ethylene and cytokinin in Arabidopsis. Experimental evidence suggests that PLS acts on or close to the ethylene receptor ETR1, and a mathematical model describing possible PLS-ethylene pathway interactions is developed, and used to make quantitative predictions about PLS-hormone interactions. Modelling correctly predicts experimental results for the effect of the pls gene mutation on endogenous cytokinin concentration. Modelling also reveals a role for PLS in auxin biosynthesis in addition to a role in auxin transport. The model reproduces available mutants, and with new experimental data provides new insights into how PLS regulates auxin concentration, by controlling the relative contribution of auxin transport and biosynthesis and by integrating auxin, ethylene and cytokinin signalling. Modelling further reveals that a bell-shaped dose-response relationship between endogenous auxin and root length is established via PLS. This combined modelling and experimental analysis provides new insights into the integration of hormonal signals in plants. link: http://identifiers.org/pubmed/20531403

Parameters:

NameDescription
k_cytokinin = 10.0 per_secReaction: CK_ex => CK, Rate Law: compartment_1*k_cytokinin*CK_ex
k4 = 1.0 per_uM_per_secReaction: Ra => Ra_star; Auxin, Rate Law: compartment_1*k4*Auxin*Ra
k12 = 0.1 per_uM_per_sec; k12a = 0.1 per_uM_per_secReaction: => ET; Auxin, CK, Rate Law: compartment_1*(k12+k12a*Auxin*CK)
k11 = 5.0 per_uM_per_secReaction: Re_star => Re; ET, Rate Law: compartment_1*k11*Re_star*ET
k2a = 2.8 per_sec; k2b = 1.0 uM; k2c = 0.01 uM; k2 = 0.2 per_uM_per_secReaction: => Auxin; ET, CK, PLSp, Rate Law: compartment_1*(k2+k2a*ET/(1+CK/k2b)*PLSp/(k2c+PLSp))
k1a = 1.0 per_uM_per_sec; k1 = 1.0 uMReaction: => Auxin; X, Rate Law: compartment_1*k1a/(1+X/k1)
k8 = 1.0 per_secReaction: => PLSp; PLSm, Rate Law: compartment_1*k8*PLSm
k9 = 1.0 per_secReaction: PLSp =>, Rate Law: compartment_1*k9*PLSp
k14 = 3.0 per_uM_per_secReaction: CTR1 => CTR1_star; Re_star, Rate Law: compartment_1*k14*Re_star*CTR1
k10 = 3.0E-4 per_sec; k10a = 0.5 per_uM_per_secReaction: Re => Re_star; PLSp, Rate Law: compartment_1*(k10+k10a*PLSp)*Re
k_ethylene = 0.5 per_secReaction: ACC => ET, Rate Law: compartment_1*k_ethylene*ACC
k16a = 1.0 per_sec; k16 = 0.3 per_uM_per_secReaction: => X; CTR1_star, Rate Law: compartment_1*(k16-k16a*CTR1_star)
k17 = 0.1 per_secReaction: X =>, Rate Law: compartment_1*k17*X
k13 = 1.0 per_secReaction: ET =>, Rate Law: compartment_1*k13*ET
k6 = 0.3 per_sec; k6a = 0.2 uMReaction: => PLSm; Ra_star, ET, Rate Law: compartment_1*k6*Ra_star/(1+ET/k6a)
k18a = 1.0 per_uM_per_sec; k18 = 0.1 uMReaction: => CK; Auxin, Rate Law: compartment_1*k18a/(1+Auxin/k18)
k5 = 1.0 per_secReaction: Ra_star => Ra, Rate Law: compartment_1*k5*Ra_star
k7 = 1.0 per_secReaction: PLSm =>, Rate Law: compartment_1*k7*PLSm
k19 = 1.0 per_secReaction: CK =>, Rate Law: compartment_1*k19*CK
k_auxin = 70.0 per_secReaction: IAA => Auxin, Rate Law: compartment_1*k_auxin*IAA
k3a = 0.45 per_uM_per_sec; k3 = 2.0 per_secReaction: Auxin => ; X, Rate Law: compartment_1*(k3+k3a*X)*Auxin
k15 = 0.085 per_secReaction: CTR1_star => CTR1, Rate Law: compartment_1*k15*CTR1_star

States:

NameDescription
Ra[auxin binding]
Ra star[auxin binding]
XX
PLSp[3770598; Peptide POLARIS]
Auxin[auxin]
IAA[indole-3-acetic acid; Indole-3-acetate]
ACC[1-aminocyclopropanecarboxylic acid]
PLSm[3770598]
Re star[Ethylene receptor 1]
Re[Ethylene receptor 1; Ethylene receptor 2]
CTR1[Serine/threonine-protein kinase CTR1]
CK ex[cytokinin]
ET[ethene; Ethylene]
CK[cytokinin]
CTR1 star[Serine/threonine-protein kinase CTR1]

Liu2011_Complement_System: BIOMD0000000303v0.0.1

Model of the Complement SystemThis is the continuous deterministic (ODE) model of the complement system described in the…

Details

The complement system is key to innate immunity and its activation is necessary for the clearance of bacteria and apoptotic cells. However, insufficient or excessive complement activation will lead to immune-related diseases. It is so far unknown how the complement activity is up- or down- regulated and what the associated pathophysiological mechanisms are. To quantitatively understand the modulatory mechanisms of the complement system, we built a computational model involving the enhancement and suppression mechanisms that regulate complement activity. Our model consists of a large system of Ordinary Differential Equations (ODEs) accompanied by a dynamic Bayesian network as a probabilistic approximation of the ODE dynamics. Applying Bayesian inference techniques, this approximation was used to perform parameter estimation and sensitivity analysis. Our combined computational and experimental study showed that the antimicrobial response is sensitive to changes in pH and calcium levels, which determines the strength of the crosstalk between CRP and L-ficolin. Our study also revealed differential regulatory effects of C4BP. While C4BP delays but does not decrease the classical complement activation, it attenuates but does not significantly delay the lectin pathway activation. We also found that the major inhibitory role of C4BP is to facilitate the decay of C3 convertase. In summary, the present work elucidates the regulatory mechanisms of the complement system and demonstrates how the bio-pathway machinery maintains the balance between activation and inhibition. The insights we have gained could contribute to the development of therapies targeting the complement system. link: http://identifiers.org/pubmed/21283780

Parameters:

NameDescription
kg04_1 = 1.1; kg04_2 = 2000.0Reaction: C2 => C2a + C2b; GlcNac_HF_MASP, Rate Law: compartment*kg04_1*GlcNac_HF_MASP*C2/(kg04_2+C2)
kg03_2 = 829.115813389061; kg03_1 = 66.3776807395383Reaction: C4 => C4a + C4b; GlcNac_HF_MASP, Rate Law: compartment*kg03_1*GlcNac_HF_MASP*C4/(kg03_2+C4)
kd08_2 = 0.1; kd08_1 = 0.0368010796682635Reaction: PC_CRP_LF_C1 + MASP => PC_CRP_LF_C1_MASP, Rate Law: compartment*(kd08_1*PC_CRP_LF_C1*MASP-kd08_2*PC_CRP_LF_C1_MASP)
kf01_2 = 0.069020578737621; kf01_1 = 0.969998277173144Reaction: C4BP + PC_CRP => C4BP_PC_CRP, Rate Law: compartment*(kf01_1*C4BP*PC_CRP-kf01_2*C4BP_PC_CRP)
kg01_1 = 0.091011109910329; kg01_2 = 0.0508205528375529Reaction: X + HF => GlcNac_HF, Rate Law: compartment*(kg01_1*X*HF-kg01_2*GlcNac_HF)
ke01_1 = 7.07E-5; ke01_2 = 1.0E-4Reaction: GlcNac_LF + CRP => GlcNac_LF_CRP, Rate Law: compartment*(ke01_1*GlcNac_LF*CRP-ke01_2*GlcNac_LF_CRP)
kf04_1 = 0.613416050428938; kf04_2 = 0.983691204042155Reaction: C4BP + C4b => C4BP_C4b, Rate Law: compartment*(kf04_1*C4BP*C4b-kf04_2*C4BP_C4b)
k1_4=0.0470911Reaction: C4b_C2a =>, Rate Law: compartment*k1_4*C4b_C2a
kd06_1 = 2.0; kd06_2 = 500.0Reaction: C4 => C4a + C4b; PC_CRP_LF_C1, Rate Law: compartment*kd06_1*PC_CRP_LF_C1*C4/(kd06_2+C4)
ke07_2 = 2000.0; ke07_1 = 1.1Reaction: C2 => C2a + C2b; GlcNac_LF_CRP_MASP, Rate Law: compartment*ke07_1*GlcNac_LF_CRP_MASP*C2/(ke07_2+C2)
kc03_1 = 0.414004459949002; kc03_2 = 0.99647572245388Reaction: C3b => dC3b, Rate Law: compartment*(kc03_1*C3b-kc03_2*dC3b)
kf02_2 = 0.069020578737621; kf02_1 = 0.969998277173144Reaction: C4BP + GlcNac_LF_CRP => C4BP_GlcNac_LF_CRP, Rate Law: compartment*(kf02_1*C4BP*GlcNac_LF_CRP-kf02_2*C4BP_GlcNac_LF_CRP)
kd01_2 = 7.23E-5; kd01_1 = 7.07E-5Reaction: PC_CRP + LF => PC_CRP_LF, Rate Law: compartment*(kd01_1*PC_CRP*LF-kd01_2*PC_CRP_LF)
k1_4=3.42266E-4Reaction: C4BP =>, Rate Law: compartment*k1_4*C4BP
k1_4=0.492901Reaction: C3b =>, Rate Law: compartment*k1_4*C3b
ke04_2 = 2500.0; ke04_1 = 10.5Reaction: C2 => C2a + C2b; GlcNac_LF_CRP_C1, Rate Law: compartment*ke04_1*GlcNac_LF_CRP_C1*C2/(ke04_2+C2)
kd05_1 = 7.4E-4; kd05_2 = 0.0011Reaction: PC_CRP_LF + C1 => PC_CRP_LF_C1, Rate Law: compartment*(kd05_1*PC_CRP_LF*C1-kd05_2*PC_CRP_LF_C1)
kd02_1 = 0.0368010796682635; kd02_2 = 0.1Reaction: PC_CRP_LF + MASP => PC_CRP_LF_MASP, Rate Law: compartment*(kd02_1*PC_CRP_LF*MASP-kd02_2*PC_CRP_LF_MASP)
ke02_2 = 0.0011; ke02_1 = 7.4E-4Reaction: GlcNac_LF_CRP + C1 => GlcNac_LF_CRP_C1, Rate Law: compartment*(ke02_1*GlcNac_LF_CRP*C1-ke02_2*GlcNac_LF_CRP_C1)
ke03_2 = 500.0; ke03_1 = 2.0Reaction: C4 => C4a + C4b; GlcNac_LF_CRP_C1, Rate Law: compartment*ke03_1*GlcNac_LF_CRP_C1*C4/(ke03_2+C4)
kd10_1 = 71.1705760475931; kd10_2 = 3796.22684377655Reaction: C4 => C4a + C4b; PC_CRP_LF_C1_MASP, Rate Law: compartment*kd10_1*PC_CRP_LF_C1_MASP*C4/(kd10_2+C4)
ka03_2 = 500.0; ka03_1 = 2.0Reaction: C4 => C4a + C4b; PC_CRP_C1, Rate Law: compartment*ka03_1*PC_CRP_C1*C4/(ka03_2+C4)
kf06_1 = 0.613416050428938; kf06_2 = 0.983691204042155Reaction: C4b_C2a + C4BP => C4b_C2a_C4BP, Rate Law: compartment*(kf06_1*C4b_C2a*C4BP-kf06_2*C4b_C2a_C4BP)
kd03_2 = 829.115813389061; kd03_1 = 66.3776807395383Reaction: C4 => C4a + C4b; PC_CRP_LF_MASP, Rate Law: compartment*kd03_1*PC_CRP_LF_MASP*C4/(kd03_2+C4)
kc04_1 = 0.977836567576895; kc04_2 = 0.199162432258527Reaction: C4b_C2a => dC4b_C2a, Rate Law: compartment*(kc04_1*C4b_C2a-kc04_2*dC4b_C2a)
kc01_1 = 0.64564661669102; kc01_2 = 0.194551104058408Reaction: C4b + C2a => C4b_C2a, Rate Law: compartment*(kc01_1*C4b*C2a-kc01_2*C4b_C2a)
kd04_1 = 1.1; kd04_2 = 2000.0Reaction: C2 => C2a + C2b; PC_CRP_LF_MASP, Rate Law: compartment*kd04_1*PC_CRP_LF_MASP*C2/(kd04_2+C2)
ke05_1 = 0.0368010796682635; ke05_2 = 0.1Reaction: GlcNac_LF_CRP + MASP => GlcNac_LF_CRP_MASP, Rate Law: compartment*(ke05_1*GlcNac_LF_CRP*MASP-ke05_2*GlcNac_LF_CRP_MASP)
kb02_2 = 0.1; kb02_1 = 0.0368010796682635Reaction: GlcNac_LF + MASP => GlcNac_LF_MASP, Rate Law: compartment*(kb02_1*GlcNac_LF*MASP-kb02_2*GlcNac_LF_MASP)
kf07_1 = 0.613416050428938; kf07_2 = 0.983691204042155Reaction: dC4b_C2a + C4BP => dC4b_C2a_C4BP, Rate Law: compartment*(kf07_1*dC4b_C2a*C4BP-kf07_2*dC4b_C2a_C4BP)
kd07_2 = 2500.0; kd07_1 = 10.5Reaction: C2 => C2a + C2b; PC_CRP_LF_C1, Rate Law: compartment*kd07_1*PC_CRP_LF_C1*C2/(kd07_2+C2)
kb01_2 = 0.0508205528375529; kb01_1 = 0.091011109910329Reaction: GlcNac + LF => GlcNac_LF, Rate Law: compartment*(kb01_1*GlcNac*LF-kb01_2*GlcNac_LF)
ke06_1 = 66.3776807395383; ke06_2 = 829.115813389061Reaction: C4 => C4a + C4b; GlcNac_LF_CRP_MASP, Rate Law: compartment*ke06_1*GlcNac_LF_CRP_MASP*C4/(ke06_2+C4)
ka02_2 = 0.0011; ka02_1 = 7.4E-4Reaction: PC_CRP + C1 => PC_CRP_C1, Rate Law: compartment*(ka02_1*PC_CRP*C1-ka02_2*PC_CRP_C1)
kf05 = 0.980777558937884Reaction: C4b_C2a + C4BP => C4b + C2a + C4BP, Rate Law: compartment*kf05*C4b_C2a*C4BP
kb04_1 = 1.1; kb04_2 = 2000.0Reaction: C2 => C2a + C2b; GlcNac_LF_MASP, Rate Law: compartment*kb04_1*GlcNac_LF_MASP*C2/(kb04_2+C2)
kb03_2 = 829.115813389061; kb03_1 = 66.3776807395383Reaction: C4 => C4a + C4b; GlcNac_LF_MASP, Rate Law: compartment*kb03_1*GlcNac_LF_MASP*C4/(kb03_2+C4)
kf03 = 0.0613537204215846Reaction: C4b_C2a + C4BP => iC4b_C2a + C4BP, Rate Law: compartment*kf03*C4b_C2a*C4BP
ka04_2 = 2500.0; ka04_1 = 10.5Reaction: C2 => C2a + C2b; PC_CRP_C1, Rate Law: compartment*ka04_1*PC_CRP_C1*C2/(ka04_2+C2)
ka01_1 = 0.0275999; ka01_2 = 0.0109Reaction: PC + CRP => PC_CRP, Rate Law: compartment*(ka01_1*PC*CRP-ka01_2*PC_CRP)
kg02_1 = 0.0368010796682635; kg02_2 = 0.1Reaction: GlcNac_HF + MASP => GlcNac_HF_MASP, Rate Law: compartment*(kg02_1*GlcNac_HF*MASP-kg02_2*GlcNac_HF_MASP)
kc02 = 5.91152775857994E-4Reaction: C4b_C2a + C3 => C4b_C2a + C3a + C3b, Rate Law: compartment*kc02*C4b_C2a*C3
kd11_2 = 5972.30640657865; kd11_1 = 38.9625903487667Reaction: C2 => C2a + C2b; PC_CRP_LF_C1_MASP, Rate Law: compartment*kd11_1*PC_CRP_LF_C1_MASP*C2/(kd11_2+C2)
k1_4=0.0; k2=0.0Reaction: C4BP + PC_CRP_LF => C4BP_PC_CRP_LF, Rate Law: compartment*(k1_4*C4BP*PC_CRP_LF-k2*C4BP_PC_CRP_LF)
kd09_1 = 7.4E-4; kd09_2 = 0.0011Reaction: PC_CRP_LF_MASP + C1 => PC_CRP_LF_C1_MASP, Rate Law: compartment*(kd09_1*PC_CRP_LF_MASP*C1-kd09_2*PC_CRP_LF_C1_MASP)

States:

NameDescription
dC4b C2a C4BP[C4b-binding protein alpha chain; cell surface; C2a [extracellular region]; thioester-C1010-Q1013-C4b [extracellular region]]
C3[Complement factor 3 [extracellular region]; Complement C3]
dC3b[Cell surface:C3b [plasma membrane]; cell surface]
GlcNac LF CRP C1[N-acetyl-D-glucosamine; C-reactive protein; Ficolin-2; C1Q:2xC1R:2xC1S [extracellular region]]
PC CRP[phosphocholine; C-reactive protein]
PC CRP LF[phosphocholine; C-reactive protein; Ficolin-2]
GlcNac LF CRP MASP[N-acetyl-D-glucosamine; Mannan-binding lectin serine protease 2; Ficolin-2; C-reactive protein]
C4b[Complement C4-B; Complement C4-A; thioester-C1010-Q1013-C4b [extracellular region]]
PC CRP LF MASP[phosphocholine; Ficolin-2; C-reactive protein; Mannan-binding lectin serine protease 2]
C4a[Complement C4-A; Complement C4-B; C4a [extracellular region]]
LF[Ficolin-2]
C2b[C2b [extracellular region]]
C4BP C4b[C4b-binding protein alpha chain; thioester-C1010-Q1013-C4b [extracellular region]]
C2[Complement C2; C2 [extracellular region]]
C1[C1Q:2xC1R:2xC1S [extracellular region]]
PC[phosphocholine; Choline phosphate]
C4b C2a[C2a [extracellular region]; thioester-C1010-Q1013-C4b [extracellular region]]
iC4b C2a[thioester-C1010-Q1013-C4b [extracellular region]; C2a [extracellular region]]
GlcNac HF[N-acetyl-D-glucosamine]
GlcNac LF MASP[N-acetyl-D-glucosamine; Ficolin-2; Mannan-binding lectin serine protease 2]
C3b[C3b [extracellular region]]
GlcNac[N-acetyl-D-glucosamine; N-Acetyl-D-glucosamine]
GlcNac LF[N-acetyl-D-glucosamine; Ficolin-2]
C4BP[C4b-binding protein alpha chain]
C3a[C3a [extracellular region]; Complement C3]
PC CRP LF C1[phosphocholine; C-reactive protein; Ficolin-2; C1Q:2xC1R:2xC1S [extracellular region]]
XX
PC CRP C1[phosphocholine; C-reactive protein; C1Q:2xC1R:2xC1S [extracellular region]]
C4BP PC CRP[phosphocholine; C4b-binding protein alpha chain; C-reactive protein]
PC CRP LF C1 MASP[phosphocholine; Mannan-binding lectin serine protease 2; Ficolin-2; C1Q:2xC1R:2xC1S [extracellular region]]
GlcNac LF CRP[N-acetyl-D-glucosamine; Ficolin-2; C-reactive protein]
HFHF
C2a[C2a [extracellular region]]
CRP[C-reactive protein; IPR001759; CRP(19-224) [extracellular region]]
dC4b C2a[cell surface]
C4BP GlcNac LF CRP[N-acetyl-D-glucosamine; C4b-binding protein alpha chain; Ficolin-2; C-reactive protein]
MASP[Mannan-binding lectin serine protease 2]
C4BP PC CRP LF[phosphocholine; C4b-binding protein alpha chain; Ficolin-2; C-reactive protein]
GlcNac HF MASP[N-acetyl-D-glucosamine; Mannan-binding lectin serine protease 2]
C4b C2a C4BP[thioester-C1010-Q1013-C4b [extracellular region]; C4b-binding protein alpha chain; C2a [extracellular region]]

Liu2012 - Genome-scale metabolic network of Scheffersomyces stipitis (iTL885): MODEL1507180026v0.0.1

Liu2012 - Genome-scale metabolic network of Scheffersomyces stipitis (iTL885)This model is described in the article: [A…

Details

As one of the best xylose utilization microorganisms, Scheffersomyces stipitis exhibits great potential for the efficient lignocellulosic biomass fermentation. Therefore, a comprehensive understanding of its unique physiological and metabolic characteristics is required to further improve its performance on cellulosic ethanol production.A constraint-based genome-scale metabolic model for S. stipitis CBS 6054 was developed on the basis of its genomic, transcriptomic and literature information. The model iTL885 consists of 885 genes, 870 metabolites, and 1240 reactions. During the reconstruction process, 36 putative sugar transporters were reannotated and the metabolisms of 7 sugars were illuminated. Essentiality study was conducted to predict essential genes on different growth media. Key factors affecting cell growth and ethanol formation were investigated by the use of constraint-based analysis. Furthermore, the uptake systems and metabolic routes of xylose were elucidated, and the optimization strategies for the overproduction of ethanol were proposed from both genetic and environmental perspectives.Systems biology modelling has proven to be a powerful tool for targeting metabolic changes. Thus, this systematic investigation of the metabolism of S. stipitis could be used as a starting point for future experiment designs aimed at identifying the metabolic bottlenecks of this important yeast. link: http://identifiers.org/pubmed/22998943

Liu2012-Hybrid modeling and simulation of stochastic effects on progression through the eukaryotic cell cycle.: MODEL2004140002v0.0.1

The eukaryotic cell cycle is regulated by a complicated chemical reaction network. Although many deterministic models ha…

Details

The eukaryotic cell cycle is regulated by a complicated chemical reaction network. Although many deterministic models have been proposed, stochastic models are desired to capture noise in the cell resulting from low numbers of critical species. However, converting a deterministic model into one that accurately captures stochastic effects can result in a complex model that is hard to build and expensive to simulate. In this paper, we first apply a hybrid (mixed deterministic and stochastic) simulation method to such a stochastic model. With proper partitioning of reactions between deterministic and stochastic simulation methods, the hybrid method generates the same primary characteristics and the same level of noise as Gillespie's stochastic simulation algorithm, but with better efficiency. By studying the results generated by various partitionings of reactions, we developed a new strategy for hybrid stochastic modeling of the cell cycle. The new approach is not limited to using mass-action rate laws. Numerical experiments demonstrate that our approach is consistent with characteristics of noisy cell cycle progression, and yields cell cycle statistics in accord with experimental observations. link: http://identifiers.org/pubmed/22280742

Liu2017 - chemotherapy targeted model of tumor immune system: BIOMD0000000930v0.0.1

Its a mathematical model reflecting chemotherapy response in tumor immune interaction system. Model encoded by Sarubini…

Details

Considering the targeted chemotherapy, a mathematical model of tumor-immune system was constructed on the basis of de Pillis’s model. In this paper, we conducted qualitative analysis on the mathematical model, including the positivity and boundedness of solutions, local stability and global stability of equi- librium solutions. Some numerical simulations were given to illustrate the analytic results. Comparing the targeted chemotherapy model with regular chemotherapy model, we found that the targeted chemother- apy was benefit to kill tumor cells. link: http://identifiers.org/doi/10.1016/j.chaos.2017.03.002

Parameters:

NameDescription
gamma = 0.9Reaction: Chemotherapeutic_drug_concentration__M =>, Rate Law: Tumor_Microenvironment*gamma*Chemotherapeutic_drug_concentration__M
Vm = 0.45Reaction: => Chemotherapeutic_drug_concentration__M, Rate Law: Tumor_Microenvironment*Vm
beta = 0.012Reaction: Circulating_lymphocyte_population__C =>, Rate Law: Tumor_Microenvironment*beta*Circulating_lymphocyte_population__C
c1 = 3.41E-10Reaction: Tumor_Cell_Population__T => ; Effector_immune_cell_population__N, Rate Law: Tumor_Microenvironment*c1*Effector_immune_cell_population__N*Tumor_Cell_Population__T
Kn = 0.6; eta = 0.0Reaction: Effector_immune_cell_population__N => ; Chemotherapeutic_drug_concentration__M, Rate Law: Tumor_Microenvironment*Kn*(1-eta)*Chemotherapeutic_drug_concentration__M*Effector_immune_cell_population__N
kt = 3.2E-9Reaction: Chemotherapeutic_drug_concentration__M => ; Tumor_Cell_Population__T, Rate Law: Tumor_Microenvironment*kt*Tumor_Cell_Population__T*Chemotherapeutic_drug_concentration__M
a = 0.431; b = 1.02E-14Reaction: => Tumor_Cell_Population__T, Rate Law: Tumor_Microenvironment*a*Tumor_Cell_Population__T*(1-b*Tumor_Cell_Population__T)
p = 2.0E-11Reaction: Effector_immune_cell_population__N => ; Tumor_Cell_Population__T, Rate Law: Tumor_Microenvironment*p*Tumor_Cell_Population__T*Effector_immune_cell_population__N
alpha2 = 7.5E8Reaction: => Circulating_lymphocyte_population__C, Rate Law: Tumor_Microenvironment*alpha2
mu = 0.0412Reaction: Effector_immune_cell_population__N =>, Rate Law: Tumor_Microenvironment*mu*Effector_immune_cell_population__N
alpha1 = 12000.0Reaction: => Effector_immune_cell_population__N, Rate Law: Tumor_Microenvironment*alpha1
s = 20.2; g = 0.015Reaction: => Effector_immune_cell_population__N; Tumor_Cell_Population__T, Rate Law: Tumor_Microenvironment*g*Tumor_Cell_Population__T/(s+Tumor_Cell_Population__T)*Effector_immune_cell_population__N
Kc = 0.6; eta = 0.0Reaction: Circulating_lymphocyte_population__C => ; Chemotherapeutic_drug_concentration__M, Rate Law: Tumor_Microenvironment*Kc*(1-eta)*Chemotherapeutic_drug_concentration__M*Circulating_lymphocyte_population__C
Kt = 0.8Reaction: Tumor_Cell_Population__T => ; Chemotherapeutic_drug_concentration__M, Rate Law: Tumor_Microenvironment*Kt*Chemotherapeutic_drug_concentration__M*Tumor_Cell_Population__T

States:

NameDescription
Tumor Cell Population T[Neoplastic Cell]
Effector immune cell population N[OBI_1110016]
Circulating lymphocyte population C[BTO:0000775; OBI_0000181]
Chemotherapeutic drug concentration M[drug]

Livshitz2007_CardiacMyocytes: MODEL0406270966v0.0.1

This a model from the article: Regulation of Ca2+ and electrical alternans in cardiac myocytes: role of CAMKII and rep…

Details

Alternans of cardiac repolarization is associated with arrhythmias and sudden death. At the cellular level, alternans involves beat-to-beat oscillation of the action potential (AP) and possibly Ca(2+) transient (CaT). Because of experimental difficulty in independently controlling the Ca(2+) and electrical subsystems, mathematical modeling provides additional insights into mechanisms and causality. Pacing protocols were conducted in a canine ventricular myocyte model with the following results: 1) CaT alternans results from refractoriness of the sarcoplasmic reticulum Ca(2+) release system; alternation of the L-type calcium current has a negligible effect; 2) CaT-AP coupling during late AP occurs through the sodium-calcium exchanger and underlies AP duration (APD) alternans; 3) increased Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activity extends the range of CaT and APD alternans to slower frequencies and increases alternans magnitude; its decrease suppresses CaT and APD alternans, exerting an antiarrhythmic effect; and 4) increase of the rapid delayed rectifier current (I(Kr)) also suppresses APD alternans but without suppressing CaT alternans. Thus CaMKII inhibition eliminates APD alternans by eliminating its cause (CaT alternans) while I(Kr) enhancement does so by weakening CaT-APD coupling. The simulations identify combined CaMKII inhibition and I(Kr) enhancement as a possible antiarrhythmic intervention. link: http://identifiers.org/pubmed/17277017

LiX2019 - macrophage polarization and tumor cell plasticity: MODEL1909230002v0.0.1

This model is based on: Computational Modeling of the Crosstalk Between Macrophage Polarization and Tumor Cell Plastici…

Details

Tumor microenvironments contain multiple cell types interacting among one another via different signaling pathways. Furthermore, both cancer cells and different immune cells can display phenotypic plasticity in response to these communicating signals, thereby leading to complex spatiotemporal patterns that can impact therapeutic response. Here, we investigate the crosstalk between cancer cells and macrophages in a tumor microenvironment through in silico (computational) co-culture models. In particular, we investigate how macrophages of different polarization (M1 vs. M2) can interact with epithelial-mesenchymal plasticity of cancer cells, and conversely, how cancer cells exhibiting different phenotypes (epithelial vs. mesenchymal) can influence the polarization of macrophages. Based on interactions documented in the literature, an interaction network of cancer cells and macrophages is constructed. The steady states of the network are then analyzed. Various interactions were removed or added into the constructed-network to test the functions of those interactions. Also, parameters in the mathematical models were varied to explore their effects on the steady states of the network. In general, the interactions between cancer cells and macrophages can give rise to multiple stable steady-states for a given set of parameters and each steady state is stable against perturbations. Importantly, we show that the system can often reach one type of stable steady states where cancer cells go extinct. Our results may help inform efficient therapeutic strategies. link: http://identifiers.org/pubmed/30729096

Liò2012_Modelling osteomyelitis_Control Model: BIOMD0000000923v0.0.1

Background This work focuses on the computational modelling of osteomyelitis, a bone pathology caused by bacteria infect…

Details

BACKGROUND: This work focuses on the computational modelling of osteomyelitis, a bone pathology caused by bacteria infection (mostly Staphylococcus aureus). The infection alters the RANK/RANKL/OPG signalling dynamics that regulates osteoblasts and osteoclasts behaviour in bone remodelling, i.e. the resorption and mineralization activity. The infection rapidly leads to severe bone loss, necrosis of the affected portion, and it may even spread to other parts of the body. On the other hand, osteoporosis is not a bacterial infection but similarly is a defective bone pathology arising due to imbalances in the RANK/RANKL/OPG molecular pathway, and due to the progressive weakening of bone structure. RESULTS: Since both osteoporosis and osteomyelitis cause loss of bone mass, we focused on comparing the dynamics of these diseases by means of computational models. Firstly, we performed meta-analysis on a gene expression data of normal, osteoporotic and osteomyelitis bone conditions. We mainly focused on RANKL/OPG signalling, the TNF and TNF receptor superfamilies and the NF-kB pathway. Using information from the gene expression data we estimated parameters for a novel model of osteoporosis and of osteomyelitis. Our models could be seen as a hybrid ODE and probabilistic verification modelling framework which aims at investigating the dynamics of the effects of the infection in bone remodelling. Finally we discuss different diagnostic estimators defined by formal verification techniques, in order to assess different bone pathologies (osteopenia, osteoporosis and osteomyelitis) in an effective way. CONCLUSIONS: We present a modeling framework able to reproduce aspects of the different bone remodeling defective dynamics of osteomyelitis and osteoporosis. We report that the verification-based estimators are meaningful in the light of a feed forward between computational medicine and clinical bioinformatics. link: http://identifiers.org/pubmed/23095605

Parameters:

NameDescription
s = 100.0; beta2 = 0.02; g22 = 0.0; g12 = 1.0; f22 = 0.2; f12 = 0.0; alpha2 = 4.0Reaction: => Osteoblasts__O_b; Osteoclasts__O_c, B, Rate Law: compartment*(alpha2*Osteoclasts__O_c^(g12*(1+f12*B/s))*Osteoblasts__O_b^(g22-f22*B/s)-beta2*Osteoblasts__O_b)
O_bbar = 177.91; O_cbar = 1.78; k2 = 6.395E-4; k1 = 0.0748Reaction: => Bone_Density__z; Osteoclasts__O_c, Osteoblasts__O_b, Rate Law: compartment*((-k1)*piecewise(Osteoclasts__O_c-O_cbar, (Osteoclasts__O_c-O_cbar) >= 0, 0)+k2*piecewise(Osteoblasts__O_b-O_bbar, (Osteoblasts__O_b-O_bbar) >= 0, 0))
s = 100.0; V = 0.007; gamma_B = 0.005Reaction: => B, Rate Law: compartment*(gamma_B-V)*B*log(10, s/B)
s = 100.0; g11 = 1.1; beta1 = 0.2; alpha1 = 3.0; f11 = 0.005; g21 = -0.5; f21 = 0.005Reaction: => Osteoclasts__O_c; B, Osteoblasts__O_b, Rate Law: compartment*(alpha1*Osteoclasts__O_c^(g11*(1+f11*B/s))*Osteoblasts__O_b^(g21*(1+f21*B/s))-beta1*Osteoclasts__O_c)

States:

NameDescription
B[C50921]
Bone Density z[0016250]
Osteoclasts O c[0011016]
Osteoblasts O b[0011012]

LlorénsRico2016 - Effects of cis-Encoded antisense RNAs (asRNAs) - Case1: MODEL1511170000v0.0.1

LlorénsRico2016 - Effects of cis-Encoded antisense RNAs (asRNAs) - Case1Three putative effects of the asRNAs were consi…

Details

cis-Encoded antisense RNAs (asRNAs) are widespread along bacterial transcriptomes. However, the role of most of these RNAs remains unknown, and there is an ongoing discussion as to what extent these transcripts are the result of transcriptional noise. We show, by comparative transcriptomics of 20 bacterial species and one chloroplast, that the number of asRNAs is exponentially dependent on the genomic AT content and that expression of asRNA at low levels exerts little impact in terms of energy consumption. A transcription model simulating mRNA and asRNA production indicates that the asRNA regulatory effect is only observed above certain expression thresholds, substantially higher than physiological transcript levels. These predictions were verified experimentally by overexpressing nine different asRNAs in Mycoplasma pneumoniae. Our results suggest that most of the antisense transcripts found in bacteria are the consequence of transcriptional noise, arising at spurious promoters throughout the genome. link: http://identifiers.org/pubmed/26973873

LlorénsRico2016 - Effects of cis-Encoded antisense RNAs (asRNAs) - Case2: MODEL1511170001v0.0.1

LlorénsRico2016 - Effects of cis-Encoded antisense RNAs (asRNAs) - Case1Three putative effects of the asRNAs were consi…

Details

cis-Encoded antisense RNAs (asRNAs) are widespread along bacterial transcriptomes. However, the role of most of these RNAs remains unknown, and there is an ongoing discussion as to what extent these transcripts are the result of transcriptional noise. We show, by comparative transcriptomics of 20 bacterial species and one chloroplast, that the number of asRNAs is exponentially dependent on the genomic AT content and that expression of asRNA at low levels exerts little impact in terms of energy consumption. A transcription model simulating mRNA and asRNA production indicates that the asRNA regulatory effect is only observed above certain expression thresholds, substantially higher than physiological transcript levels. These predictions were verified experimentally by overexpressing nine different asRNAs in Mycoplasma pneumoniae. Our results suggest that most of the antisense transcripts found in bacteria are the consequence of transcriptional noise, arising at spurious promoters throughout the genome. link: http://identifiers.org/pubmed/26973873

LlorénsRico2016 - Effects of cis-Encoded antisense RNAs (asRNAs) - Case3: MODEL1511170002v0.0.1

LlorénsRico2016 - Effects of cis-Encoded antisense RNAs (asRNAs) - Case3Three putative effects of the asRNAs were consi…

Details

cis-Encoded antisense RNAs (asRNAs) are widespread along bacterial transcriptomes. However, the role of most of these RNAs remains unknown, and there is an ongoing discussion as to what extent these transcripts are the result of transcriptional noise. We show, by comparative transcriptomics of 20 bacterial species and one chloroplast, that the number of asRNAs is exponentially dependent on the genomic AT content and that expression of asRNA at low levels exerts little impact in terms of energy consumption. A transcription model simulating mRNA and asRNA production indicates that the asRNA regulatory effect is only observed above certain expression thresholds, substantially higher than physiological transcript levels. These predictions were verified experimentally by overexpressing nine different asRNAs in Mycoplasma pneumoniae. Our results suggest that most of the antisense transcripts found in bacteria are the consequence of transcriptional noise, arising at spurious promoters throughout the genome. link: http://identifiers.org/pubmed/26973873

Lo2005 - Stochastic Modeling of Blood Coagulation Initiation: MODEL1805160001v0.0.1

Simulation results using a stochastic approach to Hockin et al. (2002) mathematical model of the blood coagulation casca…

Details

A kinetic Monte Carlo simulation was developed using the deterministic reaction network developed by the Mann laboratory for tissue-factor (TF)-initiated blood coagulation. The model predicted thrombin dynamics in recalcified whole blood (3-fold diluted) pretreated with convulxin (platelet GPVI activator) and picomolar levels of TF (0-14 pM). The model did not accurately predict coagulation times at low TF (0-0.7 pM). The simulation revealed that approximately 0.2 pM TF was the critical concentration to cause 50% of reactions containing 3-fold diluted whole blood to reach a clotting threshold of 0.05 U/ml thrombin by 1 h. Simulations of 1 nl of blood (5 pM TF) revealed small stochastic variations in thrombin initiation time, while 16.6 pl simulations were highly stochastic at this level of TF (50 molecules/16.6 pl). Further experiment and simulation will require evaluation of mechanisms of coagulation kinetics at subpicomolar levels of TF. link: http://identifiers.org/pubmed/16432310

Loccisano2011-pharmacokinetics of PFOA and PFOS in the monkey: MODEL2003190002v0.0.1

Perfluoroalkyl acid carboxylates and sulfonates (PFAAs) have many consumer and industrial applications. The persistence…

Details

Perfluoroalkyl acid carboxylates and sulfonates (PFAAs) have many consumer and industrial applications. The persistence and widespread distribution of these compounds in humans have brought them under intense scrutiny. Limited pharmacokinetic data is available in humans; however, human data exists for two communities with drinking water contaminated by PFAAs. Also, there is toxicological and pharmacokinetic data for monkeys, which can be quite useful for cross-species extrapolation to humans. The goal of this research was to develop a physiologically-based pharmacokinetic (PBPK) model for PFOA and PFOS for monkeys and then scale this model to humans in order to describe available human drinking water data. The monkey model simulations were consistent with available PK data for monkeys. The monkey model was then extrapolated to the human and then used to successfully simulate the data collected from residents of two communities exposed to PFOA in drinking water. Human PFOS data is minimal; however, using the half-life estimated from occupational exposure, our model exhibits reasonable agreement with the available human serum PFOS data. It is envisioned that our PBPK model will be useful in supporting human health risk assessments for PFOA and PFOS by aiding in understanding of human pharmacokinetics. link: http://identifiers.org/pubmed/21168463

Locke2005 - Circadian Clock: BIOMD0000000055v0.0.1

Locke2005 - Circadian ClockSBML model of the interlocked feedback loop network The model describes the circuit depicted…

Details

Circadian clocks involve feedback loops that generate rhythmic expression of key genes. Molecular genetic studies in the higher plant Arabidopsis thaliana have revealed a complex clock network. The first part of the network to be identified, a transcriptional feedback loop comprising TIMING OF CAB EXPRESSION 1 (TOC1), LATE ELONGATED HYPOCOTYL (LHY) and CIRCADIAN CLOCK ASSOCIATED 1 (CCA1), fails to account for significant experimental data. We develop an extended model that is based upon a wider range of data and accurately predicts additional experimental results. The model comprises interlocking feedback loops comparable to those identified experimentally in other circadian systems. We propose that each loop receives input signals from light, and that each loop includes a hypothetical component that had not been explicitly identified. Analysis of the model predicted the properties of these components, including an acute light induction at dawn that is rapidly repressed by LHY and CCA1. We found this unexpected regulation in RNA levels of the evening-expressed gene GIGANTEA (GI), supporting our proposed network and making GI a strong candidate for this component. link: http://identifiers.org/pubmed/16729048

Parameters:

NameDescription
k2 = 1.5644; m2 = 20.44; Fch_0=8.0Reaction: cLc =>, Rate Law: compartment*m2*cLc/(k2+cLc)
r3 = 0.3166Reaction: cTc => cTn, Rate Law: compartment*r3*cTc
r4 = 2.1509Reaction: cTn => cTc, Rate Law: compartment*r4*cTn
r1 = 16.8363Reaction: cLc => cLn, Rate Law: compartment*r1*cLc
m3 = 3.6888; k3 = 1.2765Reaction: cLn =>, Rate Law: compartment*m3*cLn/(k3+cLn)
p2 = 4.324Reaction: => cTc; cTm, Rate Law: compartment*p2*cTm
r2 = 0.1687Reaction: cLn => cLc, Rate Law: compartment*r2*cLn
r7 = 2.2123Reaction: cYc => cYn, Rate Law: compartment*r7*cYc
m9 = 10.1132; k7 = 6.5585Reaction: cXm =>, Rate Law: compartment*m9*cXm/(k7+cXm)
k9 = 17.1111; m11 = 3.3442Reaction: cXn =>, Rate Law: compartment*m11*cXn/(k9+cXn)
p1 = 0.8295Reaction: => cLc; cLm, Rate Law: compartment*p1*cLm
p4 = 0.2485Reaction: => cYc; cYm, Rate Law: compartment*p4*cYm
k4 = 2.5734; m4 = 3.8231Reaction: cTm =>, Rate Law: compartment*m4*cTm/(k4+cTm)
m8 = 4.0424; dayLength = 12.0; k6 = 0.4033; m7 = 0.0492Reaction: cTn =>, Rate Law: compartment*((1-ceil(sin(pi*t/dayLength+0.001)/2))*m7+m8)*cTn/(k6+cTn)
r8 = 0.2002Reaction: cYn => cYc, Rate Law: compartment*r8*cYn
p3 = 2.147Reaction: => cXc; cXm, Rate Law: compartment*p3*cXm
m14 = 0.6114; k12 = 1.8066Reaction: cYn =>, Rate Law: compartment*m14*cYn/(k12+cYn)
dayLength = 12.0; p5 = 0.5Reaction: => cPn, Rate Law: compartment*(1-ceil(sin(pi*t/dayLength+0.001)/2))*p5
r5 = 1.0352Reaction: cXc => cXn, Rate Law: compartment*r5*cXc
dayLength = 12.0; q3 = 1.0Reaction: cPn =>, Rate Law: compartment*q3*ceil(sin(pi*t/dayLength+0.001)/2)*cPn
g5 = 1.17803247; e = 3.6064; dayLength = 12.0; n4 = 0.0857; g6 = 0.064455137; q2 = 2.40178; n5 = 0.1649; f = 1.0237Reaction: => cYm; cPn, cTn, cLn, Rate Law: compartment*(ceil(sin(pi*t/dayLength+0.001)/2)*q2*cPn+(ceil(sin(pi*t/dayLength+0.001)/2)*n4+n5)*g5^e/(g5^e+cTn^e))*g6^f/(g6^f+cLn^f)
a = 3.3064; Fch_0=8.0; n1 = 5.1694; g1 = 0.876738488Reaction: => cLm; cXn, Rate Law: compartment*n1*cXn^a/(g1^a+cXn^a)
k13 = 1.2; m15 = 1.2Reaction: cPn =>, Rate Law: compartment*m15*cPn/(k13+cPn)
dayLength = 12.0; m5 = 0.0013; m6 = 3.1741; k5 = 2.7454Reaction: cTc =>, Rate Law: compartment*((1-ceil(sin(pi*t/dayLength+0.001)/2))*m5+m6)*cTc/(k5+cTc)
k11 = 1.8258; m13 = 0.1347Reaction: cYc =>, Rate Law: compartment*m13*cYc/(k11+cYc)
dayLength = 12.0; q1 = 2.4514Reaction: => cLm; cPn, Rate Law: compartment*ceil(sin(pi*t/dayLength+0.001)/2)*q1*cPn
k1 = 1.817; m1 = 1.5283Reaction: cLm =>, Rate Law: compartment*m1*cLm/(k1+cLm)
g2 = 0.036805783; b = 1.0258; n2 = 3.0087; c = 1.0258; g3 = 0.26593318Reaction: => cTm; cYn, cLn, Rate Law: compartment*n2*cYn^b/(g2^b+cYn^b)*g3^c/(g3^c+cLn^c)
r6 = 3.3017Reaction: cXn => cXc, Rate Law: compartment*r6*cXn
m10 = 0.2179; k8 = 0.6632Reaction: cXc =>, Rate Law: compartment*m10*cXc/(k8+cXc)
k10 = 1.7303; m12 = 4.297Reaction: cYm =>, Rate Law: compartment*m12*cYm/(k10+cYm)
n3 = 0.2431; d = 1.4422; g4 = 0.538811228Reaction: => cXm; cTn, Rate Law: compartment*n3*cTn^d/(g4^d+cTn^d)

States:

NameDescription
cXm[messenger RNA; RNA]
cTc[Two-component response regulator-like APRR1]
cTn[Two-component response regulator-like APRR1]
cXcX protein in cytoplasm
cXn[transcription factor complex]
cYm[messenger RNA; RNA]
cLn[Protein LHY]
cYn[transcription factor complex]
cPnlight sensitive protein P
cYcY protein in cytoplasm
cLc[Protein LHY]
cLm[messenger RNA; RNA]
cTm[messenger RNA; RNA]

Locke2006_CircClock_LL: BIOMD0000000089v0.0.1

This a model from the article: Experimental validation of a predicted feedback loop in the multi-oscillator clock of…

Details

Our computational model of the circadian clock comprised the feedback loop between LATE ELONGATED HYPOCOTYL (LHY), CIRCADIAN CLOCK ASSOCIATED 1 (CCA1) and TIMING OF CAB EXPRESSION 1 (TOC1), and a predicted, interlocking feedback loop involving TOC1 and a hypothetical component Y. Experiments based on model predictions suggested GIGANTEA (GI) as a candidate for Y. We now extend the model to include a recently demonstrated feedback loop between the TOC1 homologues PSEUDO-RESPONSE REGULATOR 7 (PRR7), PRR9 and LHY and CCA1. This three-loop network explains the rhythmic phenotype of toc1 mutant alleles. Model predictions fit closely to new data on the gi;lhy;cca1 mutant, which confirm that GI is a major contributor to Y function. Analysis of the three-loop network suggests that the plant clock consists of morning and evening oscillators, coupled intracellularly, which may be analogous to coupled, morning and evening clock cells in Drosophila and the mouse. link: http://identifiers.org/pubmed/17102804

Parameters:

NameDescription
light = 1.0; q4 = 2.4514Reaction: => cAm; cPn, Rate Law: light*q4*cPn*compartment
light = 1.0; k6 = 0.4033; m7 = 0.0492Reaction: cTn =>, Rate Law: compartment*(1-light)*m7*cTn/(k6+cTn)
light = 1.0; q3 = 1.0Reaction: cPn =>, Rate Law: q3*light*cPn*compartment
r3 = 0.3166; r4 = 2.1509Reaction: cTc => cTn, Rate Law: compartment*((-r4)*cTn+r3*cTc)
m3 = 3.6888; k3 = 1.2765Reaction: cLn =>, Rate Law: compartment*m3*cLn/(k3+cLn)
p2 = 4.324Reaction: => cTc; cTm, Rate Law: p2*compartment*cTm
m18 = 0.0156; k16 = 0.6104Reaction: cAn =>, Rate Law: compartment*m18*cAn/(k16+cAn)
r8 = 0.2002; r7 = 2.2123Reaction: cYc => cYn, Rate Law: compartment*(r7*cYc-r8*cYn)
r6 = 3.3017; r5 = 1.0352Reaction: cXc => cXn, Rate Law: compartment*(r5*cXc-r6*cXn)
k2 = 1.5644; m2 = 20.44Reaction: cLc =>, Rate Law: compartment*m2*cLc/(k2+cLc)
m9 = 10.1132; k7 = 6.5585Reaction: cXm =>, Rate Law: compartment*m9*cXm/(k7+cXm)
k9 = 17.1111; m11 = 3.3442Reaction: cXn =>, Rate Law: compartment*m11*cXn/(k9+cXn)
p1 = 0.8295Reaction: => cLc; cLm, Rate Law: compartment*p1*cLm
r9 = 0.2528; r10 = 0.2212Reaction: cAc => cAn, Rate Law: compartment*(r9*cAc-r10*cAn)
k15 = 0.0703; m17 = 4.4505Reaction: cAc =>, Rate Law: compartment*m17*cAc/(k15+cAc)
m8 = 4.0424; k6 = 0.4033Reaction: cTn =>, Rate Law: m8*compartment*cTn/(k6+cTn)
p4 = 0.2485Reaction: => cYc; cYm, Rate Law: compartment*p4*cYm
k4 = 2.5734; m4 = 3.8231Reaction: cTm =>, Rate Law: compartment*m4*cTm/(k4+cTm)
k1 = 2.392; m1 = 1.999Reaction: cLm =>, Rate Law: compartment*m1*cLm/(k1+cLm)
r1 = 16.8363; r2 = 0.1687Reaction: cLc => cLn, Rate Law: compartment*(r1*cLc-r2*cLn)
b = 1.0258; n2 = 3.0087; g3 = 0.2658; g2 = 0.0368; c = 1.0258Reaction: => cTm; cYn, cLn, Rate Law: compartment*n2*cYn^b/(g2^b+cYn^b)*g3^c/(g3^c+cLn^c)
m14 = 0.6114; k12 = 1.8066Reaction: cYn =>, Rate Law: compartment*m14*cYn/(k12+cYn)
p3 = 2.147Reaction: => cXc; cXm, Rate Law: compartment*p3*cXm
light = 1.0; m5 = 0.0013; k5 = 2.7454Reaction: cTc =>, Rate Law: compartment*(1-light)*m5*cTc/(k5+cTc)
p6 = 0.2907Reaction: => cAc; cAm, Rate Law: compartment*p6*cAm
m6 = 3.1741; k5 = 2.7454Reaction: cTc =>, Rate Law: m6*compartment*cTc/(k5+cTc)
n3 = 0.2431; d = 1.4422; g4 = 0.5388Reaction: => cXm; cTn, Rate Law: compartment*n3*cTn^d/(g4^d+cTn^d)
light = 1.0; p5 = 0.5Reaction: => cPn, Rate Law: (1-light)*p5*compartment
k13 = 1.2; m15 = 1.2Reaction: cPn =>, Rate Law: compartment*m15*cPn/(k13+cPn)
k11 = 1.8258; m13 = 0.1347Reaction: cYc =>, Rate Law: compartment*m13*cYc/(k11+cYc)
alpha = 4.0; n1 = 7.8142; g1 = 3.1383; a = 1.2479; light = 1.0; g0 = 1.0; n0 = 0.05; q1 = 4.1954Reaction: => cLm; cAn, cXn, cPn, Rate Law: compartment*g0^alpha/(g0^alpha+cAn^alpha)*(light*(q1*cPn+n0)+n1*cXn^a/(g1^a+cXn^a))
n6 = 8.0706; g7 = 4.0E-4; g = 1.0258Reaction: => cAm; cLn, Rate Law: compartment*n6*cLn^g/(g7^g+cLn^g)
k10 = 1.7303; m12 = 4.297Reaction: cYm =>, Rate Law: compartment*m12*cYm/(k10+cYm)
m16 = 12.2398; k14 = 10.3617Reaction: cAm =>, Rate Law: compartment*m16*cAm/(k14+cAm)
m10 = 0.2179; k8 = 0.6632Reaction: cXc =>, Rate Law: compartment*m10*cXc/(k8+cXc)
e = 3.6064; light = 1.0; n4 = 0.0857; q2 = 2.4017; n5 = 0.1649; g5 = 1.178; f = 1.0237; g6 = 0.0645Reaction: => cYm; cTn, cLn, cPn, Rate Law: compartment*(light*q2*cPn+(light*n4+n5)*g5^e/(g5^e+cTn^e))*g6^f/(g6^f+cLn^f)

States:

NameDescription
cXm[messenger RNA]
cTc[Two-component response regulator-like APRR1]
cTn[Two-component response regulator-like APRR1]
cXcX protein in cytoplasm
cAc[Two-component response regulator-like APRR7; Two-component response regulator-like APRR9]
cXnX protein in nucleus
cYm[messenger RNA]
cLn[Protein CCA1; Protein LHY]
cYn[Protein GIGANTEA]
cPnlight sensitive protein P
cYc[Protein GIGANTEA]
cAn[Two-component response regulator-like APRR7; Two-component response regulator-like APRR9]
cLc[Protein LHY; Protein CCA1]
cLm[messenger RNA]
cTm[messenger RNA]
cAm[messenger RNA]

Locke2008_Circadian_Clock: BIOMD0000000185v0.0.1

The model reproduces Fig 2A of the paper. Model successfully reproduced using Jarnac and MathSBML. To the extent possib…

Details

BACKGROUND: Virtually all living organisms have evolved a circadian (~24 hour) clock that controls physiological and behavioural processes with exquisite precision throughout the day/night cycle. The suprachiasmatic nucleus (SCN), which generates these ~24 h rhythms in mammals, consists of several thousand neurons. Each neuron contains a gene-regulatory network generating molecular oscillations, and the individual neuron oscillations are synchronised by intercellular coupling, presumably via neurotransmitters. Although this basic mechanism is currently accepted and has been recapitulated in mathematical models, several fundamental questions about the design principles of the SCN remain little understood. For example, a remarkable property of the SCN is that the phase of the SCN rhythm resets rapidly after a 'jet lag' type experiment, i.e. when the light/dark (LD) cycle is abruptly advanced or delayed by several hours. RESULTS: Here, we describe an extensive parameter optimization of a previously constructed simplified model of the SCN in order to further understand its design principles. By examining the top 50 solutions from the parameter optimization, we show that the neurotransmitters' role in generating the molecular circadian rhythms is extremely important. In addition, we show that when a neurotransmitter drives the rhythm of a system of coupled damped oscillators, it exhibits very robust synchronization and is much more easily entrained to light/dark cycles. We were also able to recreate in our simulations the fast rhythm resetting seen after a 'jet lag' type experiment. CONCLUSION: Our work shows that a careful exploration of parameter space for even an extremely simplified model of the mammalian clock can reveal unexpected behaviours and non-trivial predictions. Our results suggest that the neurotransmitter feedback loop plays a crucial role in the robustness and phase resetting properties of the mammalian clock, even at the single neuron level. link: http://identifiers.org/pubmed/18312618

Parameters:

NameDescription
k3 = 0.1177 hr_1Reaction: => Y1; X1, Rate Law: compartment*k3*X1
k7 = 0.2282 hr_1Reaction: => V1; X1, Rate Law: compartment*k7*X1
F = 0.0 nM; vc = 6.7924 nM_hr_1; K = 1.0 dimensionless; Kc = 4.8283 nMReaction: => X1, Rate Law: compartment*vc*K*F/(Kc+K*F)
k5 = 0.3352 hr_1Reaction: => Z1; Y1, Rate Law: compartment*k5*Y1
K2 = 0.291 nM; v_2 = 8.4297 nM_hr_1Reaction: X1 =>, Rate Law: compartment*v_2*X1/(K2+X1)
v_4 = 1.0841 nM_hr_1; K4 = 8.1343 nMReaction: Y1 =>, Rate Law: compartment*v_4*Y1/(K4+Y1)
L = 0.0 nM_hr_1Reaction: => X1, Rate Law: compartment*L
K1 = 2.7266 nM; n = 5.6645 dimensionless; v_1 = 6.8355 nM_hr_1Reaction: => X1; Z1, Rate Law: compartment*v_1*K1^n/(K1^n+Z1^n)
v_6 = 4.6645 nM_hr_1; K6 = 9.9849 nMReaction: Z1 =>, Rate Law: compartment*v_6*Z1/(K6+Z1)
v_8 = 3.5216 nM_hr_1; K8 = 7.4519 nMReaction: V1 =>, Rate Law: compartment*v_8*V1/(K8+V1)

States:

NameDescription
V2[Neuropeptide F; neuropeptide hormone activity]
Y2[Circadian locomoter output cycles protein kaput]
Z2Transcriptional repressor
X2[Circadian locomoter output cycles protein kaput; messenger RNA]
Y1[Circadian locomoter output cycles protein kaput]
X1[messenger RNA; Circadian locomoter output cycles protein kaput]
Z1Transcriptional repressor
V1[Neuropeptide F; neuropeptide hormone activity]

Lockwood2006 - Alzheimer's Disease PBPK model: BIOMD0000000673v0.0.1

Lockwood2006 - AlzheimersDisease PBPK modelA mathematical model to predict the effectiveness of CI-1017 (muscarinic agon…

Details

Clinical trial simulation (CTS) was used to select a robust design to test the hypothesis that a new treatment was effective for Alzheimer's disease (AD). Typically, a parallel group, placebo controlled, 12-week trial in 200-400 AD patients would be used to establish drug effect relative to placebo (i.e., Ho: Drug Effect = 0). We evaluated if a crossover design would allow smaller and shorter duration trials.A family of plausible drug and disease models describing the time course of the AD assessment scale (ADAS-Cog) was developed based on Phase I data and literature reports of other treatments for AD. The models included pharmacokinetic, pharmacodynamic, disease progression, and placebo components. Eight alternative trial designs were explored via simulation. One hundred replicates of each combination of drug and disease model and trial design were simulated. A 'positive trial' reflecting drug activity was declared considering both a dose trend test (p < 0.05) and pair-wise comparisons to placebo (p < 0.025).A 4 x 4 Latin Square design was predicted to have at least 80% power to detect activity across a range of drug and disease models. The trial design was subsequently implemented and the trial was completed. Based on the results of the actual trial, a conclusive decision about further development was taken. The crossover design provided enhanced power over a parallel group design due to the lower residual variability.CTS aided the decision to use a more efficient proof of concept trial design, leading to savings of up to US 4 M dollars in direct costs and a firm decision 8-12 months earlier than a 12-week parallel group trial. link: http://identifiers.org/pubmed/16906456

Loira2012 - Metabolic Network of Y.lipolytica: MODEL1111190000v0.0.1

This model is from the article: A genome-scale metabolic model of the lipid-accumulating yeast Yarrowia lipolytica N…

Details

BACKGROUND: Yarrowia lipolytica is an oleaginous yeast which has emerged as an important microorganism for several biotechnological processes, such as the production of organic acids, lipases and proteases. It is also considered a good candidate for single-cell oil production. Although some of its metabolic pathways are well studied, its metabolic engineering is hindered by the lack of a genome-scale model that integrates the current knowledge about its metabolism. RESULTS: Combining in silico tools and expert manual curation, we have produced an accurate genome-scale metabolic model for Y. lipolytica. Using a scaffold derived from a functional metabolic model of the well-studied but phylogenetically distant yeast S. cerevisiae, we mapped conserved reactions, rewrote gene associations, added species-specific reactions and inserted specialized copies of scaffold reactions to account for species-specific expansion of protein families. We used physiological measures obtained under lab conditions to validate our predictions. CONCLUSIONS: Y. lipolytica iNL895 represents the first well-annotated metabolic model of an oleaginous yeast, providing a base for future metabolic improvement, and a starting point for the metabolic reconstruction of other species in the Yarrowia clade and other oleaginous yeasts. link: http://identifiers.org/pubmed/22558935

Lolas2016 - tumour-induced neoneurogenesis and perineural tumour growth: BIOMD0000000750v0.0.1

The paper describes a model of tumour-induced neoneurogenesis and perineural tumour growth. Created by COPASI 4.25 (Bu…

Details

It is well-known that tumours induce the formation of a lymphatic and a blood vasculature around themselves. A similar but far less studied process occurs in relation to the nervous system and is referred to as neoneurogenesis. The relationship between tumour progression and the nervous system is still poorly understood and is likely to involve a multitude of factors. It is therefore relevant to study tumour-nerve interactions through mathematical modelling: this may reveal the most significant factors of the plethora of interacting elements regulating neoneurogenesis. The present work is a first attempt to model the neurobiological aspect of cancer development through a system of differential equations. The model confirms the experimental observations that a tumour is able to promote nerve formation/elongation around itself, and that high levels of nerve growth factor and axon guidance molecules are recorded in the presence of a tumour. Our results also reflect the observation that high stress levels (represented by higher norepinephrine release by sympathetic nerves) contribute to tumour development and spread, indicating a mutually beneficial relationship between tumour cells and neurons. The model predictions suggest novel therapeutic strategies, aimed at blocking the stress effects on tumour growth and dissemination. link: http://identifiers.org/pubmed/26861829

Parameters:

NameDescription
delta = 0.0129 mm^3; dt = 0.0127 1/dReaction: Tp => ; A, Rate Law: tumor_microenvironment*dt*(1+delta*A)*Tp
cn = 0.41 1/(mm^3*d)Reaction: => Nn, Rate Law: tumor_microenvironment*cn
y4 = 1.47E-5 mm^3/d; y3 = 1.0E-5 mm^3/dReaction: A => ; Tp, S, P, Rate Law: tumor_microenvironment*(y3*Tp+y4*(S+P))*A
y5 = 0.002 mm^3/dReaction: Nn => ; Tp, Rate Law: tumor_microenvironment*y5*Tp*Nn
dt = 0.0127 1/dReaction: Tm =>, Rate Law: tumor_microenvironment*dt*Tm
ks = 0.26 1/mm^3; rs = 0.06 1/dReaction: => S, Rate Law: tumor_microenvironment*rs*(1-S/ks)*S
da = 49.91 1/dReaction: Na =>, Rate Law: tumor_microenvironment*da*Na
sigma4 = 0.01 d; sigma3 = 7.79 d/mm^3Reaction: => S; A, Rate Law: tumor_microenvironment*A*S/(sigma3+sigma4*A)
rtm = 1.0E-4 1/dReaction: => Tm, Rate Law: tumor_microenvironment*rtm*Tm
y1 = 5.57E-5 mm^3/d; y2 = 0.05 mm^3/dReaction: G => ; Tp, S, P, Rate Law: tumor_microenvironment*(y1*Tp+y2*(S+P))*G
dA = 2.4 1/dReaction: A =>, Rate Law: tumor_microenvironment*dA*A
sa = 0.73 1/dReaction: => Na; P, Rate Law: tumor_microenvironment*sa*P
dn = 1.66 1/dReaction: Nn =>, Rate Law: tumor_microenvironment*dn*Nn
sA = 0.00542 1/dReaction: => A; Tp, Rate Law: tumor_microenvironment*sA*Tp
sigma1 = 129.0 d/mm^3; sigma2 = 50.0 dReaction: => S; G, Rate Law: tumor_microenvironment*G*S/(sigma1+sigma2*G)
y6 = 0.001 mm^3/dReaction: Na => ; Tp, Rate Law: tumor_microenvironment*y6*Tp*Na
sg = 0.00222 1/dReaction: => G; Tp, Rate Law: tumor_microenvironment*sg*Tp
kp = 0.03 1/mm^3; rp = 7.0 1/dReaction: => P, Rate Law: tumor_microenvironment*rp*(1-P/kp)*P
t2 = 2.39 d; rtp = 4.81E-4 1/d; t1 = 134.27 d/mm^3; ONn = 6666.66666666667 1/mm^3; kt = 1000000.0 1/mm^3Reaction: => Tp; G, Rate Law: tumor_microenvironment*7/8*Tp*(rtp+G/(t1+t2*G))*(1-Tp/kt)*(Tp/ONn-1)
ca = 3990.0 1/(mm^3*d)Reaction: => Na, Rate Law: tumor_microenvironment*ca
u0 = 0.22 1/d; u1 = 9.8E-6 mm^3/d; u2 = 0.002 mm^3/dReaction: Tp => Tm; A, Na, Rate Law: tumor_microenvironment*(u0+u1*A+u2*Na)*Tp
sn = 1.6 1/dReaction: => Nn; S, Rate Law: tumor_microenvironment*sn*S
p3 = 1.0 d/mm^3; p4 = 0.01 dReaction: => P; A, Rate Law: tumor_microenvironment*A*P/(p3+p4*A)
dg = 22.18 1/dReaction: G =>, Rate Law: tumor_microenvironment*dg*G
p2 = 0.1 d; p1 = 0.33 1/dReaction: => P; G, Rate Law: tumor_microenvironment*G*P/(p1+p2*G)

States:

NameDescription
Na[acetylcholine]
A[axon guidance]
S[sympathetic neuron]
P[parasympathetic neuron]
Tp[neoplastic cell]
Nn[noradrenaline]
G[Growth Factor]
Tm[neoplastic cell]

Lombardo2018 - Expression of PDL1 in Neuroblastoma Cancer Cell: MODEL1812070002v0.0.1

The model reproduces the time profiles of PDL1 in a Neuroblastoma Cancer Cell, considering the activating mutation of AL…

Details

Immunotherapy is a promising new therapeutic approach for neuroblastoma (NBM): an anti‐GD2 vaccine combined with orally administered soluble beta‐glucan is undergoing a phase II clinical trial and nivolumab and ipilimumab are being tested in recurrent and refractory tumors. Unfortunately, predictive biomarkers of response to immunotherapy are currently not available for NBM patients. The aim of this study was to create a computational network model simulating the different intracellular pathways involved in NBM, in order to predict how the tumor phenotype may be influenced to increase the sensitivity to anti‐programmed cell death‐ligand‐1 (PD‐L1)/programmed cell death‐1 (PD‐1) immunotherapy. The model runs on COPASI software. In order to determine the influence of intracellular signaling pathways on the expression of PD‐L1 in NBM, we first developed an integrated network of protein kinase cascades. Michaelis–Menten kinetics were associated to each reaction in order to tailor the different enzymes kinetics, creating a system of ordinary differential equations (ODEs). The data of this study offers a first tool to be considered in the therapeutic management of the NBM patient undergoing immunotherapeutic treatment. link: http://identifiers.org/doi/10.3390/brainsci9090221

Lopez2014 - A Validated Mathematical Model of Tumor Growth Including Tumor-Host Interaction and Cell-Mediated Immune Response: BIOMD0000000784v0.0.1

This is a dynamical model of cancer growth that includes three interacting cell populations of tumor cells, healthy host…

Details

We consider a dynamical model of cancer growth including three interacting cell populations of tumor cells, healthy host cells and immune effector cells. The tumor-immune and the tumor-host interactions are characterized to reproduce experimental results. A thorough dynamical analysis of the model is carried out, showing its capability to explain theoretical and empirical knowledge about tumor development. A chemotherapy treatment reproducing different experiments is also introduced. We believe that this simple model can serve as a foundation for the development of more complicated and specific cancer models. link: http://identifiers.org/pubmed/25348062

Parameters:

NameDescription
d_3 = 0.112Reaction: z_Effector_Cells =>, Rate Law: compartment*d_3*z_Effector_Cells
r_2 = 0.35Reaction: => y_Healthy_Cells, Rate Law: compartment*r_2*y_Healthy_Cells*(1-y_Healthy_Cells)
D_x_z = 0.456947412742685Reaction: x_Tumor_Cells =>, Rate Law: compartment*D_x_z*x_Tumor_Cells
a_21 = 0.954Reaction: y_Healthy_Cells => ; x_Tumor_Cells, Rate Law: compartment*a_21*x_Tumor_Cells*y_Healthy_Cells
a_31 = 5.25Reaction: z_Effector_Cells => ; x_Tumor_Cells, Rate Law: compartment*a_31*x_Tumor_Cells*z_Effector_Cells
a_12 = 0.195Reaction: x_Tumor_Cells => ; y_Healthy_Cells, Rate Law: compartment*a_12*y_Healthy_Cells*x_Tumor_Cells
v=1.0Reaction: => z_Effector_Cells, Rate Law: compartment*v
g = 0.29; h = 7.95E-11; D_x_z = 0.456947412742685Reaction: => z_Effector_Cells; x_Tumor_Cells, Rate Law: compartment*g*D_x_z^2*x_Tumor_Cells^2/(h+D_x_z^2*x_Tumor_Cells^2)

States:

NameDescription
x Tumor Cells[neoplastic cell]
z Effector Cells[effector T cell]
y Healthy Cells[cell]

Louzoun2014 - A mathematical model for pancreatic cancer growth and treatments: MODEL1909100002v0.0.1

This is a mathematical model of pancreatic cancer, geared towards examining the efficacy of tumor-suppressing drugs in t…

Details

Pancreatic cancer is one of the most deadly types of cancer and has extremely poor prognosis. This malignancy typically induces only limited cellular immune responses, the magnitude of which can increase with the number of encountered cancer cells. On the other hand, pancreatic cancer is highly effective at evading immune responses by inducing polarization of pro-inflammatory M1 macrophages into anti-inflammatory M2 macrophages, and promoting expansion of myeloid derived suppressor cells, which block the killing of cancer cells by cytotoxic T cells. These factors allow immune evasion to predominate, promoting metastasis and poor responsiveness to chemotherapies and immunotherapies. In this paper we develop a mathematical model of pancreatic cancer, and use it to qualitatively explain a variety of biomedical and clinical data. The model shows that drugs aimed at suppressing cancer growth are effective only if the immune induced cancer cell death lies within a specific range, that is, the immune system has a specific window of opportunity to effectively suppress cancer under treatment. The model results suggest that tumor growth rate is affected by complex feedback loops between the tumor cells, endothelial cells and the immune response. The relative strength of the different loops determines the cancer growth rate and its response to immunotherapy. The model could serve as a starting point to identify optimal nodes for intervention against pancreatic cancer. link: http://identifiers.org/pubmed/24594371

Luan2007 - Blood coagulation model combining previous models and platelet activation: MODEL1806050001v0.0.1

Mathematical model combining previous models (Jones1994, Leipoldt1995, Kuhursky2001, Hockin2002) into one model with pla…

Details

The role that mechanistic mathematical modeling and systems biology will play in molecular medicine and clinical development remains uncertain. In this study, mathematical modeling and sensitivity analysis were used to explore the working hypothesis that mechanistic models of human cascades, despite model uncertainty, can be computationally screened for points of fragility, and that these sensitive mechanisms could serve as therapeutic targets. We tested our working hypothesis by screening a model of the well-studied coagulation cascade, developed and validated from literature. The predicted sensitive mechanisms were then compared with the treatment literature. The model, composed of 92 proteins and 148 protein-protein interactions, was validated using 21 published datasets generated from two different quiescent in vitro coagulation models. Simulated platelet activation and thrombin generation profiles in the presence and absence of natural anticoagulants were consistent with measured values, with a mean correlation of 0.87 across all trials. Overall state sensitivity coefficients, which measure the robustness or fragility of a given mechanism, were calculated using a Monte Carlo strategy. In the absence of anticoagulants, fluid and surface phase factor X/activated factor X (fX/FXa) activity and thrombin-mediated platelet activation were found to be fragile, while fIX/FIXa and fVIII/FVIIIa activation and activity were robust. Both anti-fX/FXa and direct thrombin inhibitors are important classes of anticoagulants; for example, anti-fX/FXa inhibitors have FDA approval for the prevention of venous thromboembolism following surgical intervention and as an initial treatment for deep venous thrombosis and pulmonary embolism. Both in vitro and in vivo experimental evidence is reviewed supporting the prediction that fIX/FIXa activity is robust. When taken together, these results support our working hypothesis that computationally derived points of fragility of human relevant cascades could be used as a rational basis for target selection despite model uncertainty. link: http://identifiers.org/pubmed/17658944

Luan2010 - Blood Coagulation Model (extension of Luan2007): MODEL1806250001v0.0.1

Mathematical model of blood coagulation. Extension of Luan2007. New reactions, e.g., the activation of platelet by throm…

Details

The role of mechanistic modeling and systems biology in molecular medicine remains unclear. In this study, we explored whether uncertain models could be used to understand how a network responds to a therapeutic intervention. As a proof of concept, we modeled and analyzed the response of the human coagulation cascade to recombinant factor VIIa (rFVIIa) and prothrombin (fII) addition in normal and hemophilic plasma. An ensemble of parametrically uncertain human coagulation models was developed (N = 437). Each model described the time evolution of 193 proteins and protein complexes interconnected by 301 interactions under quiescent flow. The 467 unknown model parameters were estimated, using multiobjective optimization, from published in vitro coagulation studies. The model ensemble was validated using published in vitro thrombin measurements and thrombin measurements taken from coronary artery disease patients. Sensitivity analysis was then used to rank-order the importance of model parameters as a function of experimental or physiological conditions. A novel strategy for the systematic comparison of ranks identified a family of fX/FXa and fII/FIIa interactions that became more sensitive with decreasing fVIII/fIX. The fragility of these interactions was preserved following the addition of exogenous rFVIIa and fII. This suggested that exogenous rFVIIa did not alter the qualitative operation of the cascade. Rather, exogenous rFVIIa and fII took advantage of existing fluid and interfacial fX/FXa and fII/FIIa sensitivity to restore normal coagulation in low fVIII/fIX conditions. The proposed rFVIIa mechanism of action was consistent with experimental literature not used in model training. Thus, we demonstrated that an ensemble of uncertain models could unravel key facets of the mechanism of action of a focused intervention. Whereas the current study was limited to coagulation, perhaps the general strategy used could be extended to other molecular networks relevant to human health. link: http://identifiers.org/pubmed/20844798

Luo1991_VentricularCardiacAction: MODEL0479527919v0.0.1

This a model from the article: A model of the ventricular cardiac action potential. Depolarization, repolarization, an…

Details

A mathematical model of the membrane action potential of the mammalian ventricular cell is introduced. The model is based, whenever possible, on recent single-cell and single-channel data and incorporates the possibility of changing extracellular potassium concentration [K]o. The fast sodium current, INa, is characterized by fast upstroke velocity (Vmax = 400 V/sec) and slow recovery from inactivation. The time-independent potassium current, IK1, includes a negative-slope phase and displays significant crossover phenomenon as [K]o is varied. The time-dependent potassium current, IK, shows only a minimal degree of crossover. A novel potassium current that activates at plateau potentials is included in the model. The simulated action potential duplicates the experimentally observed effects of changes in [K]o on action potential duration and rest potential. Physiological simulations focus on the interaction between depolarization and repolarization (i.e., premature stimulation). Results demonstrate the importance of the slow recovery of INa in determining the response of the cell. Simulated responses to periodic stimulation include monotonic Wenckebach patterns and alternans at normal [K]o, whereas at low [K]o nonmonotonic Wenckebach periodicities, aperiodic patterns, and enhanced supernormal excitability that results in unstable responses ("chaotic activity") are observed. The results are consistent with recent experimental observations, and the model simulations relate these phenomena to the underlying ionic channel kinetics. link: http://identifiers.org/pubmed/1709839

Luo1994_CardiacVentricularActionPotential: MODEL0912160003v0.0.1

This a model from the article: A dynamic model of the cardiac ventricular action potential. I. Simulations of ionic cu…

Details

A mathematical model of the cardiac ventricular action potential is presented. In our previous work, the membrane Na+ current and K+ currents were formulated. The present article focuses on processes that regulate intracellular Ca2+ and depend on its concentration. The model presented here for the mammalian ventricular action potential is based mostly on the guinea pig ventricular cell. However, it provides the framework for modeling other types of ventricular cells with appropriate modifications made to account for species differences. The following processes are formulated: Ca2+ current through the L-type channel (ICa), the Na(+)-Ca2+ exchanger, Ca2+ release and uptake by the sarcoplasmic reticulum (SR), buffering of Ca2+ in the SR and in the myoplasm, a Ca2+ pump in the sarcolemma, the Na(+)-K+ pump, and a nonspecific Ca(2+)-activated membrane current. Activation of ICa is an order of magnitude faster than in previous models. Inactivation of ICa depends on both the membrane voltage and [Ca2+]i. SR is divided into two subcompartments, a network SR (NSR) and a junctional SR (JSR). Functionally, Ca2+ enters the NSR and translocates to the JSR following a monoexponential function. Release of Ca2+ occurs at JSR and can be triggered by two different mechanisms, Ca(2+)-induced Ca2+ release and spontaneous release. The model provides the basis for the study of arrhythmogenic activity of the single myocyte including afterdepolarizations and triggered activity. It can simulate cellular responses under different degrees of Ca2+ overload. Such simulations are presented in our accompanying article in this issue of Circulation Research. link: http://identifiers.org/pubmed/7514509

M


Ma2002_cAMP_oscillations: BIOMD0000000229v0.0.1

This a model from the article: Quantifying robustness of biochemical network models. Ma L, Iglesias PA. BMC Bioinf…

Details

BACKGROUND: Robustness of mathematical models of biochemical networks is important for validation purposes and can be used as a means of selecting between different competing models. Tools for quantifying parametric robustness are needed. RESULTS: Two techniques for describing quantitatively the robustness of an oscillatory model were presented and contrasted. Single-parameter bifurcation analysis was used to evaluate the stability robustness of the limit cycle oscillation as well as the frequency and amplitude of oscillations. A tool from control engineering–the structural singular value (SSV)–was used to quantify robust stability of the limit cycle. Using SSV analysis, we find very poor robustness when the model's parameters are allowed to vary. CONCLUSION: The results show the usefulness of incorporating SSV analysis to single parameter sensitivity analysis to quantify robustness. link: http://identifiers.org/pubmed/12482327

Parameters:

NameDescription
k2 = 0.9Reaction: ACA => ; PKA, Rate Law: k2*ACA*PKA
k14 = 4.5Reaction: CAR1 =>, Rate Law: k14*CAR1
k3 = 2.5Reaction: => PKA; incAMP, Rate Law: k3*incAMP
k9 = 0.3Reaction: => incAMP; ACA, Rate Law: k9*ACA
k10 = 0.8Reaction: incAMP => ; REGA, Rate Law: k10*REGA*incAMP
k6 = 0.8Reaction: ERK2 => ; PKA, Rate Law: k6*PKA*ERK2
k11 = 0.7Reaction: => excAMP; ACA, Rate Law: k11*ACA
k12 = 4.9Reaction: excAMP =>, Rate Law: k12*excAMP
k5 = 0.6Reaction: => ERK2; CAR1, Rate Law: k5*CAR1
k7 = 1.0Reaction: => REGA, Rate Law: k7
k1 = 2.0Reaction: => ACA; CAR1, Rate Law: k1*CAR1
k13 = 23.0Reaction: => CAR1; excAMP, Rate Law: k13*excAMP
k8 = 1.3Reaction: REGA => ; ERK2, Rate Law: k8*ERK2*REGA
k4 = 1.5Reaction: PKA =>, Rate Law: k4*PKA

States:

NameDescription
excAMP[3',5'-cyclic AMP; C000575]
PKA[cAMP-dependent protein kinase regulatory subunit; IPR002373]
REGA[3',5'-cyclic-nucleotide phosphodiesterase regA]
ERK2[Extracellular signal-regulated kinase 2; IPR008349]
CAR1[Cyclic AMP receptor 1]
ACA[IPR008172]
incAMP[3',5'-cyclic AMP; C000575]

Ma2005-digital response of p53 to DNA damage-ATM Activation Module: MODEL2005130001v0.0.1

Recent observations show that the single-cell response of p53 to ionizing radiation (IR) is “digital” in that it is the…

Details

Recent observations show that the single-cell response of p53 to ionizing radiation (IR) is "digital" in that it is the number of oscillations rather than the amplitude of p53 that shows dependence on the radiation dose. We present a model of this phenomenon. In our model, double-strand break (DSB) sites induced by IR interact with a limiting pool of DNA repair proteins, forming DSB-protein complexes at DNA damage foci. The persisting complexes are sensed by ataxia telangiectasia mutated (ATM), a protein kinase that activates p53 once it is phosphorylated by DNA damage. The ATM-sensing module switches on or off the downstream p53 oscillator, consisting of a feedback loop formed by p53 and its negative regulator, Mdm2. In agreement with experiments, our simulations show that by assuming stochasticity in the initial number of DSBs and the DNA repair process, p53 and Mdm2 exhibit a coordinated oscillatory dynamics upon IR stimulation in single cells, with a stochastic number of oscillations whose mean increases with IR dose. The damped oscillations previously observed in cell populations can be explained as the aggregate behavior of single cells. link: http://identifiers.org/pubmed/16186499

MacDonald2011_GeneticMetabolicDeterminants_BuchnereAphidicola: MODEL1012300000v0.0.1

This model is from the article: Genetic and metabolic determinants of nutritional phenotype in an insect-bacterial sym…

Details

The pervasive influence of resident microorganisms on the phenotype of their hosts is exemplified by the intracellular bacterium Buchnera aphidicola, which provides its aphid partner with essential amino acids (EAAs). We investigated variation in the dietary requirement for EAAs among four pea aphid (Acyrthosiphon pisum) clones. Buchnera-derived nitrogen contributed to the synthesis of all EAAs for which aphid clones required a dietary supply, and to none of the EAAs for which all four clones had no dietary requirement, suggesting that low total dietary nitrogen may select for reduced synthesis of certain EAAs in some aphid clones. The sequenced Buchnera genomes showed that the EAA nutritional phenotype (i.e. the profile of dietary EAAs required by the aphid) cannot be attributed to sequence variation of Buchnera genes coding EAA biosynthetic enzymes. Metabolic modelling by flux balance analysis demonstrated that EAA output from Buchnera can be determined precisely by the flux of host metabolic precursors to Buchnera. Specifically, the four EAA nutritional phenotypes could be reproduced by metabolic models with unique profiles of host inputs, dominated by variation in supply of aspartate, homocysteine and glutamate. This suggests that the nutritional phenotype of the symbiosis is determined principally by host metabolism and transporter genes that regulate nutrient supply to Buchnera. Intraspecific variation in the nutritional phenotype of symbioses is expected to mediate partitioning of plant resources among aphid genotypes, potentially promoting the genetic subdivision of aphid populations. In this way, microbial symbioses may play an important role in the evolutionary diversification of phytophagous insects. link: http://identifiers.org/pubmed/21392141

MacGregor2005_HypothalamicSystems: MODEL9811206584v0.0.1

This a model from the article: Modelling the hypothalamic control of growth hormone secretion. MacGregor DJ, Leng G.…

Details

Here, we construct a mathematical model of the hypothalamic systems that control the secretion of growth hormone (GH). The work extends a recent model of the pituitary GH system, adding representations of the hypothalamic GH-releasing hormone (GHRH) and somatostatin neurones, each modelled as a single synchronised unit. An unpatterned stochastic input drives the GHRH neurones generating pulses of GHRH release that trigger GH pulses. Delayed feedback from GH results in increased somatostatin release, which inhibits both GH secretion and GHRH release, producing an overall pattern of 3-h pulses of GH secretion that is very similar to the secretory profile observed in male rats. Rather than directly stimulating somatostatin release, GH feedback triggers a priming effect, increasing releasable stores of somatostatin. Varying this priming effect to reduce the effect of GH can reproduce the less pulsatile form of GH release observed in the female rat. The model behaviour is tested by comparison with experimental observations with a range of different experimental protocols involving GHRH injections and somatostatin and GH infusion. link: http://identifiers.org/pubmed/16280026

Machado2014 - Curcumin production pathway in Escherichia coli: BIOMD0000000565v0.0.1

Machado2014 - Curcumin production pathway in Escherichia coliThis model is described in the article: [A kinetic model f…

Details

Curcumin is a natural compound obtained from turmeric, and is well known for its pharmacological effects. In this work, we design a heterologous pathway for industrial production of curcumin in Escherichia coli. A kinetic model of the pathway is then developed and connected to a kinetic model of the central carbon metabolism of E. coli. This model is used for optimization of the mutant strain through a rational design approach, and two manipulation targets are identified for overexpression. Dynamic simulations are then performed to compare the curcumin production profiles of the different mutant strains. Our results show that it is possible to obtain a significant improvement in the curcumin production rates with the proposed mutants. The kinetic model here developed can be an important framework to optimize curcumin production at an industrial scale and add value to its biomedical potential. link: http://identifiers.org/pubmed/25218090

Parameters:

NameDescription
KGAPDHgap=0.683 milli Molar; cnad = 1.47 milli Molar; KGAPDHnad=0.252 milli Molar; KGAPDHnadh=1.09 milli Molar; cnadh = 0.1 milli Molar; KGAPDHpgp=1.04E-5 milli Molar; rmaxGAPDH=921.5942861 mM per second; KGAPDHeq=0.63 dimensionlessReaction: cgap => cpgp; cgap, cpgp, Rate Law: cytosol*rmaxGAPDH*(cgap*cnad-cpgp*cnadh/KGAPDHeq)/((KGAPDHgap*(1+cpgp/KGAPDHpgp)+cgap)*(KGAPDHnad*(1+cnadh/KGAPDHnadh)+cnad))
VALDOblf=2.0 dimensionless; kALDOgapinh=0.6 milli Molar; kALDOeq=0.144 milli Molar; kALDOfdp=1.75 milli Molar; rmaxALDO=17.41464425 mM per second; kALDOgap=0.088 milli Molar; kALDOdhap=0.088 milli MolarReaction: cfdp => cdhap + cgap; cfdp, cgap, cdhap, Rate Law: cytosol*rmaxALDO*(cfdp-cgap*cdhap/kALDOeq)/(kALDOfdp+cfdp+kALDOgap*cdhap/(kALDOeq*VALDOblf)+kALDOdhap*cgap/(kALDOeq*VALDOblf)+cfdp*cgap/kALDOgapinh+cgap*cdhap/(VALDOblf*kALDOeq))
catp = 4.27 milli Molar; rmaxG1PAT=0.007525458026 mM per second; KG1PATg1p=3.2 milli Molar; KG1PATfdp=0.119 milli Molar; KG1PATatp=4.42 milli Molar; nG1PATfdp=1.2 milli MolarReaction: cg1p => ; cfdp, cg1p, cfdp, Rate Law: cytosol*rmaxG1PAT*cg1p*catp*(1+(cfdp/KG1PATfdp)^nG1PATfdp)/((KG1PATatp+catp)*(KG1PATg1p+cg1p))
rmaxR5PI=4.83841193 second inverse; KR5PIeq=4.0 dimensionlessReaction: cribu5p => crib5p; cribu5p, crib5p, Rate Law: cytosol*rmaxR5PI*(cribu5p-crib5p/KR5PIeq)
cnadph = 0.062 milli Molar; KG6PDHnadp=0.0246 milli Molar; rmaxG6PDH=1.380196955 mM per second; KG6PDHg6p=14.4 milli Molar; cnadp = 0.195 milli Molar; KG6PDHnadphg6pinh=6.43 milli Molar; KG6PDHnadphnadpinh=0.01 milli MolarReaction: cg6p => cpg; cg6p, Rate Law: cytosol*rmaxG6PDH*cg6p*cnadp/((cg6p+KG6PDHg6p)*(1+cnadph/KG6PDHnadphg6pinh)*(KG6PDHnadp*(1+cnadph/KG6PDHnadphnadpinh)+cnadp))
KG3PDHdhap=1.0 milli Molar; rmaxG3PDH=0.01162042696 mM per secondReaction: cdhap => ; cdhap, Rate Law: cytosol*rmaxG3PDH*cdhap/(KG3PDHdhap+cdhap)
Dil = 0.0 second inverseReaction: cfdp => ; cfdp, Rate Law: cytosol*Dil*cfdp
rmaxMurSynth=0.0 mM per secondReaction: cf6p =>, Rate Law: cytosol*rmaxMurSynth
rmaxTA=10.87164108 per mM per second; KTAeq=1.05 dimensionlessReaction: cgap + csed7p => cf6p + ce4p; cgap, csed7p, ce4p, cf6p, Rate Law: cytosol*rmaxTA*(cgap*csed7p-ce4p*cf6p/KTAeq)
KPFKf6ps=0.325 milli Molar; KPFKampa=19.1 milli Molar; nPFK=11.1 dimensionless; catp = 4.27 milli Molar; KPFKadpb=3.89 milli Molar; KPFKampb=3.2 milli Molar; KPFKadpc=4.14 milli Molar; KPFKatps=0.123 milli Molar; cadp = 0.595 milli Molar; rmaxPFK=1840.584747 mM per second; camp = 0.955 milli Molar; KPFKpep=3.26 milli Molar; KPFKadpa=128.0 milli Molar; LPFK=5629067.0 dimensionlessReaction: cf6p => cfdp; cpep, cf6p, cpep, Rate Law: cytosol*rmaxPFK*catp*cf6p/((catp+KPFKatps*(1+cadp/KPFKadpc))*(cf6p+KPFKf6ps*(1+cpep/KPFKpep+cadp/KPFKadpb+camp/KPFKampb)/(1+cadp/KPFKadpa+camp/KPFKampa))*(1+LPFK/(1+cf6p*(1+cadp/KPFKadpa+camp/KPFKampa)/(KPFKf6ps*(1+cpep/KPFKpep+cadp/KPFKadpb+camp/KPFKampb)))^nPFK))
KENOpep=0.135 milli Molar; KENOpg2=0.1 milli Molar; KENOeq=6.73 milli Molar; rmaxENO=330.4476151 mM per secondReaction: cpg2 => cpep; cpg2, cpep, Rate Law: cytosol*rmaxENO*(cpg2-cpep/KENOeq)/(KENOpg2*(1+cpep/KENOpep)+cpg2)
kTISeq=1.39 dimensionless; kTISdhap=2.8 milli Molar; rmaxTIS=68.67474392 mM per second; kTISgap=0.3 milli MolarReaction: cdhap => cgap; cdhap, cgap, Rate Law: cytosol*rmaxTIS*(cdhap-cgap/kTISeq)/(kTISdhap*(1+cgap/kTISgap)+cdhap)
Km_4CL=0.026; kcat_4CL=9.572; E_4CL=100.0Reaction: fer => fercoa; fer, Rate Law: E_4CL*kcat_4CL*fer/(Km_4CL+fer)
KPDHpyr=1159.0 milli Molar; nPDH=3.68 dimensionless; rmaxPDH=270.27734 mM per second; Ki_PDH_accoa=0.0222222 milli MolarReaction: cpyr => accoa; cpyr, accoa, Rate Law: cytosol*rmaxPDH*cpyr^nPDH/(KPDHpyr*(1+accoa/Ki_PDH_accoa)+cpyr^nPDH)
KPTSa2=0.01 milli Molar; nPTSg6p=3.66 dimensionless; rmaxPTS=7829.78 mM per second; KPTSa3=245.3 dimensionless; KPTSa1=3082.3 milli Molar; KPTSg6p=2.15 milli MolarReaction: cglcex + cpep => cg6p + cpyr; cglcex, cpep, cpyr, cg6p, Rate Law: extracellular*rmaxPTS*cglcex*cpep/cpyr/((KPTSa1+KPTSa2*cpep/cpyr+KPTSa3*cglcex+cglcex*cpep/cpyr)*(1+cg6p^nPTSg6p/KPTSg6p))
Keq_FER_t=1.0 dimensionless; k_FER_t=1000.0 second inverseReaction: fer_ext => fer; fer_ext, fer, Rate Law: extracellular*k_FER_t*(fer_ext-fer/Keq_FER_t)
Dil = 0.0 second inverse; cfeed_fer=500.0 milli MolarReaction: => fer_ext; fer_ext, Rate Law: extracellular*Dil*(cfeed_fer-fer_ext)
rmaxMetSynth=0.0022627 mM per secondReaction: => cpyr, Rate Law: cytosol*rmaxMetSynth
KPGKeq=1934.4 dimensionless; KPGKadp=0.185 milli Molar; catp = 4.27 milli Molar; KPGKatp=0.653 milli Molar; cadp = 0.595 milli Molar; rmaxPGK=3021.773771 mM per second; KPGKpg3=0.473 milli Molar; KPGKpgp=0.0468 milli MolarReaction: cpgp => cpg3; cpgp, cpg3, Rate Law: cytosol*rmaxPGK*(cadp*cpgp-catp*cpg3/KPGKeq)/((KPGKadp*(1+catp/KPGKatp)+cadp)*(KPGKpgp*(1+cpg3/KPGKpg3)+cpgp))
Ki_ACCOAC_malcoa=0.1 milli Molar; rmaxACCOAC=0.04634 mM per second; K_ACCOAC_accoa=3.0E-4 milli MolarReaction: accoa => malcoa; accoa, malcoa, Rate Law: cytosol*rmaxACCOAC*accoa/(K_ACCOAC_accoa*(1+malcoa/Ki_ACCOAC_malcoa)+accoa)
KRu5Peq=1.4 dimensionless; rmaxRu5P=6.739029475 second inverseReaction: cribu5p => cxyl5p; cribu5p, cxyl5p, Rate Law: cytosol*rmaxRu5P*(cribu5p-cxyl5p/KRu5Peq)
Km_DCS_malcoa=0.0084; Km_DCS_fercoa=0.046; E_DCS=100.0; n_DCS_fercoa=1.8; kcat_DCS=0.01343Reaction: fercoa + malcoa => ferdicoa; fercoa, malcoa, Rate Law: E_DCS*kcat_DCS*fercoa^n_DCS_fercoa/(Km_DCS_fercoa^n_DCS_fercoa+fercoa^n_DCS_fercoa)*malcoa/(Km_DCS_malcoa+malcoa)
npepCxylasefdp=4.21 dimensionless; rmaxpepCxylase=0.1070205858 mM per second; KpepCxylasefdp=0.7 milli Molar; KpepCxylasepep=4.07 milli MolarReaction: cpep => ; cfdp, cpep, cfdp, Rate Law: cytosol*rmaxpepCxylase*cpep*(1+(cfdp/KpepCxylasefdp)^npepCxylasefdp)/(KpepCxylasepep+cpep)
Dil = 0.0 second inverse; cfeed_glc=110.96 milli MolarReaction: => cglcex; cglcex, Rate Law: extracellular*Dil*(cfeed_glc-cglcex)
k_CUR_t=1000.0 second inverse; Keq_CUR_t=1.0 dimensionlessReaction: cur => cur_ext; cur, cur_ext, Rate Law: cytosol*k_CUR_t*(cur-cur_ext/Keq_CUR_t)
rmaxTrpSynth=0.001037 mM per secondReaction: => cpyr + cgap, Rate Law: cytosol*rmaxTrpSynth
E_CURS=100.0; kcat_CURS=0.02163; Km_CURS_ferdicoa=0.018; Km_CURS_fercoa=0.018Reaction: fercoa + ferdicoa => cur; fercoa, ferdicoa, Rate Law: E_CURS*kcat_CURS*fercoa/(Km_CURS_fercoa+fercoa)*ferdicoa/(Km_CURS_ferdicoa+ferdicoa)
KPGluMupg3=0.2 milli Molar; KPGluMueq=0.188 dimensionless; KPGluMupg2=0.369 milli Molar; rmaxPGluMu=89.04965407 mM per secondReaction: cpg3 => cpg2; cpg3, cpg2, Rate Law: cytosol*rmaxPGluMu*(cpg3-cpg2/KPGluMueq)/(KPGluMupg3*(1+cpg2/KPGluMupg2)+cpg3)
rmaxTKb=86.55855855 per mM per second; KTKbeq=10.0 dimensionlessReaction: ce4p + cxyl5p => cgap + cf6p; cxyl5p, ce4p, cf6p, cgap, Rate Law: cytosol*rmaxTKb*(cxyl5p*ce4p-cf6p*cgap/KTKbeq)
LPK=1000.0 dimensionless; KPKatp=22.5 milli Molar; catp = 4.27 milli Molar; KPKpep=0.31 milli Molar; cadp = 0.595 milli Molar; rmaxPK=0.06113150238 mM per second; camp = 0.955 milli Molar; KPKfdp=0.19 milli Molar; KPKadp=0.26 milli Molar; KPKamp=0.2 milli Molar; nPK=4.0 dimensionlessReaction: cpep => cpyr; cfdp, cpep, cfdp, Rate Law: cytosol*rmaxPK*cpep*(cpep/KPKpep+1)^(nPK-1)*cadp/(KPKpep*(LPK*((1+catp/KPKatp)/(cfdp/KPKfdp+camp/KPKamp+1))^nPK+(cpep/KPKpep+1)^nPK)*(cadp+KPKadp))
KPGIf6p=0.266 milli Molar; KPGIf6ppginh=0.2 milli Molar; KPGIg6ppginh=0.2 milli Molar; KPGIg6p=2.9 milli Molar; KPGIeq=0.1725 dimensionless; rmaxPGI=650.9878687 mM per secondReaction: cg6p => cf6p; cpg, cg6p, cf6p, cpg, Rate Law: cytosol*rmaxPGI*(cg6p-cf6p/KPGIeq)/(KPGIg6p*(1+cf6p/(KPGIf6p*(1+cpg/KPGIf6ppginh))+cpg/KPGIg6ppginh)+cg6p)
KSynth1pep=1.0 milli Molar; rmaxSynth1=0.01953897003 mM per secondReaction: cpep => ; cpep, Rate Law: cytosol*rmaxSynth1*cpep/(KSynth1pep+cpep)
KDAHPSpep=0.0053 milli Molar; KDAHPSe4p=0.035 milli Molar; nDAHPSpep=2.2 dimensionless; rmaxDAHPS=0.1079531227 mM per second; nDAHPSe4p=2.6 dimensionlessReaction: ce4p + cpep => ; ce4p, cpep, Rate Law: cytosol*rmaxDAHPS*ce4p^nDAHPSe4p*cpep^nDAHPSpep/((KDAHPSe4p+ce4p^nDAHPSe4p)*(KDAHPSpep+cpep^nDAHPSpep))
KSynth4malcoa=1.0 milli Molar; rmaxSynth4=0.092372 mM per secondReaction: malcoa => ; malcoa, Rate Law: cytosol*rmaxSynth4*malcoa/(KSynth4malcoa+malcoa)
rmaxSynth3=0.284 mM per second; KSynth3accoa=1.0 milli MolarReaction: accoa => ; accoa, Rate Law: cytosol*rmaxSynth3*accoa/(KSynth3accoa+accoa)
KSynth2pyr=1.0 milli Molar; rmaxSynth2=0.07361855055 mM per secondReaction: cpyr => ; cpyr, Rate Law: cytosol*rmaxSynth2*cpyr/(KSynth2pyr+cpyr)
rmaxTKa=9.473384783 per mM per second; KTKaeq=1.2 dimensionlessReaction: crib5p + cxyl5p => cgap + csed7p; crib5p, cxyl5p, csed7p, cgap, Rate Law: cytosol*rmaxTKa*(crib5p*cxyl5p-csed7p*cgap/KTKaeq)
rmaxSerSynth=0.025712107 mM per second; KSerSynthpg3=1.0 milli MolarReaction: cpg3 => ; cpg3, Rate Law: cytosol*rmaxSerSynth*cpg3/(KSerSynthpg3+cpg3)
cnadph = 0.062 milli Molar; KPGDHpg=37.5 milli Molar; KPGDHatpinh=208.0 milli Molar; catp = 4.27 milli Molar; rmaxPGDH=16.23235977 mM per second; KPGDHnadp=0.0506 milli Molar; KPGDHnadphinh=0.0138 milli Molar; cnadp = 0.195 milli MolarReaction: cpg => cribu5p; cpg, Rate Law: cytosol*rmaxPGDH*cpg*cnadp/((cpg+KPGDHpg)*(cnadp+KPGDHnadp*(1+cnadph/KPGDHnadphinh)*(1+catp/KPGDHatpinh)))
rmaxRPPK=0.01290045226 mM per second; KRPPKrib5p=0.1 milli MolarReaction: crib5p => ; crib5p, Rate Law: cytosol*rmaxRPPK*crib5p/(KRPPKrib5p+crib5p)
KPGMeq=0.196 dimensionless; rmaxPGM=0.8398242773 mM per second; KPGMg6p=1.038 milli Molar; KPGMg1p=0.0136 milli MolarReaction: cg6p => cg1p; cg6p, cg1p, Rate Law: cytosol*rmaxPGM*(cg6p-cg1p/KPGMeq)/(KPGMg6p*(1+cg1p/KPGMg1p)+cg6p)

States:

NameDescription
cur ext[curcumin]
crib5p[aldehydo-D-ribose 5-phosphate(2-)]
fer[ferulic acid]
cdhap[glycerone phosphate(2-)]
cpg3[3-phospho-D-glyceric acid]
cpyr[pyruvic acid]
fer ext[ferulic acid]
csed7p[sedoheptulose 7-phosphate]
ferdicoa[feruloylacetyl-CoA]
accoa[acetyl-CoA]
cxyl5p[D-xylulose 5-phosphate(2-)]
cpg2[2-phosphoglyceric acid]
cglcex[glucose]
cpg[6-phospho-D-gluconic acid]
malcoa[malonyl-CoA]
cg1p[D-glucopyranose 1-phosphate]
cpep[phosphoenolpyruvic acid]
cg6p[D-glucose 6-phosphate]
ce4p[D-erythrose 4-phosphate]
cribu5p[D-ribulose 5-phosphate(2-)]
cf6p[D-fructose 6-phosphate]
cfdp[alpha-D-fructofuranose 1,6-bisphosphate]
cpgp[683]
cur[curcumin]
cgap[glyceraldehyde 3-phosphate]
fercoa[feruloyl-CoA]

Mackenzie1996_NaGlucoseCotransporter_Kidney: MODEL1006230076v0.0.1

This a model from the article: Biophysical characteristics of the pig kidney Na+/glucose cotransporter SGLT2 reveal a…

Details

The Na+-dependent, low affinity glucose transporter SGLT2 cloned from pig kidney is 76% identical (at the amino acid level) to its high affinity homologue SGLT1. Using two-microelectrode voltage clamp, we have characterized the presteady-state and steady-state kinetics of SGLT2 expressed in Xenopus oocytes. The kinetic properties of the steady-state sugar-evoked currents as a function of external Na+ and alpha-methyl-D-glucopyranoside (alphaMG) concentrations were consistent with an ordered, simultaneous transport model in which Na+ binds first. Na+ binding was voltage-dependent and saturated with hyperpolarizing voltages. Phlorizin was a potent inhibitor of the sugar-evoked currents (KiPz approximately 10 microM) and blocked an inward Na+ current in the absence of sugar. SGLT2 exhibited Na+-dependent presteady-state currents with time constants 3-7 ms. Charge movements were described by Boltzmann relations with apparent valence approximately 1 and maximal charge transfer approximately 11 nC, and were reduced by the addition of sugar or phlorizin. The differences between SGLT1 and SGLT2 were that (i) the apparent affinity constant (K0.5) for alphaMG (approximately 3 mM) was an order of magnitude higher for SGLT2; (ii) SGLT2 excluded galactose, suggesting discrete sugar binding; (iii) K0.5 for Na+ was lower in SGLT2; and (iv) the Hill coefficient for Na+ was 1 for SGLT2 but 2 for SGLT1. Simulations of the six-state kinetic model previously proposed for SGLT1 indicated that many of the kinetic properties observed in SGLT2 are expected by simply reducing the Na+/glucose coupling from 2 to 1. link: http://identifiers.org/pubmed/8955098

MacNamara2012 - Signal transduction: MODEL1305240000v0.0.1

MacNamara2012 - Signal transductionA toy model of signal tranduction to illustrate how different logic formalizms (Boole…

Details

Despite the current wealth of high-throughput data, our understanding of signal transduction is still incomplete. Mathematical modeling can be a tool to gain an insight into such processes. Detailed biochemical modeling provides deep understanding, but does not scale well above relatively a few proteins. In contrast, logic modeling can be used where the biochemical knowledge of the system is sparse and, because it is parameter free (or, at most, uses relatively a few parameters), it scales well to large networks that can be derived by manual curation or retrieved from public databases. Here, we present an overview of logic modeling formalisms in the context of training logic models to data, and specifically the different approaches to modeling qualitative to quantitative data (state) and dynamics (time) of signal transduction. We use a toy model of signal transduction to illustrate how different logic formalisms (Boolean, fuzzy logic and differential equations) treat state and time. Different formalisms allow for different features of the data to be captured, at the cost of extra requirements in terms of computational power and data quality and quantity. Through this demonstration, the assumptions behind each formalism are discussed, as well as their advantages and disadvantages and possible future developments. link: http://identifiers.org/pubmed/22871648

Macnamara2015/1 - virotherapy full model: BIOMD0000000766v0.0.1

The paper describes a full model of oncolytic virotherapy. Created by COPASI 4.25 (Build 207) This model is descri…

Details

The main priority when designing cancer immuno-therapies has been to seek viable biological mechanisms that lead to permanent cancer eradication or cancer control. Understanding the delicate balance between the role of effector and memory cells on eliminating cancer cells remains an elusive problem in immunology. Here we make an initial investigation into this problem with the help of a mathematical model for oncolytic virotherapy; although the model can in fact be made general enough to be applied also to other immunological problems. According to this model, we find that long-term cancer control is associated with a large number of persistent effector cells (irrespective of the initial peak in effector cell numbers). However, this large number of persistent effector cells is sustained by a relatively large number of memory cells. Moreover, the results of the mathematical model suggest that cancer control from a dormant state cannot be predicted by the size of the memory population. link: http://identifiers.org/pubmed/25882747

Parameters:

NameDescription
dv = 0.0038 1/d; hu = 1.0 1Reaction: U => I; V, Rate Law: tme*dv*V*U/(hu+U)
de = 0.1 1/dReaction: E =>, Rate Law: tme*de*E
hv = 10000.0 1; pe = 0.4 1/dReaction: => E; M, U, V, Rate Law: tme*pe*M*(U+V)/(U+V+hv)
r = 0.927 1/d; K = 1.8182E8 1Reaction: => U; I, Rate Law: tme*r*U*(1-(U+I)/K)
he = 1000.0 1; du = 2.0 1/dReaction: U => ; E, Rate Law: tme*du*U*E/(he+E)
delta = 1.0 1/dReaction: I =>, Rate Law: tme*delta*I
w = 2.042 1/dReaction: V =>, Rate Law: tme*w*V
hv = 10000.0 1; pm = 2.5 1/d; m = 10000.0 1Reaction: => M; V, Rate Law: tme*pm*V*M*(1-M/m)/(hv+V)
dt = 5.0E-9 1/dReaction: E => ; U, Rate Law: tme*dt*U*E
delta = 1.0 1/d; b = 1000.0 1Reaction: => V; I, Rate Law: tme*delta*b*I

States:

NameDescription
U[malignant cell]
I[malignant cell]
M[Effector Memory Immune Cell]
V[Oncolytic Virus]
E[Effector Immune Cell]

Macnamara2015/2 - virotherapy virus-free submodel: BIOMD0000000767v0.0.1

The paper describes a submodel of oncolytic virotherapy. Created by COPASI 4.25 (Build 207) This model is describ…

Details

The main priority when designing cancer immuno-therapies has been to seek viable biological mechanisms that lead to permanent cancer eradication or cancer control. Understanding the delicate balance between the role of effector and memory cells on eliminating cancer cells remains an elusive problem in immunology. Here we make an initial investigation into this problem with the help of a mathematical model for oncolytic virotherapy; although the model can in fact be made general enough to be applied also to other immunological problems. According to this model, we find that long-term cancer control is associated with a large number of persistent effector cells (irrespective of the initial peak in effector cell numbers). However, this large number of persistent effector cells is sustained by a relatively large number of memory cells. Moreover, the results of the mathematical model suggest that cancer control from a dormant state cannot be predicted by the size of the memory population. link: http://identifiers.org/pubmed/25882747

Parameters:

NameDescription
de = 0.1 1/dReaction: E =>, Rate Law: tme*de*E
hv = 10000.0 1; pe = 0.4 1/dReaction: => E; M, U, Rate Law: tme*pe*M*U/(U+hv)
r = 0.927 1/d; K = 1.8182E8 1Reaction: => U, Rate Law: tme*r*U*(1-U/K)
he = 1000.0 1; du = 2.0 1/dReaction: U => ; E, Rate Law: tme*du*U*E/(he+E)
dt = 5.0E-9 1/dReaction: E => ; U, Rate Law: tme*dt*U*E

States:

NameDescription
U[malignant cell]
E[Effector Immune Cell]

Maeda2006_MyosinPhosphorylation: BIOMD0000000088v0.0.1

The model reproduces Fig 2B, D, F, and 2H. The dynamics correspond to a stimulus of 1 U/ml of thrombin which is equal to…

Details

Sustained contraction of cells depends on sustained Rho-associated kinase (Rho-kinase) activation. We developed a computational model of the Rho-kinase pathway to understand the systems characteristics. Thrombin-dependent in vivo transient responses of Rho activation and Ca2+ increase could be reproduced in silico. Low and high thrombin stimulation induced transient and sustained phosphorylation, respectively, of myosin light chain (MLC) and myosin phosphatase targeting subunit 1 (MYPT1) in vivo. The transient phosphorylation of MLC and MYPT1 could be reproduced in silico, but their sustained phosphorylation could not. This discrepancy between in vivo and in silico in the sustained responses downstream of Rho-kinase indicates that a missing pathway(s) may be responsible for the sustained Rho-kinase activation. We found, experimentally, that the sustained phosphorylation of MLC and MYPT1 exhibit all-or-none responses. Bromoenol lactone, a specific inhibitor of Ca2+ -independent phospholipase A2 (iPLA2), inhibited sustained phosphorylation of MLC and MYPT1, which indicates that sustained Rho-kinase activation requires iPLA2 activity. Thus, the systems analysis of the Rho-kinase pathway identified a novel iPLA2-dependent mechanism of the sustained Rho-kinase activation, which exhibits an all-or-none response. link: http://identifiers.org/pubmed/16923126

Parameters:

NameDescription
kb=0.01 per_sec; kf=10.0 per_uM_per_secReaction: s279 + s289 => s294, Rate Law: c1*(kf*s279*s289-kb*s294)
Km=5.0 microMolar; ratio=4.0 dimensionless; Vmax=15.0 per_secReaction: s2 + s130 => s174, Rate Law: c1*((1+ratio)*Vmax*s130*s2/Km-Vmax*ratio*s174)
Km=0.1 microMolar; ratio=0.028278 dimensionless; Vmax=17.505 per_secReaction: s252 + s351 => s496, Rate Law: c1*((1+ratio)*Vmax*s252*s351/Km-Vmax*ratio*s496)
Vmax=8.66 per_secReaction: s456 => s252 + s359, Rate Law: c1*Vmax*s456
Vmax=3.94 per_sec; Km=0.0014 microMolar; ratio=4.0 dimensionlessReaction: s310 + s349 => s332, Rate Law: c1*((1+ratio)*Vmax*s310*s349/Km-Vmax*ratio*s332)
kb=0.1 per_sec; kf=0.01 per_uM_per_secReaction: s351 + s349 => s355, Rate Law: c1*(kf*s351*s349-kb*s355)
ratio=0.4261 dimensionless; Vmax=8.66 per_sec; Km=2.47 microMolarReaction: s252 + s358 => s456, Rate Law: c1*((1+ratio)*Vmax*s252*s358/Km-Vmax*ratio*s456)
kb=0.1 per_sec; kf=1.0 per_secReaction: s565 => s324, Rate Law: c1*(kf*s565-kb*s324)
kb=170.0 per_sec; kf=10.0 per_uM_per_secReaction: s278 + s135 => s279, Rate Law: c1*(kf*s278*s135-kb*s279)
kf=0.5 per_secReaction: s352 => s355, Rate Law: c1*kf*s352
Vmax=3.94 per_secReaction: s332 => s310 + s350, Rate Law: c1*Vmax*s332
Vmax=10.0 per_secReaction: s568 => s152 + s153 + s569, Rate Law: c1*Vmax*s568
kb=50.0 per_sec; kf=10.0 per_uM_per_secReaction: s276 + s135 => s277, Rate Law: c1*(kf*s276*s135-kb*s277)
Vmax=3.67 per_sec; Km=10.019 microMolar; ratio=1.7299 dimensionlessReaction: s359 + s295 => s506, Rate Law: c1*((1+ratio)*Vmax*s295*s359/Km-Vmax*ratio*s506)
kf=100.0 per_uM_per_sec; kb=0.62 per_secReaction: s351 + s350 => s352, Rate Law: c1*(kf*s351*s350-kb*s352)
kb=500.0 per_sec; kf=10.0 per_uM_per_secReaction: s279 + s135 => s280, Rate Law: c1*(kf*s279*s135-kb*s280)
Vmax=1.28 per_sec; Km=4.5099 microMolar; ratio=16.617 dimensionlessReaction: s124 + s358 => s361, Rate Law: c1*((1+ratio)*Vmax*s124*s358/Km-Vmax*ratio*s361)
Km=0.04 microMolar; ratio=4.0 dimensionless; Vmax=4.9 per_secReaction: s432 + s135 => s446, Rate Law: c1*((1+ratio)*Vmax*s432*s135/Km-Vmax*ratio*s446)
kb=3.0 per_sec; kf=30.0 per_uM2_per_secReaction: s430 + s135 => s444, Rate Law: c1*(kf*s135*s135*s430-kb*s444)
kf=4.63E-5 per_secReaction: s310 => s331, Rate Law: c1*kf*s310
Vmax=9.317 per_secReaction: s470 => s359 + s351, Rate Law: c1*Vmax*s470
kf=0.004 per_uM_per_sec; kb=8.6348 per_secReaction: s566 + s153 => s565, Rate Law: c1*(kf*s566*s153-kb*s565)
kf=0.1 per_uM_per_sec; kb=0.5 per_secReaction: s277 + s289 => s292, Rate Law: c1*(kf*s277*s289-kb*s292)
Km=58.099 microMolar; ratio=28.795 dimensionless; Vmax=1.95 per_secReaction: s359 + s570 => s477, Rate Law: c1*((1+ratio)*Vmax*s359*s570/Km-Vmax*ratio*s477)
kf=1.0 per_secReaction: s444 => s172 + s430, Rate Law: c1*kf*s444
Vmax=3.67 per_sec; Km=148.08 microMolar; ratio=39.349 dimensionlessReaction: s358 + s289 => s487, Rate Law: c1*((1+ratio)*Vmax*s289*s358/Km-Vmax*ratio*s487)
kb=3.5026 per_sec; kf=1.2705 per_secReaction: s566 => s314, Rate Law: c1*(kf*s566-kb*s314)
kf=10.0 per_uM_per_sec; kb=45.0 per_secReaction: s277 + s135 => s278, Rate Law: c1*(kf*s277*s135-kb*s278)
kf=0.01 per_uM_per_secReaction: s421 + s440 => s442, Rate Law: c1*kf*s421*s440
kb=1.0 per_sec; kf=2.5201 per_uM_per_secReaction: s424 + s187 => s443, Rate Law: c1*(kf*s424*s187-kb*s443)
kf=3.0E-4 per_uM_per_sec; kb=0.1 per_secReaction: s309 + s153 => s311, Rate Law: c1*(kf*s309*s153-kb*s311)
kf=0.1 per_uM_per_sec; kb=0.45 per_secReaction: s278 + s289 => s293, Rate Law: c1*(kf*s278*s289-kb*s293)
Vmax=9.317 per_sec; Km=16.0 microMolar; ratio=7.5865 dimensionlessReaction: s359 + s351 => s480, Rate Law: c1*((1+ratio)*Vmax*s351*s359/Km-Vmax*ratio*s480)
kb=50.0 per_sec; kf=1.0 per_secReaction: s309 => s310, Rate Law: c1*(kf*s309-kb*s310)
Vmax=3.67 per_secReaction: s506 => s360 + s295, Rate Law: c1*Vmax*s506
Vmax=17.505 per_secReaction: s496 => s252 + s570, Rate Law: c1*Vmax*s496
Vmax=1.28 per_secReaction: s361 => s359 + s124, Rate Law: c1*Vmax*s361
kf=0.0133 per_secReaction: s564 => s569 + s440, Rate Law: c1*kf*s564
Km=19.841 microMolar; ratio=4.0 dimensionless; Vmax=10.0 per_secReaction: s569 + s151 => s568, Rate Law: c1*((1+ratio)*Vmax*s569*s151/Km-ratio*Vmax*s568)

States:

NameDescription
s278[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2; Calmodulin; Calcium cation]
s442[Guanine nucleotide-binding protein G(T) subunit gamma-T1; Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Guanine nucleotide-binding protein subunit alpha-11]
s310[Protein kinase C alpha type]
s329csa39_degraded
s330csa36_degraded
s289[Myosin light chain kinase 2, skeletal/cardiac muscle]
s294[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin; Calcium cation]
s338[Protein phosphatase 1 regulatory subunit 14A; Protein kinase C alpha type]
s359[Myosin light chain phosphate; Myosin light chain 1/3, skeletal muscle isoform]
s446[calcium(2+); Calcium cation]
s456[GTP; Transforming protein RhoA; Rho-associated protein kinase 1; Myosin light chain 1/3, skeletal muscle isoform; GTP]
s568[1-phosphatidyl-1D-myo-inositol 4,5-bisphosphate; calcium(2+); 1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase beta-1; 1-Phosphatidyl-D-myo-inositol 4,5-bisphosphate; Calcium cation]
s292[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin; Calcium cation]
s546[calcium(2+); Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain 1/3, skeletal muscle isoform; Calmodulin; Calcium cation]
s311[diglyceride; Protein kinase C alpha type; Diacylglycerol]
s2[Prothrombin]
s565[diglyceride; calcium(2+); Protein kinase C alpha type; Diacylglycerol; Calcium cation]
s314[Protein kinase C alpha type]
s277[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2; Calmodulin; Calcium cation]
s360[Myosin light chain; Myosin light chain 1/3, skeletal muscle isoform]
s324[Protein kinase C alpha type]
s309[Protein kinase C alpha type]
s358[Myosin light chain phosphate; Myosin light chain 1/3, skeletal muscle isoform]
s280[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2; Calmodulin; Calcium cation]
s512[calcium(2+); Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain 1/3, skeletal muscle isoform; Calmodulin; Calcium cation]
s331csa35_degraded
s174[Proteinase-activated receptor 1; IPR000935]
s135[calcium(2+); Calcium cation]
s295[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin; Calcium cation]
s506[calcium(2+); Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain 1/3, skeletal muscle isoform; Calmodulin; Calcium cation]
s252[GTP; Rho-associated protein kinase 1; Transforming protein RhoA; GTP]
s50[GDP; GDP]
s566[calcium(2+); Protein kinase C alpha type; Calcium cation]
s187[GTP; Guanine nucleotide-binding protein subunit alpha-11; GTP]
s513[calcium(2+); Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain 1/3, skeletal muscle isoform; Calmodulin; Calcium cation]
s352[Protein phosphatase 1 regulatory subunit 12A; Protein phosphatase 1 regulatory subunit 14A]
s293[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin; Calcium cation]
s496[Rho-associated protein kinase 1; Protein phosphatase 1 regulatory subunit 12A]
s332[Protein phosphatase 1 regulatory subunit 14A; Protein kinase C alpha type]
s520[calcium(2+); Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain 1/3, skeletal muscle isoform; Calmodulin; Calcium cation]
s564[GTP; calcium(2+); 1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase beta-1; Guanine nucleotide-binding protein subunit alpha-11; GTP; Calcium cation]
s279[calcium(2+); Calmodulin-3Calmodulin-1Calmodulin-2; Calmodulin; Calcium cation]
s444[calcium(2+); Calcium cation]
s335[Protein phosphatase 1 regulatory subunit 14A; Protein kinase C alpha type]
s349[Protein phosphatase 1 regulatory subunit 14A]
s539[calcium(2+); Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain 1/3, skeletal muscle isoform; Calmodulin; Calcium cation]
s526[calcium(2+); Myosin light chain kinase 2, skeletal/cardiac muscle; Calmodulin-3Calmodulin-1Calmodulin-2; Myosin light chain 1/3, skeletal muscle isoform; Calmodulin; Calcium cation]

Maeda2019_AmmoniumTransportAssimilation: MODEL1901090001v0.0.1

The E. coli ammonium transport and assimilation network. This SBML model simulates Kim's experiment with glucose as a ca…

Details

The complex ammonium transport and assimilation network of E. coli involves the ammonium transporter AmtB, the regulatory proteins GlnK and GlnB, and the central N-assimilating enzymes together with their highly complex interactions. The engineering and modelling of such a complex network seem impossible because functioning depends critically on a gamut of data known at patchy accuracy. We developed a way out of this predicament, which employs: (i) a constrained optimization-based technology for the simultaneous fitting of models to heterogeneous experimental data sets gathered through diverse experimental set-ups, (ii) a ‘rubber band method’ to deal with different degrees of uncertainty, both in experimentally determined or estimated parameter values and in measured transient or steady-state variables (training data sets), (iii) integration of human expertise to decide on accuracies of both parameters and variables, (iv) massive computation employing a fast algorithm and a supercomputer, (v) an objective way of quantifying the plausibility of models, which makes it possible to decide which model is the best and how much better that model is than the others. We applied the new technology to the ammonium transport and assimilation network, integrating recent and older data of various accuracies, from different expert laboratories. The kinetic model objectively ranked best, has E. coli's AmtB as an active transporter of ammonia to be assimilated with GlnK minimizing the futile cycling that is an inevitable consequence of intracellular ammonium accumulation. It is 130 times better than a model with facilitated passive transport of ammonia. link: http://identifiers.org/doi/10.1038/s41540-019-0091-6

Mager2005 - Quasi-equilibrium pharmacokinetic model for drugs exhibiting target-mediated drug disposition: BIOMD0000000765v0.0.1

This model was developed with the aim of constructing an equilibrium model of the pharmacokinetic behaviour of a drug ex…

Details

The aim of this study is to derive and evaluate an equilibrium model of a previously developed general pharmacokinetic model for drugs exhibiting target-mediated drug disposition (TMDD).A quasi-equilibrium solution to the system of ordinary differential equations that describe the kinetics of TMDD was obtained. Computer simulations of the equilibrium model were carried out to generate plasma concentration-time profiles resulting from a large range of intravenous bolus doses. Additionally, the final model was fitted to previously published pharmacokinetic profiles of leukemia inhibitory factor (LIF), a cytokine that seems to exhibit TMDD, following intravenous administration of 12.5, 25, 100, 250, 500, or 750 microg/kg in sheep.Simulations show that pharmacokinetic profiles display steeper distribution phases for lower doses and similar terminal disposition phases, but with slight underestimation at early time points as theoretically expected. The final model well-described LIF pharmacokinetics, and the final parameters, which were estimated with relatively good precision, were in good agreement with literature values.An equilibrium model of TMDD is developed that recapitulates the essential features of the full general model and eliminates the need for estimating drug-binding microconstants that are often difficult or impossible to identify from typical in vivo pharmacokinetic data. link: http://identifiers.org/pubmed/16180117

Parameters:

NameDescription
k_on = 0.1Reaction: R + C => RC, Rate Law: Central*k_on*R*C
k_tp = 0.0Reaction: A_T =>, Rate Law: Peripheral_Tissue*k_tp*A_T
k_off = 0.1Reaction: => R + C; RC, Rate Law: Central*k_off*RC
k_pt = 0.0; V_c = 10.0Reaction: => A_T; C, Rate Law: k_pt*C*V_c
k_off = 0.1; k_int = 0.0Reaction: RC =>, Rate Law: Central*(k_off+k_int)*RC
k_tp = 0.0; V_c = 10.0Reaction: => C; A_T, Rate Law: k_tp*A_T/V_c
k_syn = 0.0Reaction: => R, Rate Law: Central*k_syn
k_pt = 0.0; k_el = 1.0Reaction: C =>, Rate Law: Central*(k_el+k_pt)*C
k_deg = 0.566Reaction: R =>, Rate Law: Central*k_deg*R

States:

NameDescription
C[drug; 7-Methylxanthine]
A T[drug; Tissue]
R[Receptor]
RC[receptor complex]

Magnus1997_MitoCa_BetaCellMinimalModel: MODEL1201140004v0.0.1

This a model from the article: Minimal model of beta-cell mitochondrial Ca2+ handling. Magnus G, Keizer J. Am J Phys…

Details

We develop a simplified, but useful, mathematical model to describe Ca2+ handling by mitochondria in the pancreatic beta-cell. The model includes the following six transport mechanisms in the inner mitochondrial membrane: proton pumping via respiration and proton uptake by way of the F1Fzero-ATPase (adapted from D. Pietrobon and S. Caplan. Biochemistry 24: 5764-5778, 1985), a proton leak, adenine nucleotide exchange, the Ca2+ uniporter, and Na+/Ca2+ exchange. Each mechanism is developed separately into a kinetic model for the rate of transport, with parameters taken from experiments on isolated mitochondrial preparations. These mechanisms are combined in a modular fashion first to describe state 4 (nonphosphorylating) and state 3 (phosphorylating) mitochondria with mitochondrial NADH and Ca2+ concentrations as fixed parameters and then to describe Ca2+ handling with variable mitochondrial Ca2+ concentration. Simulations are compared to experimental measurements and agree well with the threshold for Ca2+ uptake, measured mitochondrial Ca2+ levels, and the influence of Ca2+ on oxygen uptake. In the absence of Ca2+ activation of mitochondrial dehydrogenases, the simulations predict a significant reduction in the rate of production of ATP that involves a "short circuit" via Ca2+ uptake through the uniporter. This effect suggests a potential role for mitochondrial Ca2+ handling in determining the ATP-ADP ratio in the pancreatic beta-cell. link: http://identifiers.org/pubmed/9277370

Mahadevan2006 - Genome-scale metabolic network of Geobacter sulfurreducens (iRM588): MODEL1507180000v0.0.1

Mahadevan2006 - Genome-scale metabolic network of Geobacter sulfurreducens (iRM588)This model is described in the articl…

Details

Geobacter sulfurreducens is a well-studied representative of the Geobacteraceae, which play a critical role in organic matter oxidation coupled to Fe(III) reduction, bioremediation of groundwater contaminated with organics or metals, and electricity production from waste organic matter. In order to investigate G. sulfurreducens central metabolism and electron transport, a metabolic model which integrated genome-based predictions with available genetic and physiological data was developed via the constraint-based modeling approach. Evaluation of the rates of proton production and consumption in the extracellular and cytoplasmic compartments revealed that energy conservation with extracellular electron acceptors, such as Fe(III), was limited relative to that associated with intracellular acceptors. This limitation was attributed to lack of cytoplasmic proton consumption during reduction of extracellular electron acceptors. Model-based analysis of the metabolic cost of producing an extracellular electron shuttle to promote electron transfer to insoluble Fe(III) oxides demonstrated why Geobacter species, which do not produce shuttles, have an energetic advantage over shuttle-producing Fe(III) reducers in subsurface environments. In silico analysis also revealed that the metabolic network of G. sulfurreducens could synthesize amino acids more efficiently than that of Escherichia coli due to the presence of a pyruvate-ferredoxin oxidoreductase, which catalyzes synthesis of pyruvate from acetate and carbon dioxide in a single step. In silico phenotypic analysis of deletion mutants demonstrated the capability of the model to explore the flexibility of G. sulfurreducens central metabolism and correctly predict mutant phenotypes. These results demonstrate that iterative modeling coupled with experimentation can accelerate the understanding of the physiology of poorly studied but environmentally relevant organisms and may help optimize their practical applications. link: http://identifiers.org/pubmed/16461711

Mahajan2008_CardiacActionPotential_Arrhythmias: MODEL1006230048v0.0.1

This a model from the article: A rabbit ventricular action potential model replicating cardiac dynamics at rapid heart…

Details

Mathematical modeling of the cardiac action potential has proven to be a powerful tool for illuminating various aspects of cardiac function, including cardiac arrhythmias. However, no currently available detailed action potential model accurately reproduces the dynamics of the cardiac action potential and intracellular calcium (Ca(i)) cycling at rapid heart rates relevant to ventricular tachycardia and fibrillation. The aim of this study was to develop such a model. Using an existing rabbit ventricular action potential model, we modified the L-type calcium (Ca) current (I(Ca,L)) and Ca(i) cycling formulations based on new experimental patch-clamp data obtained in isolated rabbit ventricular myocytes, using the perforated patch configuration at 35-37 degrees C. Incorporating a minimal seven-state Markovian model of I(Ca,L) that reproduced Ca- and voltage-dependent kinetics in combination with our previously published dynamic Ca(i) cycling model, the new model replicates experimentally observed action potential duration and Ca(i) transient alternans at rapid heart rates, and accurately reproduces experimental action potential duration restitution curves obtained by either dynamic or S1S2 pacing. link: http://identifiers.org/pubmed/18160660

mahaney2000_SERCAregulation: MODEL4816599063v0.0.1

This model originates from BioModels Database: A Database of Annotated Published Models. It is copyright (c) 2005-2011 T…

Details

Kinetics studies of the cardiac Ca-ATPase expressed in Sf21 cells (Spodoptera frugiperda insect cells) have been carried out to test the hypotheses that phospholamban inhibits Ca-ATPase cycling by decreasing the rate of the E1.Ca to E1'.Ca transition and/or the rate of phosphoenzyme hydrolysis. Three sample types were studied: Ca-ATPase expressed alone, Ca-ATPase coexpressed with wild-type phospholamban (the natural pentameric inhibitor), and Ca-ATPase coexpressed with the L37A-phospholamban mutant (a more potent monomeric inhibitor, in which Leu(37) is replaced by Ala). Phospholamban coupling to the Ca-ATPase was controlled using a monoclonal antibody against phospholamban. Gel electrophoresis and immunoblotting confirmed an equivalent ratio of Ca-ATPase and phospholamban in each sample (1 mol Ca-ATPase to 1.5 mol phospholamban). Steady-state ATPase activity assays at 37 degrees C, using 5 mM MgATP, showed that the phospholamban-containing samples had nearly equivalent maximum activity ( approximately 0.75 micromol. nmol Ca-ATPase(-1).min(-1) at 15 microM Ca(2+)), but that wild-type phospholamban and L37A-phospholamban increased the Ca-ATPase K(Ca) values by 200 nM and 400 nM, respectively. When steady-state Ca-ATPase phosphoenzyme levels were measured at 0 degrees C, using 1 microM MgATP, the K(Ca) values also shifted by 200 nM and 400 nM, respectively, similar to the results obtained by measuring ATP hydrolysis at 37 degrees C. Measurements of the time course of phosphoenzyme formation at 0 degrees C, using 1 microM MgATP and 268 nM ionized [Ca(2+)], indicated that L37A-phospholamban decreased the steady-state phosphoenzyme level to a greater extent (45%) than did wild-type phospholamban (33%), but neither wild-type nor L37A-phospholamban had any effect on the apparent rate of phosphoenzyme formation relative to that of Ca-ATPase expressed alone. Measurements of inorganic phosphate (P(i)) release concomitant with the phosphoenzyme formation studies showed that L37A-phospholamban decreased the steady-state rate of P(i) release to a greater extent (45%) than did wild-type phospholamban (33%). However, independent measurements of Ca-ATPase dephosphorylation after the addition of 5 mM EGTA to the phosphorylated enzyme showed that neither wild-type phospholamban nor L37A-phospholamban had any effect on the rate of phosphoenzyme decay relative to Ca-ATPase expressed alone. Computer simulation of the kinetics data indicated that phospholamban and L37A-phospholamban decreased twofold and fourfold, respectively, the equilibrium binding of the first Ca(2+) ion to the Ca-ATPase E1 intermediate, rather than inhibiting rate of the E.Ca to E'.Ca transition or the rate of phosphoenzyme decay. Therefore, we conclude that phospholamban inhibits Ca-ATPase cycling by decreasing Ca-ATPase Ca(2+) binding to the E1 intermediate. link: http://identifiers.org/pubmed/10692318

Mahasa2016-tumor–immune surveillance: MODEL2003040001v0.0.1

We present a novel mathematical model involving various immune cell populations and tumor cellpopulations. The model des…

Details

We present a novel mathematical model involving various immune cell populations and tumor cell populations. The model describes how tumor cells evolve and survive the brief encounter with the immune system mediated by natural killer (NK) cells and the activated CD8(+) cytotoxic T lymphocytes (CTLs). The model is composed of ordinary differential equations describing the interactions between these important immune lymphocytes and various tumor cell populations. Based on up-to-date knowledge of immune evasion and rational considerations, the model is designed to illustrate how tumors evade both arms of host immunity (i.e. innate and adaptive immunity). The model predicts that (a) an influx of an external source of NK cells might play a crucial role in enhancing NK-cell immune surveillance; (b) the host immune system alone is not fully effective against progression of tumor cells; (c) the development of immunoresistance by tumor cells is inevitable in tumor immune surveillance. Our model also supports the importance of infiltrating NK cells in tumor immune surveillance, which can be enhanced by NK cell-based immunotherapeutic approaches. link: http://identifiers.org/pubmed/27317864

Makin2013 - Blood coagulation cascade model: MODEL1807230001v0.0.1

Mathematical model of the blood coagulation system including lipid binding sites, meizothrombin, thrombomodulin, cleaved…

Details

The process of human blood clotting involves a complex interaction of continuous-time/continuous-state processes and discrete-event/discrete-state phenomena, where the former comprise the various chemical rate equations and the latter comprise both threshold-limited behaviors and binary states (presence/absence of a chemical). Whereas previous blood-clotting models used only continuous dynamics and perforce addressed only portions of the coagulation cascade, we capture both continuous and discrete aspects by modeling it as a hybrid dynamical system. The model was implemented as a hybrid Petri net, a graphical modeling language that extends ordinary Petri nets to cover continuous quantities and continuous-time flows. The primary focus is simulation: (1) fidelity to the clinical data in terms of clotting-factor concentrations and elapsed time; (2) reproduction of known clotting pathologies; and (3) fine-grained predictions which may be used to refine clinical understanding of blood clotting. Next we examine sensitivity to rate-constant perturbation. Finally, we propose a method for titrating between reliance on the model and on prior clinical knowledge. For simplicity, we confine these last two analyses to a critical purely-continuous subsystem of the model. link: http://identifiers.org/pubmed/24131053

Maleckar2008_AtrialMyocyte: MODEL9810152478v0.0.1

This a model from the article: Mathematical simulations of ligand-gated and cell-type specific effects on the action p…

Details

In the mammalian heart, myocytes and fibroblasts can communicate via gap junction, or connexin-mediated current flow. Some of the effects of this electrotonic coupling on the action potential waveform of the human ventricular myocyte have been analyzed in detail. The present study employs a recently developed mathematical model of the human atrial myocyte to investigate the consequences of this heterogeneous cell-cell interaction on the action potential of the human atrium. Two independent physiological processes which alter the physiology of the human atrium have been studied. i) The effects of the autonomic transmitter acetylcholine on the atrial action potential have been investigated by inclusion of a time-independent, acetylcholine-activated K(+) current in this mathematical model of the atrial myocyte. ii) A non-selective cation current which is activated by natriuretic peptides has been incorporated into a previously published mathematical model of the cardiac fibroblast. These results identify subtle effects of acetylcholine, which arise from the nonlinear interactions between ionic currents in the human atrial myocyte. They also illustrate marked alterations in the action potential waveform arising from fibroblast-myocyte source-sink principles when the natriuretic peptide-mediated cation conductance is activated. Additional calculations also illustrate the effects of simultaneous activation of both of these cell-type specific conductances within the atrial myocardium. This study provides a basis for beginning to assess the utility of mathematical modeling in understanding detailed cell-cell interactions within the complex paracrine environment of the human atrial myocardium. link: http://identifiers.org/pubmed/19186188

Malinzi2018 - Enhancement of chemotherapy using oncolytic virotherapy: MODEL2003050002v0.0.1

&lt;notes xmlns=&quot;http://www.sbml.org/sbml/level2/version4&quot;&gt; &lt;body xmlns=&quot;http://www.w3.org/1…

Details

Oncolytic virotherapy has been emerging as a promising novel cancer treatment which may be further combined with the existing therapeutic modalities to enhance their effects. To investigate how virotherapy could enhance chemotherapy, we propose an ODE based mathematical model describing the interactions between tumour cells, the immune response, and a treatment combination with chemotherapy and oncolytic viruses. Stability analysis of the model with constant chemotherapy treatment rates shows that without any form of treatment, a tumour would grow to its maximum size. It also demonstrates that chemotherapy alone is capable of clearing tumour cells provided that the drug efficacy is greater than the intrinsic tumour growth rate. Furthermore, virotherapy alone may not be able to clear tumour cells from body tissue but would rather enhance chemotherapy if viruses with high viral potency are used. To assess the combined effect of virotherapy and chemotherapy we use the forward sensitivity index to perform a sensitivity analysis, with respect to chemotherapy key parameters, of the virus basic reproductive number and the tumour endemic equilibrium. The results from this sensitivity analysis indicate the existence of a critical dose of chemotherapy above which no further significant reduction in the tumour population can be observed. Numerical simulations show that a successful combinational therapy of the chemotherapeutic drugs and viruses depends mostly on the virus burst size, infection rate, and the amount of drugs supplied. Optimal control analysis was performed, by means of the Pontryagin's maximum principle, to further refine predictions of the model with constant treatment rates by accounting for the treatment costs and sides effects. Results from this analysis suggest that the optimal drug and virus combination correspond to half their maximum tolerated doses. This is in agreement with the results from stability and sensitivity analyses. link: http://identifiers.org/pubmed/30418793

Malinzi2018 - tumour-immune interaction model: BIOMD0000000809v0.0.1

The paper describes a spatio-temporal mathematical model, in the form of a moving boundary problem, to explain cancer do…

Details

A spatio-temporal mathematical model, in the form of a moving boundary problem, to explain cancer dormancy is developed. Analysis of the model is carried out for both temporal and spatio-temporal cases. Stability analysis and numerical simulations of the temporal model replicate experimental observations of immune-induced tumour dormancy. Travelling wave solutions of the spatio-temporal model are determined using the hyperbolic tangent method and minimum wave speeds of invasion are calculated. Travelling wave analysis depicts that cell invasion dynamics are mainly driven by their motion and growth rates. A stability analysis of the spatio-temporal model shows a possibility of dynamical stabilization of the tumour-free steady state. Simulation results reveal that the tumour swells to a dormant level. link: http://identifiers.org/pubmed/30537482

Parameters:

NameDescription
myu_1 = 1.0Reaction: u =>, Rate Law: compartment*myu_1*u
sigma_2 = 0.5; sigma_1 = 0.3Reaction: => y, Rate Law: compartment*sigma_1*y*(1-sigma_2*y)
phi_2 = 0.25; phi_1 = 1.3398Reaction: => x, Rate Law: compartment*phi_1*x*(1-phi_2*x)
myu_2 = 0.24Reaction: ystar =>, Rate Law: compartment*myu_2*ystar
rho = 0.1Reaction: => ystar; x, y, Rate Law: compartment*rho*x*y
nu_1 = 0.00218Reaction: x => ; y, Rate Law: compartment*nu_1*x*y
delta = 3.0218; gamma = 2.02Reaction: => x; y, Rate Law: compartment*delta*x*y/(gamma+x)
nu_2 = 0.7279Reaction: y => ; x, Rate Law: compartment*nu_2*x*y
nu_3 = 300.0Reaction: => u; x, y, Rate Law: compartment*nu_3*x*y

States:

NameDescription
ystar[cancer; Dead]
x[Immune Cell]
u[Chemokine; Concentration]
y[cancer]

Malinzi2019 - chemovirotherapy: BIOMD0000000764v0.0.1

The paper describes a model of oncolytic virothherapy. Created by COPASI 4.25 (Build 207) This model is described…

Details

A mathematical model for the treatment of cancer using chemovirotherapy is developed with the aim of determining the efficacy of three drug infusion methods: constant, single bolus, and periodic treatments. The model is in the form of ODEs and is further extended into DDEs to account for delays as a result of the infection of tumor cells by the virus and chemotherapeutic drug responses. Analysis of the model is carried out for each of the three drug infusion methods. Analytic solutions are determined where possible and stability analysis of both steady state solutions for the ODEs and DDEs is presented. The results indicate that constant and periodic drug infusion methods are more efficient compared to a single bolus injection. Numerical simulations show that with a large virus burst size, irrespective of the drug infusion method, chemovirotherapy is highly effective compared to either treatments. The simulations further show that both delays increase the period within which a tumor can be cleared from body tissue. link: http://identifiers.org/pubmed/30984283

Parameters:

NameDescription
bet = 1955.03421309873 1Reaction: U + V => I, Rate Law: tme*bet*U*V
k1=1.0Reaction: I =>, Rate Law: tme*k1*I
alph = 0.402737047898338 1Reaction: => U; I, Rate Law: tme*alph*U*((1-U)-I)
y = 0.00195503421309873 1Reaction: V =>, Rate Law: tme*y*V
k = 97.7517106549365 1Reaction: => C, Rate Law: tme*k
p = 8.13294232649072 1Reaction: C =>, Rate Law: tme*p*C
d0 = 9.77517106549365E-4 1Reaction: U => ; C, Rate Law: tme*d0*C*U
d1 = 0.00117302052785924 1Reaction: I => ; C, Rate Law: tme*d1*C*I
b = 2.0 1Reaction: => V; I, Rate Law: tme*b*I

States:

NameDescription
I[malignant cell]
U[malignant cell]
CC
V[Oncolytic Virus]

Malkov2020 - SEIRS model of COVID-19 transmission with reinfection: BIOMD0000000979v0.0.1

Epidemiological models of COVID-19 transmission assume that recovered individuals have a fully protected immunity. To da…

Details

Epidemiological models of COVID-19 transmission assume that recovered individuals have a fully protected immunity. To date, there is no definite answer about whether people who recover from COVID-19 can be reinfected with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In the absence of a clear answer about the risk of reinfection, it is instructive to consider the possible scenarios. To study the epidemiological dynamics with the possibility of reinfection, I use a Susceptible-Exposed-Infectious-Resistant-Susceptible model with the time-varying transmission rate. I consider three different ways of modeling reinfection. The crucial feature of this study is that I explore both the difference between the reinfection and no-reinfection scenarios and how the mitigation measures affect this difference. The principal results are the following. First, the dynamics of the reinfection and no-reinfection scenarios are indistinguishable before the infection peak. Second, the mitigation measures delay not only the infection peak, but also the moment when the difference between the reinfection and no-reinfection scenarios becomes prominent. These results are robust to various modeling assumptions. link: http://identifiers.org/pubmed/32982082

Malkov2020 - SEIRS model of COVID-19 transmission with time-varying R values and reinfection: BIOMD0000000980v0.0.1

Epidemiological models of COVID-19 transmission assume that recovered individuals have a fully pro- tected immunity. To…

Details

Epidemiological models of COVID-19 transmission assume that recovered individuals have a fully protected immunity. To date, there is no definite answer about whether people who recover from COVID-19 can be reinfected with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In the absence of a clear answer about the risk of reinfection, it is instructive to consider the possible scenarios. To study the epidemiological dynamics with the possibility of reinfection, I use a Susceptible-Exposed-Infectious-Resistant-Susceptible model with the time-varying transmission rate. I consider three different ways of modeling reinfection. The crucial feature of this study is that I explore both the difference between the reinfection and no-reinfection scenarios and how the mitigation measures affect this difference. The principal results are the following. First, the dynamics of the reinfection and no-reinfection scenarios are indistinguishable before the infection peak. Second, the mitigation measures delay not only the infection peak, but also the moment when the difference between the reinfection and no-reinfection scenarios becomes prominent. These results are robust to various modeling assumptions. link: http://identifiers.org/pubmed/32982082

Maltsev2009_PacemakerCellModel_nonSteadyState: MODEL1006230069v0.0.1

This a model from the article: Synergism of coupled subsarcolemmal Ca2+ clocks and sarcolemmal voltage clocks confers…

Details

Recent experimental studies have demonstrated that sinoatrial node cells (SANC) generate spontaneous, rhythmic, local subsarcolemmal Ca(2+) releases (Ca(2+) clock), which occur during late diastolic depolarization (DD) and interact with the classic sarcolemmal voltage oscillator (membrane clock) by activating Na(+)-Ca(2+) exchanger current (I(NCX)). This and other interactions between clocks, however, are not captured by existing essentially membrane-delimited cardiac pacemaker cell numerical models. Using wide-scale parametric analysis of classic formulations of membrane clock and Ca(2+) cycling, we have constructed and initially explored a prototype rabbit SANC model featuring both clocks. Our coupled oscillator system exhibits greater robustness and flexibility than membrane clock operating alone. Rhythmic spontaneous Ca(2+) releases of sarcoplasmic reticulum (SR)-based Ca(2+) clock ignite rhythmic action potentials via late DD I(NCX) over much broader ranges of membrane clock parameters [e.g., L-type Ca(2+) current (I(CaL)) and/or hyperpolarization-activated ("funny") current (I(f)) conductances]. The system Ca(2+) clock includes SR and sarcolemmal Ca(2+) fluxes, which optimize cell Ca(2+) balance to increase amplitudes of both SR Ca(2+) release and late DD I(NCX) as SR Ca(2+) pumping rate increases, resulting in a broad pacemaker rate modulation (1.8-4.6 Hz). In contrast, the rate modulation range via membrane clock parameters is substantially smaller when Ca(2+) clock is unchanged or lacking. When Ca(2+) clock is disabled, the system parametric space for fail-safe SANC operation considerably shrinks: without rhythmic late DD I(NCX) ignition signals membrane clock substantially slows, becomes dysrhythmic, or halts. In conclusion, the Ca(2+) clock is a new critical dimension in SANC function. A synergism of the coupled function of Ca(2+) and membrane clocks confers fail-safe SANC operation at greatly varying rates. link: http://identifiers.org/pubmed/19136600

Maltsev2009_PacemakerCellModel_SteadyState: MODEL1006230104v0.0.1

This a model from the article: Synergism of coupled subsarcolemmal Ca2+ clocks and sarcolemmal voltage clocks confers…

Details

Recent experimental studies have demonstrated that sinoatrial node cells (SANC) generate spontaneous, rhythmic, local subsarcolemmal Ca(2+) releases (Ca(2+) clock), which occur during late diastolic depolarization (DD) and interact with the classic sarcolemmal voltage oscillator (membrane clock) by activating Na(+)-Ca(2+) exchanger current (I(NCX)). This and other interactions between clocks, however, are not captured by existing essentially membrane-delimited cardiac pacemaker cell numerical models. Using wide-scale parametric analysis of classic formulations of membrane clock and Ca(2+) cycling, we have constructed and initially explored a prototype rabbit SANC model featuring both clocks. Our coupled oscillator system exhibits greater robustness and flexibility than membrane clock operating alone. Rhythmic spontaneous Ca(2+) releases of sarcoplasmic reticulum (SR)-based Ca(2+) clock ignite rhythmic action potentials via late DD I(NCX) over much broader ranges of membrane clock parameters [e.g., L-type Ca(2+) current (I(CaL)) and/or hyperpolarization-activated ("funny") current (I(f)) conductances]. The system Ca(2+) clock includes SR and sarcolemmal Ca(2+) fluxes, which optimize cell Ca(2+) balance to increase amplitudes of both SR Ca(2+) release and late DD I(NCX) as SR Ca(2+) pumping rate increases, resulting in a broad pacemaker rate modulation (1.8-4.6 Hz). In contrast, the rate modulation range via membrane clock parameters is substantially smaller when Ca(2+) clock is unchanged or lacking. When Ca(2+) clock is disabled, the system parametric space for fail-safe SANC operation considerably shrinks: without rhythmic late DD I(NCX) ignition signals membrane clock substantially slows, becomes dysrhythmic, or halts. In conclusion, the Ca(2+) clock is a new critical dimension in SANC function. A synergism of the coupled function of Ca(2+) and membrane clocks confers fail-safe SANC operation at greatly varying rates. link: http://identifiers.org/pubmed/19136600

Manda2019 - Acute hepatitis B virus infection model within the host incorporating immune cells and cytokine response: MODEL1911280002v0.0.1

This is a within-host hepatitis B viral mathematical model for hepatitis B in the acute phase of infection. The model in…

Details

We formulate and analyze a within-host hepatitis B viral mathematical model for hepatitis B in the acute phase of infection. The model incorporates hepatocytes, hepatitis B virus, immune system cells and cytokine dynamics using a system of ordinary differential equations. We use the model to demonstrate the trends of the hepatitis B infection qualitatively without the effects of immune cells and cytokines. Using these trends, we tested the effects of incorporating the immune cells only and immune cells with cytokine responses at low and high inhibitions on the hepatitis B virus infection. Our results showed that it is impossible to have the immune cells work independently from cytokines when there is an acute hepatitis B virus infection. Therefore, our results suggest that incorporating immune cells and cytokine dynamics in the acute hepatitis B virus infection stage delays infection in the hepatocytes and excluding such dynamics speeds up infection during this phase. Results from this study are useful in developing strategies for control of hepatocellular carcinoma which is caused by hepatitis B virus infection. link: http://identifiers.org/doi/10.1007/s12064-019-00305-2

Mandlik2013 - Synthetic circuit of IPC synthase in Leishmania: MODEL1208030000v0.0.1

Mandlik2012 - Synthetic circuit of IPC synthase in LeishmaniaA genetic circuit for the targeted enzyme inositol phosphor…

Details

Building circuits and studying their behavior in cells is a major goal of systems and synthetic biology. Synthetic biology enables the precise control of cellular states for systems studies, the discovery of novel parts, control strategies, and interactions for the design of robust synthetic systems. To the best of our knowledge, there are no literature reports for the synthetic circuit construction for protozoan parasites. This paper describes the construction of genetic circuit for the targeted enzyme inositol phosphorylceramide synthase belonging to the protozoan parasite Leishmania. To explore the dynamic nature of the circuit designed, simulation was done followed by circuit validation by qualitative and quantitative approaches. The genetic circuit designed for inositol phosphorylceramide synthase (Biomodels Database-MODEL1208030000) shows responsiveness, oscillatory and bistable behavior, together with intrinsic robustness. link: http://identifiers.org/pubmed/24386012

Mardinoglu2013 - Genome-scale metabolic model (HMR version 1.0) - human adipocytes (iAdipocytes1809): MODEL1402200001v0.0.1

This SBML representation of the Homo sapiens adipocyte metabolic network is made available under the Creative Commons At…

Details

We evaluated the presence/absence of proteins encoded by 14 077 genes in adipocytes obtained from different tissue samples using immunohistochemistry. By combining this with previously published adipocyte-specific proteome data, we identified proteins associated with 7340 genes in human adipocytes. This information was used to reconstruct a comprehensive and functional genome-scale metabolic model of adipocyte metabolism. The resulting metabolic model, iAdipocytes1809, enables mechanistic insights into adipocyte metabolism on a genome-wide level, and can serve as a scaffold for integration of omics data to understand the genotype-phenotype relationship in obese subjects. By integrating human transcriptome and fluxome data, we found an increase in the metabolic activity around androsterone, ganglioside GM2 and degradation products of heparan sulfate and keratan sulfate, and a decrease in mitochondrial metabolic activities in obese subjects compared with lean subjects. Our study hereby shows a path to identify new therapeutic targets for treating obesity through combination of high throughput patient data and metabolic modeling. link: http://identifiers.org/pubmed/23511207