SBMLBioModels: H - K

H


Hou2020 - SEIR model of COVID-19 transmission in Wuhan: BIOMD0000000970v0.0.1

A novel coronavirus pneumonia, first identified in Wuhan City and referred to as COVID-19 by the World Health Organizati…

Details

A novel coronavirus pneumonia, first identified in Wuhan City and referred to as COVID-19 by the World Health Organization, has been quickly spreading to other cities and countries. To control the epidemic, the Chinese government mandated a quarantine of the Wuhan city on January 23, 2020. To explore the effectiveness of the quarantine of the Wuhan city against this epidemic, transmission dynamics of COVID-19 have been estimated. A well-mixed "susceptible exposed infectious recovered" (SEIR) compartmental model was employed to describe the dynamics of the COVID-19 epidemic based on epidemiological characteristics of individuals, clinical progression of COVID-19, and quarantine intervention measures of the authority. Considering infected individuals as contagious during the latency period, the well-mixed SEIR model fitting results based on the assumed contact rate of latent individuals are within 6-18, which represented the possible impact of quarantine and isolation interventions on disease infections, whereas other parameter were suppose as unchanged under the current intervention. The present study shows that, by reducing the contact rate of latent individuals, interventions such as quarantine and isolation can effectively reduce the potential peak number of COVID-19 infections and delay the time of peak infection. link: http://identifiers.org/pubmed/32243599

Houser2012_pheromone_Ste12: MODEL1204040000v0.0.1

This model is from the article: Positive roles for negative regulators in the mating response of yeast. Houser JR, F…

Details

All cells must detect and respond to changes in their environment, often through changes in gene expression. The yeast pheromone pathway has been extensively characterized, and is an ideal system for studying transcriptional regulation. Here we combine computational and experimental approaches to study transcriptional regulation mediated by Ste12, the key transcription factor in the pheromone response. Our mathematical model is able to explain multiple counterintuitive experimental results and led to several novel findings. First, we found that the transcriptional repressors Dig1 and Dig2 positively affect transcription by stabilizing Ste12. This stabilization through protein-protein interactions creates a large pool of Ste12 that is rapidly activated following pheromone stimulation. Second, we found that protein degradation follows saturating kinetics, explaining the long half-life of Ste12 in mutants expressing elevated amounts of Ste12. Finally, our model reveals a novel mechanism for robust perfect adaptation through protein-protein interactions that enhance complex stability. This mechanism allows the transcriptional response to act on a shorter time scale than upstream pathway activity. link: http://identifiers.org/pubmed/22669614

Howell2020- Compartmental Logical model of mitotic exit: MODEL2007200001v0.0.1

This model represents the main aspects of regulation of mitotic exit in budding yeast, including the MEN and FEAR networ…

Details

The transition from mitosis into the first gap phase of the cell cycle in budding yeast is controlled by the Mitotic Exit Network (MEN). The network interprets spatiotemporal cues about the progression of mitosis and ensures that release of Cdc14 phosphatase occurs only after completion of key mitotic events. The MEN has been studied intensively; however, a unified understanding of how localisation and protein activity function together as a system is lacking. In this paper, we present a compartmental, logical model of the MEN that is capable of representing spatial aspects of regulation in parallel to control of enzymatic activity. We show that our model is capable of correctly predicting the phenotype of the majority of mutants we tested, including mutants that cause proteins to mislocalise. We use a continuous time implementation of the model to demonstrate that Cdc14 Early Anaphase Release (FEAR) ensures robust timing of anaphase, and we verify our findings in living cells. Furthermore, we show that our model can represent measured cell–cell variation in Spindle Position Checkpoint (SPoC) mutants. This work suggests a general approach to incorporate spatial effects into logical models. We anticipate that the model itself will be an important resource to experimental researchers, providing a rigorous platform to test hypotheses about regulation of mitotic exit. link: http://identifiers.org/doi/10.1371/journal.pbio.3000917

Hu2018 - Dynamics of tumor-CD4+-cytokine-host cells interactions with treatments: BIOMD0000000797v0.0.1

This is a proposed mathematical model describing interactions between tumor cells, CD4+ T cells, cytokines, and host cel…

Details

Mathematical models of interactions between tumor cells, CD4+ T cells, cytokines, and host cells are proposed to investigate the role of CD4+ on tumor regression. Our results suggest that host cells along with the mechanism of production of CD4+ T cells play important roles in driving tumor dynamics. Cancer cells can be eradicated if the tumor has a small growth rate and is also not competitive. Treatments by either CD4+, cytokines, or a combination of the two are applied to study their effectiveness. It is concluded that doses of treatments along with the tumor size are critical in determining the fate of the tumor. Tumor cells can be eliminated completely if doses of treatments by cytokine are large. The treatments are in general more effective if the tumor size is smaller. Bistability is observed in all of the models with or without the treatment strategies indicating that there is a window of opportunity for clearing off the tumor cells. link: http://identifiers.org/doi/10.1016/j.amc.2017.11.009

Parameters:

NameDescription
mu_1 = 0.1Reaction: y_CD4_T_Cells =>, Rate Law: compartment*mu_1*y_CD4_T_Cells
I_2 = 0.0Reaction: => z_Cytokine, Rate Law: compartment*I_2
a_1 = 100000.0; c_1 = 0.2Reaction: x_Tumor_Cells => ; z_Cytokine, Rate Law: compartment*c_1*x_Tumor_Cells*z_Cytokine/(a_1+x_Tumor_Cells)
beta_1 = 0.835; alpha_1 = 1000.0Reaction: => y_CD4_T_Cells; x_Tumor_Cells, z_Cytokine, Rate Law: compartment*beta_1*x_Tumor_Cells*z_Cytokine/(alpha_1+x_Tumor_Cells)
I_1 = 0.0Reaction: => y_CD4_T_Cells, Rate Law: compartment*I_1
delta_2 = 1.0E-7Reaction: y_CD4_T_Cells => ; x_Tumor_Cells, Rate Law: compartment*delta_2*x_Tumor_Cells*y_CD4_T_Cells
r_1 = 0.514; b_1 = 1.02E-9Reaction: => x_Tumor_Cells, Rate Law: compartment*r_1*x_Tumor_Cells*(1-b_1*x_Tumor_Cells)
beta_2 = 5.4; alpha_2 = 1000.0Reaction: => z_Cytokine; x_Tumor_Cells, y_CD4_T_Cells, Rate Law: compartment*beta_2*x_Tumor_Cells*y_CD4_T_Cells/(alpha_2+x_Tumor_Cells)
delta_1 = 1.1E-10Reaction: x_Tumor_Cells => ; w_Healthy_Tissue, Rate Law: compartment*delta_1*x_Tumor_Cells*w_Healthy_Tissue
r_2 = 0.2822; b_2 = 1.0E-9Reaction: => w_Healthy_Tissue, Rate Law: compartment*r_2*w_Healthy_Tissue*(1-b_2*w_Healthy_Tissue)
mu_2 = 34.0Reaction: z_Cytokine =>, Rate Law: compartment*mu_2*z_Cytokine
delta_3 = 5.8E-11Reaction: w_Healthy_Tissue => ; x_Tumor_Cells, Rate Law: compartment*delta_3*x_Tumor_Cells*w_Healthy_Tissue

States:

NameDescription
x Tumor Cells[neoplastic cell]
y CD4 T Cells[CD4-positive helper T cell]
z Cytokine[Interleukin-2; Cytokine]
w Healthy Tissue[Tissue]

Hu2019 - Modeling Pancreatic Cancer Dynamics with Immunotherapy: BIOMD0000000792v0.0.1

This is a mathematical model of pancreatic cancer that includes descriptions of pancreatic cancer cells, pancreatic stel…

Details

We develop a mathematical model of pancreatic cancer that includes pancreatic cancer cells, pancreatic stellate cells, effector cells and tumor-promoting and tumor-suppressing cytokines to investigate the effects of immunotherapies on patient survival. The model is first validated using the survival data of two clinical trials. Local sensitivity analysis of the parameters indicates there exists a critical activation rate of pro-tumor cytokines beyond which the cancer can be eradicated if four adoptive transfers of immune cells are applied. Optimal control theory is explored as a potential tool for searching the best adoptive cellular immunotherapies. Combined immunotherapies between adoptive ex vivo expanded immune cells and TGF-[Formula: see text] inhibition by siRNA treatments are investigated. This study concludes that mono-immunotherapy is unlikely to control the pancreatic cancer and combined immunotherapies between anti-TGF-[Formula: see text] and adoptive transfers of immune cells can prolong patient survival. We show through numerical explorations that how these two types of immunotherapies are scheduled is important to survival. Applying TGF-[Formula: see text] inhibition first followed by adoptive immune cell transfers can yield better survival outcomes. link: http://identifiers.org/pubmed/30843136

Parameters:

NameDescription
r_1 = 0.0195; b_1 = 1.02E-11; beta_1 = 1.7857E-12Reaction: => x_PCC; y_PSC, Rate Law: compartment*(r_1+beta_1*y_PSC)*x_PCC*(1-b_1*x_PCC)
mu_3 = 0.02Reaction: z_Effector_Cells =>, Rate Law: compartment*mu_3*z_Effector_Cells
mu_4 = 0.034Reaction: w_TPC =>, Rate Law: compartment*mu_4*w_TPC
delta_1 = 0.96; m_1 = 1.0E9Reaction: x_PCC => ; z_Effector_Cells, w_TPC, Rate Law: compartment*delta_1*x_PCC*z_Effector_Cells/(m_1+w_TPC)
mu_2 = 0.015Reaction: y_PSC =>, Rate Law: compartment*mu_2*y_PSC
beta_3 = 124.5; m_3 = 1000000.0; k_3 = 2.0E10Reaction: => z_Effector_Cells; v_TSC, w_TPC, Rate Law: compartment*beta_3*z_Effector_Cells*v_TSC/((k_3+v_TSC)*(m_3+w_TPC))
mu_5 = 0.034Reaction: v_TSC =>, Rate Law: compartment*mu_5*v_TSC
r_4 = 12500.0; m_4 = 8.9E10Reaction: => w_TPC; x_PCC, y_PSC, v_TSC, Rate Law: compartment*r_4*x_PCC*y_PSC/(m_4+v_TSC)
k_5 = 1000000.0; beta_5 = 7.3Reaction: => v_TSC; x_PCC, z_Effector_Cells, Rate Law: compartment*beta_5*x_PCC*z_Effector_Cells/(k_5+x_PCC)
beta_2 = 0.125; r_2 = 0.00195; k_2 = 5.6E10; b_2 = 1.7857E-9Reaction: => y_PSC; w_TPC, Rate Law: compartment*(r_2+beta_2*w_TPC/(k_2+w_TPC))*y_PSC*(1-b_2*y_PSC)
v=0.1Reaction: => w_TPC; x_PCC, z_Effector_Cells, Rate Law: compartment*v
mu_6 = 0.66Reaction: R_siRNA =>, Rate Law: compartment*mu_6*R_siRNA
D_0 = 5.0E10Reaction: => R_siRNA, Rate Law: compartment*D_0
r_3 = 3500.0Reaction: => z_Effector_Cells, Rate Law: compartment*r_3

States:

NameDescription
y PSC[pancreatic stellate cell]
z Effector Cells[Effector Immune Cell]
v TSC[Interferon gamma; Cytokine]
w TPC[Interleukin-6; Transforming growth factor beta 1; Cytokine]
R siRNA[Small Interfering RNA]
x PCC[EFO:0002966; neoplastic cell]

Hu2019 - Pancreatic cancer dynamics: BIOMD0000000744v0.0.1

The paper describes a model on the size of pancreatic tumour. Created by COPASI 4.25 (Build 207) This model is descri…

Details

We develop a mathematical model of pancreatic cancer that includes pancreatic cancer cells, pancreatic stellate cells, effector cells and tumor-promoting and tumor- suppressing cytokines to investigate the effects of immunotherapies on patient survival. The model is first validated using the survival data of two clinical trials. Local sen- sitivity analysis of the parameters indicates there exists a critical activation rate of pro-tumor cytokines beyond which the cancer can be eradicated if four adoptive trans- fers of immune cells are applied. Optimal control theory is explored as a potential tool for searching the best adoptive cellular immunotherapies. Combined immunother- apies between adoptive ex vivo expanded immune cells and TGF-β inhibition by siRNA treatments are investigated. This study concludes that mono-immunotherapy is unlikely to control the pancreatic cancer and combined immunotherapies between anti-TGF-β and adoptive transfers of immune cells can prolong patient survival. We show through numerical explorations that how these two types of immunotherapies are scheduled is important to survival. Applying TGF-β inhibition first followed by adoptive immune cell transfers can yield better survival outcomes. link: http://identifiers.org/doi/10.1007/s11538-019-00591-3

Parameters:

NameDescription
r1 = 0.0195 1/d; b1 = 1.02E-11 1Reaction: => x, Rate Law: Pancreatic_tumor*r1*x*(1-x*b1)
m1 = 1.0E8 1; delta1 = 0.96 1/dReaction: x => ; z, w, Rate Law: Pancreatic_tumor*delta1*x*z/(m1+w)
u5 = 0.034 1/dReaction: v =>, Rate Law: Pancreatic_tumor*u5*v
u3 = 0.02 1/dReaction: z =>, Rate Law: Pancreatic_tumor*u3*z
beta3 = 124.5 1/d; m3 = 1000000.0 1; k3 = 2.0E10 1Reaction: => z; v, w, Rate Law: Pancreatic_tumor*beta3*z*v/((k3+v)*(m3+w))
r3 = 3500.0 1/dReaction: => z, Rate Law: Pancreatic_tumor*r3
beta4 = 5.85 1/d; k4 = 1000000.0 1Reaction: => w; x, z, Rate Law: Pancreatic_tumor*beta4*x*z/(k4+x)
u2 = 0.015 1/dReaction: y =>, Rate Law: Pancreatic_tumor*u2*y
u4 = 0.034 1/dReaction: w =>, Rate Law: Pancreatic_tumor*u4*w
beta1 = 3.482115E-12 1/d; b1 = 1.02E-11 1Reaction: => x; y, Rate Law: Pancreatic_tumor*beta1*y*x*(1-x*b1)
beta2 = 0.125 1/d; b2 = 1.7857E-9 1; k2 = 5.6E10 1Reaction: => y; w, Rate Law: Pancreatic_tumor*beta2*w/(k2+w)*y*(1-b2*y)
b2 = 1.7857E-9 1; r2 = 0.00195 1/dReaction: => y, Rate Law: Pancreatic_tumor*r2*y*(1-b2*y)
beta5 = 7.3 1/d; k5 = 1000000.0 1Reaction: => v; x, z, Rate Law: Pancreatic_tumor*beta5*x*z/(k5+x)
m4 = 8.9E10 1; r4 = 12500.0 1/dReaction: => w; x, y, v, Rate Law: Pancreatic_tumor*r4*x*y/(m4+v)

States:

NameDescription
v[Cytokine]
w[Cytokine]
x[Malignant Cell; malignant cell]
z[Natural Killer Cell; CD8-Positive T-Lymphocyte]
y[Pancreatic Stellate Cell; pancreatic stellate cell]

Hu2020 - E. coli translation model: MODEL2006210001v0.0.1

This translation model consists of 274 biochemical reactions, including 119 reactions with non-linear kinetics. This mec…

Details

Protein synthesis is the most expensive process in fast-growing bacteria. Experimentally observed growth rate dependencies of the translation machinery form the basis of powerful phenomenological growth laws; however, a quantitative theory on the basis of biochemical and biophysical constraints is lacking. Here, we show that the growth rate-dependence of the concentrations of ribosomes, tRNAs, mRNA, and elongation factors observed in Escherichia coli can be predicted accurately from a minimization of cellular costs in a mechanistic model of protein translation. The model is constrained only by the physicochemical properties of the molecules and has no adjustable parameters. The costs of individual components (made of protein and RNA parts) can be approximated through molecular masses, which correlate strongly with alternative cost measures such as the molecules’ carbon content or the requirement of energy or enzymes for their biosynthesis. Analogous cost minimization approaches may facilitate similar quantitative insights also for other cellular subsystems. link: http://identifiers.org/doi/10.1038/s41467-020-18948-x

Huang1996 - Ultrasensitivity in MAPK cascade: BIOMD0000000009v0.0.1

Huang1996 - Ultrasensitivity in MAPK cascadeThe temporal sequence of kinase activation, from MAPKKK (activated RAF) to t…

Details

The mitogen-activated protein kinase (MAPK) cascade is a highly conserved series of three protein kinases implicated in diverse biological processes. Here we demonstrate that the cascade arrangement has unexpected consequences for the dynamics of MAPK signaling. We solved the rate equations for the cascade numerically and found that MAPK is predicted to behave like a highly cooperative enzyme, even though it was not assumed that any of the enzymes in the cascade were regulated cooperatively. Measurements of MAPK activation in Xenopus oocyte extracts confirmed this prediction. The stimulus/response curve of the MAPK was found to be as steep as that of a cooperative enzyme with a Hill coefficient of 4-5, well in excess of that of the classical allosteric protein hemoglobin. The shape of the MAPK stimulus/ response curve may make the cascade particularly appropriate for mediating processes like mitogenesis, cell fate induction, and oocyte maturation, where a cell switches from one discrete state to another. link: http://identifiers.org/pubmed/8816754

Parameters:

NameDescription
a10=1000.0; d10=150.0Reaction: PP_K + KPase => KPase_PP_K, Rate Law: compartment*(a10*PP_K*KPase-d10*KPase_PP_K)
k6=150.0Reaction: KKPase_PP_KK => P_KK + KKPase, Rate Law: compartment*k6*KKPase_PP_KK
a1=1000.0; d1=150.0Reaction: KKK + E1 => E1_KKK, Rate Law: compartment*(a1*E1*KKK-d1*E1_KKK)
a3=1000.0; d3=150.0Reaction: KK + P_KKK => P_KKK_KK, Rate Law: compartment*(a3*KK*P_KKK-d3*P_KKK_KK)
K_PP_norm_max = 0.900049Reaction: rel_K_PP_max = K_PP_norm/K_PP_norm_max, Rate Law: missing
a6=1000.0; d6=150.0Reaction: PP_KK + KKPase => KKPase_PP_KK, Rate Law: compartment*(a6*PP_KK*KKPase-d6*KKPase_PP_KK)
k8=150.0Reaction: KPase_P_K => K + KPase, Rate Law: compartment*k8*KPase_P_K
k10=150.0Reaction: KPase_PP_K => P_K + KPase, Rate Law: compartment*k10*KPase_PP_K
k2=150.0Reaction: E1_KKK => E1 + P_KKK, Rate Law: compartment*k2*E1_KKK
a2=1000.0; d2=150.0Reaction: P_KKK + E2 => E2_P_KKK, Rate Law: compartment*(a2*E2*P_KKK-d2*E2_P_KKK)
d5=150.0; a5=1000.0Reaction: P_KK + P_KKK => P_KKK_P_KK, Rate Law: compartment*(a5*P_KK*P_KKK-d5*P_KKK_P_KK)
k3=150.0Reaction: P_KKK_KK => P_KK + P_KKK, Rate Law: compartment*k3*P_KKK_KK
a9=1000.0; d9=150.0Reaction: P_K + PP_KK => PP_KK_P_K, Rate Law: compartment*(a9*P_K*PP_KK-d9*PP_KK_P_K)
k4=150.0Reaction: KKPase_P_KK => KK + KKPase, Rate Law: compartment*k4*KKPase_P_KK
k5=150.0Reaction: P_KKK_P_KK => PP_KK + P_KKK, Rate Law: compartment*k5*P_KKK_P_KK
k9=150.0Reaction: PP_KK_P_K => PP_KK + PP_K, Rate Law: compartment*k9*PP_KK_P_K
a7=1000.0; d7=150.0Reaction: K + PP_KK => PP_KK_K, Rate Law: compartment*(a7*K*PP_KK-d7*PP_KK_K)
a4=1000.0; d4=150.0Reaction: P_KK + KKPase => KKPase_P_KK, Rate Law: compartment*(a4*P_KK*KKPase-d4*KKPase_P_KK)
k7=150.0Reaction: PP_KK_K => P_K + PP_KK, Rate Law: compartment*k7*PP_KK_K
d8=150.0; a8=1000.0Reaction: P_K + KPase => KPase_P_K, Rate Law: compartment*(a8*P_K*KPase-d8*KPase_P_K)

States:

NameDescription
KPase P K[Mitogen-activated protein kinase 1; Dual specificity protein phosphatase 1-B]
PP K[Mitogen-activated protein kinase 1]
PP KK K[Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
PP KK P K[Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
KKPaseMAPKK-Pase
KKK P normKKK_P_norm
KKPase P KK[Dual specificity mitogen-activated protein kinase kinase 1]
P KKK KK[Serine/threonine-protein kinase mos; Dual specificity mitogen-activated protein kinase kinase 1]
P K[Mitogen-activated protein kinase 1]
KKPase PP KK[Dual specificity mitogen-activated protein kinase kinase 1]
E1[IPR003577]
P KKK[Serine/threonine-protein kinase mos]
KKK[Serine/threonine-protein kinase mos]
KK PP normKK_PP_norm
rel K PP maxrelative maximal K_PP
KPase PP K[Dual specificity protein phosphatase 1-B; Mitogen-activated protein kinase 1]
E2MAPKKK inactivator
PP KK[Dual specificity mitogen-activated protein kinase kinase 1]
K PP normK_PP_norm
E1 KKK[Serine/threonine-protein kinase mos; IPR003577]
KPase[Dual specificity protein phosphatase 1-B]
P KKK P KK[Serine/threonine-protein kinase mos; Dual specificity mitogen-activated protein kinase kinase 1]
E2 P KKK[Serine/threonine-protein kinase mos]
KK[Dual specificity mitogen-activated protein kinase kinase 1]
P KK[Dual specificity mitogen-activated protein kinase kinase 1]
K[Mitogen-activated protein kinase 1]

Huang2014 - Systematic modeling for the insulin signaling network mediated by IRS1 and IRS2: MODEL1912090001v0.0.1

This is a mathematical model is which is a combination of systemic models for the insulin signaling network as mediated…

Details

The hepatic insulin signaling mediated by insulin receptor substrates IRS1 and IRS2 plays a central role in maintaining glucose homeostasis under different physiological conditions. Although functions of individual components in the signaling network have been extensively studied, our knowledge is still limited with regard to how the signals are integrated and coordinated in the complex network to render their functional roles. In this study, we construct systematic models for the insulin signaling network mediated by IRS1 and IRS2, through the integration of current knowledge in the literature into mathematical models of insulin signaling pathways. We hypothesize that the specificity of the IRS signaling mechanisms emerges from the wiring and kinetics of the entire network. A discrete dynamic model is first constructed to account for the numerous dynamic features in the system, i.e., complex feedback circuits, different regulatory time-scales and cross-talks between pathways. Our simulation shows that the wiring of the network determines different functions of IRS1 and IRS2. We further collate and reconstruct a kinetic model of the network as a system of ordinary differential equations to provide an informative model for predicting phenotypes. A sensitivity analysis is applied to identify essential regulators for the signaling process. link: http://identifiers.org/pubmed/24703981

Huarat2016 -Starvation-induced Ser/Thr protein kinase ArnS (Saci_1181) (Model 14): MODEL1607210000v0.0.1

Huarat2016 -Starvation-induced Ser/Thr protein kinase ArnS (Saci_1181) (Model 14)This model is described in the article:…

Details

Organisms have evolved motility organelles that allow them to move to favourable habitats. Cells integrate environmental stimuli into intracellular signals to motility machineries to direct this migration. Many motility organelles are complex surface appendages that have evolved a tight, hierarchical regulation of expression. In the crenearchaeon Sulfolobus acidocaldarius, biosynthesis of the archaellum is regulated by regulatory network proteins that control expression of archaellum components in a phosphorylation-dependent manner. A major trigger for archaellum expression is nutrient starvation, but although some components are known, the regulatory cascade triggered by starvation is poorly understood. In this work, the starvation-induced Ser/Thr protein kinase ArnS (Saci_1181) which is located proximally to the archaellum operon was identified. Deletion of arnS results in reduced motility, though the archaellum is properly assembled. Therefore, our experimental and modelling results indicate that ArnS plays an essential role in the precisely controlled expression of archaellum components during starvation-induced motility in Sulfolobus acidocaldarius. Furthermore they combined in vivo experiments and mathematical models to describe for the first time in archaea the dynamics of key regulators of archaellum expression. link: http://identifiers.org/pubmed/27731916

Hui2016 - Age-related changes in articular cartilage: BIOMD0000000560v0.0.1

Hui2014 - Age-related changes in articular cartilageThis model is described in the article: [Oxidative changes and sign…

Details

To use a computational approach to investigate the cellular and extracellular matrix changes that occur with age in the knee joints of mice.Knee joints from an inbred C57/BL1/6 (ICRFa) mouse colony were harvested at 3-30 months of age. Sections were stained with H&E, Safranin-O, Picro-sirius red and antibodies to matrix metalloproteinase-13 (MMP-13), nitrotyrosine, LC-3B, Bcl-2, and cleaved type II collagen used for immunohistochemistry. Based on this and other data from the literature, a computer simulation model was built using the Systems Biology Markup Language using an iterative approach of data analysis and modelling. Individual parameters were subsequently altered to assess their effect on the model.A progressive loss of cartilage matrix occurred with age. Nitrotyrosine, MMP-13 and activin receptor-like kinase-1 (ALK1) staining in cartilage increased with age with a concomitant decrease in LC-3B and Bcl-2. Stochastic simulations from the computational model showed a good agreement with these data, once transforming growth factor-β signalling via ALK1/ALK5 receptors was included. Oxidative stress and the interleukin 1 pathway were identified as key factors in driving the cartilage breakdown associated with ageing.A progressive loss of cartilage matrix and cellularity occurs with age. This is accompanied with increased levels of oxidative stress, apoptosis and MMP-13 and a decrease in chondrocyte autophagy. These changes explain the marked predisposition of joints to develop osteoarthritis with age. Computational modelling provides useful insights into the underlying mechanisms involved in age-related changes in musculoskeletal tissues. link: http://identifiers.org/pubmed/25475114

Parameters:

NameDescription
kphosNFkB = 0.001Reaction: NFkB + p38_P => NFkB_P + p38_P; NFkB, p38_P, Rate Law: kphosNFkB*NFkB*p38_P
kphosp38ROS = 1.0E-4Reaction: p38 + ROS => p38_P + ROS; p38, ROS, Rate Law: kphosp38ROS*p38*ROS
kdamLys = 5.0E-6Reaction: Lys_A + ROS => Lys_I + ROS; Lys_A, ROS, Rate Law: kdamLys*Lys_A*ROS/(10+ROS)
ksynNatP = 0.0Reaction: Source => NatP; Source, Rate Law: ksynNatP*Source
ksynMMP13Runx2 = 1.5E-6Reaction: Runx2_A => proMMP13 + Runx2_A; Runx2_A, Rate Law: ksynMMP13Runx2*Runx2_A
kinactNFkB = 0.1Reaction: NFkB + IkB => IkB_NFkB; NFkB, IkB, Rate Law: kinactNFkB*NFkB*IkB
kgenROS = 5.0E-4Reaction: Source => ROS; Source, Rate Law: kgenROS*Source
kbinBaxToBcl2Bec = 1.67E-4Reaction: Bax + Bcl2_Beclin => Bax_Bcl2_Beclin; Bax, Bcl2_Beclin, Rate Law: kbinBaxToBcl2Bec*Bax*Bcl2_Beclin
ksynRAGE = 1.0E-4Reaction: NFkB_P => NFkB_P + RAGE; NFkB_P, Rate Law: ksynRAGE*NFkB_P
ksynAggrecan = 1.0E-6Reaction: AcanmRNA => AcanmRNA + Aggrecan; AcanmRNA, Rate Law: ksynAggrecan*AcanmRNA
kactCaspp38 = 8.0E-7Reaction: Caspase_I + p38_P => Caspase_A + p38_P; Caspase_I, p38_P, Rate Law: kactCaspp38*Caspase_I*p38_P
kactTgfbMMP2 = 1.0E-7Reaction: Tgfb_I + MMP2 => Tgfb_A + MMP2; Tgfb_I, MMP2, Rate Law: kactTgfbMMP2*Tgfb_I*MMP2
kinactCasp = 3.0E-4Reaction: Caspase_A => Caspase_I; Caspase_A, Rate Law: kinactCasp*Caspase_A
kbinSmad1Smad4 = 5.0E-5Reaction: Smad1_P + Smad4 => Smad1_P_Smad4; Smad1_P, Smad4, Rate Law: kbinSmad1Smad4*Smad1_P*Smad4
ksynSmad7 = 1.0E-5Reaction: Smad2_P_Smad4 => Smad2_P_Smad4 + Smad7; Smad2_P_Smad4, Rate Law: ksynSmad7*Smad2_P_Smad4
kgenROSbyp38 = 1.0E-4Reaction: p38_P => p38_P + ROS; p38_P, Rate Law: kgenROSbyp38*p38_P
kinhibLys = 7.0E-6Reaction: Lys_A => Lys_I; Lys_A, Rate Law: kinhibLys*Lys_A
kphosSmad2 = 4.0E-5Reaction: Tgfb_Alk5_dimer + Smad2 => Tgfb_Alk5_dimer + Smad2_P; Tgfb_Alk5_dimer, Smad2, Rate Law: kphosSmad2*Tgfb_Alk5_dimer*Smad2
krelSmad1Smad4 = 0.0167Reaction: Smad1_P_Smad4 => Smad1_P + Smad4; Smad1_P_Smad4, Rate Law: krelSmad1Smad4*Smad1_P_Smad4
kactMMP13 = 1.0E-4Reaction: proMMP13 => MMP13; proMMP13, Rate Law: kactMMP13*proMMP13
kbinBcl2Beclin = 7.5E-5Reaction: Bcl2 + Beclin => Bcl2_Beclin; Bcl2, Beclin, Rate Law: kbinBcl2Beclin*Bcl2*Beclin
ksynCol2mRNASmad = 1.0E-6Reaction: Smad2_P_Smad4 => Smad2_P_Smad4 + Col2mRNA; Smad2_P_Smad4, Rate Law: ksynCol2mRNASmad*Smad2_P_Smad4
krelBcl2Beclin = 5.0E-4Reaction: Bcl2_Beclin => Bcl2 + Beclin; Bcl2_Beclin, Rate Law: krelBcl2Beclin*Bcl2_Beclin
krelAlk1Alk5 = 0.01Reaction: Alk1_Alk5 => Alk1 + Alk5; Alk1_Alk5, Rate Law: krelAlk1Alk5*Alk1_Alk5
kdegSox9mRNA = 1.0E-4Reaction: Sox9mRNA => Sink; Sox9mRNA, Rate Law: kdegSox9mRNA*Sox9mRNA
kgenROSbyDamP = 1.0E-4Reaction: DamP => DamP + ROS; DamP, Rate Law: kgenROSbyDamP*DamP
kdephosp38 = 0.01Reaction: p38_P => p38; p38_P, Rate Law: kdephosp38*p38_P
krelBecBaxBcl2 = 0.0167Reaction: Bax_Bcl2_Beclin => Beclin + Bax_Bcl2; Bax_Bcl2_Beclin, Rate Law: krelBecBaxBcl2*Bax_Bcl2_Beclin
krelSmad7Alk5 = 1.0E-6Reaction: Tgfb_Alk5_dimer_Smad7 => Tgfb_Alk5_dimer + Smad7; Tgfb_Alk5_dimer_Smad7, Rate Law: krelSmad7Alk5*Tgfb_Alk5_dimer_Smad7
kbinSmad7Alk1 = 0.5Reaction: Tgfb_Alk1_Alk5 + Smad7 => Tgfb_Alk1_Alk5_Smad7; Tgfb_Alk1_Alk5, Smad7, Rate Law: kbinSmad7Alk1*Tgfb_Alk1_Alk5*Smad7
kdegSox9 = 4.8E-5Reaction: Sox9 => Sink; Sox9, Rate Law: kdegSox9*Sox9
kdegBcl2 = 1.67E-4Reaction: Bcl2 => Sink; Bcl2, Rate Law: kdegBcl2*Bcl2
krelSmad2Smad4 = 0.0167Reaction: Smad2_P_Smad4 => Smad2_P + Smad4; Smad2_P_Smad4, Rate Law: krelSmad2Smad4*Smad2_P_Smad4
ksynIkB = 0.001Reaction: NFkB_P => NFkB_P + IkB; NFkB_P, Rate Law: ksynIkB*NFkB_P
krelSmad7Alk1 = 0.001Reaction: Tgfb_Alk1_Alk5_Smad7 => Tgfb_Alk1_Alk5 + Smad7; Tgfb_Alk1_Alk5_Smad7, Rate Law: krelSmad7Alk1*Tgfb_Alk1_Alk5_Smad7
kbinSmad2Smad4 = 1.0E-4Reaction: Smad2_P + Smad4 => Smad2_P_Smad4; Smad2_P, Smad4, Rate Law: kbinSmad2Smad4*Smad2_P*Smad4
ksynSox9 = 4.8E-4Reaction: Sox9mRNA => Sox9mRNA + Sox9; Sox9mRNA, Rate Law: ksynSox9*Sox9mRNA
kbinSmad7Alk5 = 2.0E-5Reaction: Tgfb_Alk5_dimer + Smad7 => Tgfb_Alk5_dimer_Smad7; Tgfb_Alk5_dimer, Smad7, Rate Law: kbinSmad7Alk5*Tgfb_Alk5_dimer*Smad7
kactMMP2 = 1.0E-7Reaction: proMMP2 => MMP2; proMMP2, Rate Law: kactMMP2*proMMP2
kdegMMP2 = 6.4E-6Reaction: MMP2 => Sink; MMP2, Rate Law: kdegMMP2*MMP2
ksynAlk1 = 5.0E-6Reaction: Source => Alk1; Source, Rate Law: ksynAlk1*Source
krelBaxBcl2 = 0.00167Reaction: Bax_Bcl2 => Bax + Bcl2; Bax_Bcl2, Rate Law: krelBaxBcl2*Bax_Bcl2
ksynCol2mRNASox9A = 1.0E-6Reaction: Sox9_A => Sox9_A + Col2mRNA; Sox9_A, Rate Law: ksynCol2mRNASox9A*Sox9_A
kdegBcl2Casp = 0.00167Reaction: Bcl2 + Caspase_A => Sink + Caspase_A; Bcl2, Caspase_A, Rate Law: kdegBcl2Casp*Bcl2*Caspase_A
kactRunx2 = 0.001Reaction: Runx2_I + Smad1_P_Smad4 => Runx2_A + Smad1_P_Smad4; Runx2_I, Smad1_P_Smad4, Rate Law: kactRunx2*Runx2_I*Smad1_P_Smad4
krelTgfbAlk5 = 1.0E-6Reaction: Tgfb_Alk5_dimer => Tgfb_A + Alk5_dimer; Tgfb_Alk5_dimer, Rate Law: krelTgfbAlk5*Tgfb_Alk5_dimer
kgenROSbyRAGE = 4.0E-4Reaction: RAGE => RAGE + ROS; RAGE, Rate Law: kgenROSbyRAGE*RAGE
kdephosSmad1Smad7 = 6.0E-4Reaction: Smad1_P + Smad7 => Smad1 + Smad7; Smad1_P, Smad7, Rate Law: kdephosSmad1Smad7*Smad1_P*Smad7
kdephosNFkB = 0.01Reaction: NFkB_P => NFkB; NFkB_P, Rate Law: kdephosNFkB*NFkB_P
kremROS = 3.83E-4Reaction: ROS => Sink; ROS, Rate Law: kremROS*ROS
kremROSbySOD = 1.0E-4Reaction: ROS + SOD => SOD; SOD, ROS, Rate Law: kremROSbySOD*SOD*ROS
kinactRunx2 = 5.0E-4Reaction: Runx2_A + Smad2_P_Smad4 => Runx2_I + Smad2_P_Smad4; Runx2_A, Smad2_P_Smad4, Rate Law: kinactRunx2*Runx2_A*Smad2_P_Smad4
ksynSOD = 0.002Reaction: NFkB_P => NFkB_P + SOD; NFkB_P, Rate Law: ksynSOD*NFkB_P
kactLys = 1.0E-8Reaction: Lys_I + Beclin => Lys_A + Beclin; Lys_I, Beclin, Rate Law: kactLys*Lys_I*Beclin
kbinBecToBaxBcl2 = 1.67E-5Reaction: Beclin + Bax_Bcl2 => Bax_Bcl2_Beclin; Beclin, Bax_Bcl2, Rate Law: kbinBecToBaxBcl2*Beclin*Bax_Bcl2
ksynSox9mRNA = 1.0E-5Reaction: Source => Sox9mRNA; Source, Rate Law: ksynSox9mRNA*Source
kinactBec = 5.0E-10Reaction: Beclin => Beclin_I; Beclin, Rate Law: kinactBec*Beclin
kactCaspBecI = 8.3E-7Reaction: Caspase_I + Beclin_I => Caspase_A + Beclin_I; Caspase_I, Beclin_I, Rate Law: kactCaspBecI*Caspase_I*Beclin_I
kdamNatP = 8.0E-6Reaction: NatP + ROS => DamP + ROS; NatP, ROS, Rate Law: kdamNatP*NatP*ROS/(10+ROS)
kinactTgfb = 0.05Reaction: Tgfb_A => Tgfb_I; Tgfb_A, Rate Law: kinactTgfb*Tgfb_A
kinactCaspBcl2 = 3.0E-4Reaction: Caspase_A + Bcl2_Beclin => Caspase_I + Bcl2_Beclin; Caspase_A, Bcl2_Beclin, Rate Law: kinactCaspBcl2*Caspase_A*Bcl2_Beclin
kdegIkB = 1.0E-6Reaction: ROS + IkB_NFkB => ROS + NFkB; ROS, IkB_NFkB, Rate Law: kdegIkB*ROS*IkB_NFkB
kdegBcl2ROS = 0.00167Reaction: Bcl2 + ROS => Sink + ROS; Bcl2, ROS, Rate Law: kdegBcl2ROS*Bcl2*ROS
krelBaxBcl2Bec = 0.00167Reaction: Bax_Bcl2_Beclin => Bax + Bcl2_Beclin; Bax_Bcl2_Beclin, Rate Law: krelBaxBcl2Bec*Bax_Bcl2_Beclin
kbinTgfbAlk5 = 3.0E-5Reaction: Tgfb_A + Alk5_dimer => Tgfb_Alk5_dimer; Tgfb_A, Alk5_dimer, Rate Law: kbinTgfbAlk5*Tgfb_A*Alk5_dimer
kbinBaxBcl2 = 1.67Reaction: Bax + Bcl2 => Bax_Bcl2; Bax, Bcl2, Rate Law: kbinBaxBcl2*Bax*Bcl2
ksynSox9mRNASox9A = 5.0E-6Reaction: Sox9_A => Sox9_A + Sox9mRNA; Sox9_A, Rate Law: ksynSox9mRNASox9A*Sox9_A
kbinTgfbAlk1 = 2.0E-5Reaction: Tgfb_A + Alk1_Alk5 => Tgfb_Alk1_Alk5; Tgfb_A, Alk1_Alk5, Rate Law: kbinTgfbAlk1*Tgfb_A*Alk1_Alk5
kactCasp = 1.0E-7Reaction: Caspase_I + Bax => Caspase_A + Bax; Caspase_I, Bax, Rate Law: kactCasp*Caspase_I*Bax
kactSox9 = 5.0E-6Reaction: Smad2_P_Smad4 + Sox9 => Smad2_P_Smad4 + Sox9_A; Smad2_P_Smad4, Sox9, Rate Law: kactSox9*Smad2_P_Smad4*Sox9

States:

NameDescription
Beclin[Beclin-1]
Sox9mRNA[Transcription factor SOX-9]
Bax Bcl2 Beclin I[Apoptosis regulator BAX; Apoptosis regulator Bcl-2; Beclin-1]
Runx2 A[Runt-related transcription factor 2]
Bax Bcl2 Beclin[Apoptosis regulator BAX; Apoptosis regulator Bcl-2; Beclin-1]
Caspase A[Caspase-3]
Sox9 A[Transcription factor SOX-9]
Bax[Apoptosis regulator BAX]
Bcl2 Beclin[Apoptosis regulator Bcl-2; Beclin-1]
Alk1[Serine/threonine-protein kinase receptor R3]
Beclin I[Beclin-1]
Smad2[Mothers against decapentaplegic homolog 2]
Smad2 P Smad4[Mothers against decapentaplegic homolog 2; Mothers against decapentaplegic homolog 4; phosphoprotein]
Smad4[Mothers against decapentaplegic homolog 4]
ROS[reactive oxygen species]
Tgfb A[Transforming growth factor beta-1]
Bcl2[Apoptosis regulator Bcl-2]
NFkB P[Transcription factor p65; phosphoprotein]
RAGE[Advanced glycosylation end product-specific receptor]
Lys I[lysosome]
Sox9[Transcription factor SOX-9]
Smad2 P[Mothers against decapentaplegic homolog 2; phosphoprotein]
Bcl2 Beclin I[Apoptosis regulator Bcl-2; Beclin-1]
p38[Mitogen-activated protein kinase 14]
Smad1[Mothers against decapentaplegic homolog 1]
MMP2[72 kDa type IV collagenase]
Smad1 P Smad4[Mothers against decapentaplegic homolog 1; Mothers against decapentaplegic homolog 4; phosphoprotein]
NatPNatP
NFkB[Transcription factor p65]
Smad7[Mothers against decapentaplegic homolog 7]
AcanmRNA[Aggrecan core protein]
Caspase I[Caspase-3]
MMP13[Collagenase 3]

Hund2004_VentricularEpicardialAction: MODEL0848116681v0.0.1

This a model from the article: Rate dependence and regulation of action potential and calcium transient in a canine ca…

Details

Computational biology is a powerful tool for elucidating arrhythmogenic mechanisms at the cellular level, where complex interactions between ionic processes determine behavior. A novel theoretical model of the canine ventricular epicardial action potential and calcium cycling was developed and used to investigate ionic mechanisms underlying Ca2+ transient (CaT) and action potential duration (APD) rate dependence.The Ca2+/calmodulin-dependent protein kinase (CaMKII) regulatory pathway was integrated into the model, which included a novel Ca2+-release formulation, Ca2+ subspace, dynamic chloride handling, and formulations for major ion currents based on canine ventricular data. Decreasing pacing cycle length from 8000 to 300 ms shortened APD primarily because of I(Ca(L)) reduction, with additional contributions from I(to1), I(NaK), and late I(Na). CaT amplitude increased as cycle length decreased from 8000 to 500 ms. This positive rate-dependent property depended on CaMKII activity.CaMKII is an important determinant of the rate dependence of CaT but not of APD, which depends on ion-channel kinetics. The model of CaMKII regulation may serve as a paradigm for modeling effects of other regulatory pathways on cell function. link: http://identifiers.org/pubmed/15505083

Hunziker2010_p53_StressSpecificResponse: BIOMD0000000252v0.0.1

This a model from the article: Stress-specific response of the p53-Mdm2 feedback loop Alexander Hunziker, Mogens H…

Details

The p53 signalling pathway has hundreds of inputs and outputs. It can trigger cellular senescence, cell-cycle arrest and apoptosis in response to diverse stress conditions, including DNA damage, hypoxia and nutrient deprivation. Signals from all these inputs are channeled through a single node, the transcription factor p53. Yet, the pathway is flexible enough to produce different downstream gene expression patterns in response to different stresses.We construct a mathematical model of the negative feedback loop involving p53 and its inhibitor, Mdm2, at the core of this pathway, and use it to examine the effect of different stresses that trigger p53. In response to DNA damage, hypoxia, etc., the model exhibits a wide variety of specific output behaviour - steady states with low or high levels of p53 and Mdm2, as well as spiky oscillations with low or high average p53 levels.We show that even a simple negative feedback loop is capable of exhibiting the kind of flexible stress-specific response observed in the p53 system. Further, our model provides a framework for predicting the differences in p53 response to different stresses and single nucleotide polymorphisms. link: http://identifiers.org/pubmed/20624280

Parameters:

NameDescription
S = 1000.0; k_f = 5000.0; gamma = 0.2; k_b = 7200.0; alpha = 0.1Reaction: p = ((S-k_f*p*m)-alpha*p)+(k_b+gamma)*pm, Rate Law: ((S-k_f*p*m)-alpha*p)+(k_b+gamma)*pm
beta = 0.6; k_t = 0.03Reaction: mm = k_t*p^2-beta*mm, Rate Law: k_t*p^2-beta*mm
k_f = 5000.0; delta = 11.0; gamma = 0.2; k_tl = 1.4; k_b = 7200.0Reaction: m = ((k_tl*mm-k_f*p*m)+(k_b+delta)*pm)-gamma*m, Rate Law: ((k_tl*mm-k_f*p*m)+(k_b+delta)*pm)-gamma*m
k_f = 5000.0; delta = 11.0; gamma = 0.2; k_b = 7200.0Reaction: pm = (k_f*p*m-(k_b+delta)*pm)-gamma*pm, Rate Law: (k_f*p*m-(k_b+delta)*pm)-gamma*pm

States:

NameDescription
m[MDM2; E3 ubiquitin-protein ligase Mdm2]
mm[messenger RNA; RNA]
pm[Cellular tumor antigen p53; E3 ubiquitin-protein ligase Mdm2]
p[TP53; Cellular tumor antigen p53]

Hutchinson2016 - Vascular phenotype identification and anti-angiogenic treatment recommendation: MODEL1911130007v0.0.1

This is a spatially averaged multiscale mathematical model of tumor angiogenesis. The model describes the dynamics of VE…

Details

The development of anti-angiogenic drugs for cancer therapy has yielded some promising candidates, but novel approaches for interventions to angiogenesis have led to disappointing results. In addition, there is a shortage of biomarkers that are predictive of response to anti-angiogenic treatments. Consequently, the complex biochemical and physiological basis for tumour angiogenesis remains incompletely understood. We have adopted a mathematical approach to address these issues, formulating a spatially averaged multiscale model that couples the dynamics of VEGF, Ang1, Ang2 and PDGF, with those of mature and immature endothelial cells and pericyte cells. The model reproduces qualitative experimental results regarding pericyte coverage of vessels after treatment by anti-Ang2, anti-VEGF and combination anti-VEGF/anti-Ang2 antibodies. We used the steady state behaviours of the model to characterise angiogenic and non-angiogenic vascular phenotypes, and used mechanistic perturbations representing hypothetical anti-angiogenic treatments to generate testable hypotheses regarding transitions to non-angiogenic phenotypes that depend on the pre-treatment vascular phenotype. Additionally, we predicted a synergistic effect between anti-VEGF and anti-Ang2 treatments when applied to an immature pre-treatment vascular phenotype, but not when applied to a normalised angiogenic pre-treatment phenotype. Based on these findings, we conclude that changes in vascular phenotype are predicted to be useful as an experimental biomarker of response to treatment. Further, our analysis illustrates the potential value of non-spatial mathematical models for generating tractable predictions regarding the action of anti-angiogenic therapies. link: http://identifiers.org/pubmed/26987523

Huthmacher2010_HumanErythrocyte_MetabolicNetwork: MODEL1111240001v0.0.1

This model is from the article: Antimalarial drug targets in Plasmodium falciparum predicted by st age-specific metabo…

Details

BACKGROUND: Despite enormous efforts to combat malaria the disease still afflicts up to half a billion people each year of which more than one million die. Currently no approved vaccine is available and resistances to antimalarials are widely spread. Hence, new antimalarial drugs are urgently needed. RESULTS: Here, we present a computational analysis of the metabolism of Plasmodium falciparum, the deadliest malaria pathogen. We assembled a compartmentalized metabolic model and predicted life cycle stage specific metabolism with the help of a flux balance approach that integrates gene expression data. Predicted metabolite exchanges between parasite and host were found to be in good accordance with experimental findings when the parasite's metabolic network was embedded into that of its host (erythrocyte). Knock-out simulations identified 307 indispensable metabolic reactions within the parasite. 35 out of 57 experimentally demonstrated essential enzymes were recovered and another 16 enzymes, if additionally the assumption was made that nutrient uptake from the host cell is limited and all reactions catalyzed by the inhibited enzyme are blocked. This predicted set of putative drug targets, shown to be enriched with true targets by a factor of at least 2.75, was further analyzed with respect to homology to human enzymes, functional similarity to therapeutic targets in other organisms and their predicted potency for prophylaxis and disease treatment. CONCLUSIONS: The results suggest that the set of essential enzymes predicted by our flux balance approach represents a promising starting point for further drug development. link: http://identifiers.org/pubmed/20807400

Huthmacher2010_MetabolicNetwork_P.falciparum: MODEL1111240000v0.0.1

This model is from the article: Antimalarial drug targets in Plasmodium falciparum predicted by stage-specific metabol…

Details

BACKGROUND: Despite enormous efforts to combat malaria the disease still afflicts up to half a billion people each year of which more than one million die. Currently no approved vaccine is available and resistances to antimalarials are widely spread. Hence, new antimalarial drugs are urgently needed. RESULTS: Here, we present a computational analysis of the metabolism of Plasmodium falciparum, the deadliest malaria pathogen. We assembled a compartmentalized metabolic model and predicted life cycle stage specific metabolism with the help of a flux balance approach that integrates gene expression data. Predicted metabolite exchanges between parasite and host were found to be in good accordance with experimental findings when the parasite's metabolic network was embedded into that of its host (erythrocyte). Knock-out simulations identified 307 indispensable metabolic reactions within the parasite. 35 out of 57 experimentally demonstrated essential enzymes were recovered and another 16 enzymes, if additionally the assumption was made that nutrient uptake from the host cell is limited and all reactions catalyzed by the inhibited enzyme are blocked. This predicted set of putative drug targets, shown to be enriched with true targets by a factor of at least 2.75, was further analyzed with respect to homology to human enzymes, functional similarity to therapeutic targets in other organisms and their predicted potency for prophylaxis and disease treatment. CONCLUSIONS: The results suggest that the set of essential enzymes predicted by our flux balance approach represents a promising starting point for further drug development. link: http://identifiers.org/pubmed/20807400

Hynne2001_Glycolysis: BIOMD0000000061v0.0.1

The model reproduces Fig 6 of the paper. The stoichiometry and rate of reactions involving uptake of metabolites from ex…

Details

We present a powerful, general method of fitting a model of a biochemical pathway to experimental substrate concentrations and dynamical properties measured at a stationary state, when the mechanism is largely known but kinetic parameters are lacking. Rate constants and maximum velocities are calculated from the experimental data by simple algebra without integration of kinetic equations. Using this direct approach, we fit a comprehensive model of glycolysis and glycolytic oscillations in intact yeast cells to data measured on a suspension of living cells of Saccharomyces cerevisiae near a Hopf bifurcation, and to a large set of stationary concentrations and other data estimated from comparable batch experiments. The resulting model agrees with almost all experimentally known stationary concentrations and metabolic fluxes, with the frequency of oscillation and with the majority of other experimentally known kinetic and dynamical variables. The functional forms of the rate equations have not been optimized. link: http://identifiers.org/pubmed/11744196

Parameters:

NameDescription
K15NADH=0.13 milliMolar; K15INADH=0.034 milliMolar; V15m=81.4797 mM per minute; K15DHAP=25.0 milliMolar; K15INAD=0.13 milliMolarReaction: DHAP + NADH => Glyc + NAD, Rate Law: cytosol*V15m*DHAP/(K15DHAP*(1+K15INADH/NADH*(1+NAD/K15INAD))+DHAP*(1+K15NADH/NADH*(1+NAD/K15INAD)))
Yvol=59.0 dimensionless; K2IG6P=1.2 milliMolar; K2IIG6P=7.2 milliMolar; P2=1.0 dimensionless; V2f=1014.96 mM per minute; V2r=1014.96 mM per minute; K2Glc=1.7 milliMolarReaction: GlcX => Glc; G6P, Rate Law: extracellular*V2f/Yvol*GlcX/K2Glc/(1+GlcX/K2Glc+(P2*GlcX/K2Glc+1)/(P2*Glc/K2Glc+1)*(1+Glc/K2Glc+G6P/K2IG6P+Glc*G6P/(K2Glc*K2IIG6P)))-cytosol*V2r/Yvol*Glc/K2Glc/(1+Glc/K2Glc+(P2*Glc/K2Glc+1)/(P2*GlcX/K2Glc+1)*(1+GlcX/K2Glc)+G6P/K2IG6P+Glc*G6P/(K2Glc*K2IIG6P))
K6eq=0.081 milliMolar; ratio6=5.0 dimensionless; K6FBP=0.3 milliMolar; K6DHAP=2.0 milliMolar; K6GAP=4.0 milliMolar; K6IGAP=10.0 milliMolar; V6f=2207.82 mM per minuteReaction: FBP => GAP + DHAP, Rate Law: cytosol*(V6f*FBP/(K6FBP+FBP+GAP*K6DHAP*V6f/(K6eq*V6f*ratio6)+DHAP*K6GAP*V6f/(K6eq*V6f*ratio6)+FBP*GAP/K6IGAP+GAP*DHAP*V6f/(K6eq*V6f*ratio6))-V6f*GAP*DHAP/K6eq/(K6FBP+FBP+GAP*K6DHAP*V6f/(K6eq*V6f*ratio6)+DHAP*K6GAP*V6f/(K6eq*V6f*ratio6)+FBP*GAP/K6IGAP+GAP*DHAP*V6f/(K6eq*V6f*ratio6)))
K8NADH=0.06 milliMolar; K8GAP=0.6 milliMolar; V8r=833.858 mM per minute; K8NAD=0.1 milliMolar; V8f=833.858 mM per minute; K8eq=0.0055 dimensionless; K8BPG=0.01 milliMolarReaction: GAP + NAD => NADH + BPG, Rate Law: cytosol*(V8f*GAP*NAD/K8GAP/K8NAD/((1+GAP/K8GAP+BPG/K8BPG)*(1+NAD/K8NAD+NADH/K8NADH))-V8r*BPG*NADH/K8eq/K8GAP/K8NAD/((1+GAP/K8GAP+BPG/K8BPG)*(1+NAD/K8NAD+NADH/K8NADH)))
Yvol=59.0 dimensionless; k13=16.72 minute inverseReaction: EtOH => EtOHX, Rate Law: k13/Yvol*(cytosol*EtOH-extracellular*EtOHX)
V11m=53.1328 mM per minute; K11=0.3 milliMolarReaction: Pyr => ACA, Rate Law: cytosol*V11m*Pyr/(K11+Pyr)
k9r=1528.62 mM inverse min inverse; k9f=443866.0 mM inverse min inverseReaction: BPG + ADP => PEP + ATP, Rate Law: cytosol*(k9f*BPG*ADP-k9r*PEP*ATP)
K10PEP=0.2 milliMolar; V10m=343.096 mM per minute; K10ADP=0.17 milliMolarReaction: ADP + PEP => Pyr + ATP, Rate Law: cytosol*V10m*ADP*PEP/((K10PEP+PEP)*(K10ADP+ADP))
K7eq=0.055 dimensionless; V7r=116.365 mM per minute; K7DHAP=1.23 milliMolar; K7GAP=1.27 milliMolar; V7f=116.365 mM per minuteReaction: DHAP => GAP, Rate Law: cytosol*(V7f*DHAP/(K7DHAP+DHAP+K7DHAP/K7GAP*GAP)-V7r*GAP/K7eq/(K7DHAP+DHAP+K7DHAP/K7GAP*GAP))
k24r=133.333 mM inverse min inverse; k24f=432.9 mM inverse min inverseReaction: ATP + AMP => ADP, Rate Law: cytosol*(k24f*AMP*ATP-k24r*ADP^2)
k0=0.048 minute inverseReaction: GlycX => P, Rate Law: extracellular*k0*GlycX
k22=2.25932 mM inverse min inverseReaction: ATP + G6P => ADP, Rate Law: cytosol*k22*ATP*G6P
Yvol=59.0 dimensionless; k16=1.9 minute inverseReaction: Glyc => GlycX, Rate Law: k16/Yvol*(cytosol*Glyc-extracellular*GlycX)
k20=0.00283828 mM inverse min inverseReaction: CNX + ACAX => P, Rate Law: extracellular*k20*ACAX*CNX
K12ACA=0.71 milliMolar; V12m=89.8023 mM per minute; K12NADH=0.1 milliMolarReaction: NADH + ACA => NAD + EtOH, Rate Law: cytosol*V12m*ACA*NADH/((K12NADH+NADH)*(K12ACA+ACA))
K4F6P=0.15 milliMolar; V4f=496.042 mM per minute; V4r=496.042 mM per minute; K4G6P=0.8 milliMolar; K4eq=0.13 dimensionlessReaction: G6P => F6P, Rate Law: cytosol*(V4f*G6P/(K4G6P+G6P+K4G6P/K4F6P*F6P)-V4r*F6P/K4eq/(K4G6P+G6P+K4G6P/K4F6P*F6P))
k23=3.2076 minute inverseReaction: ATP => ADP, Rate Law: cytosol*k23*ATP
K5=0.021 mM squared; V5m=45.4327 mM per minute; kappa5=0.15 dimensionlessReaction: F6P + ATP => FBP + ADP; AMP, Rate Law: cytosol*V5m*F6P^2/(K5*(1+kappa5*ATP/AMP*ATP/AMP)+F6P^2)
K3DGlc=0.37 milliMolar; K3Glc=0.0 milliMolar; V3m=51.7547 mM per minute; K3ATP=0.1 milliMolarReaction: ATP + Glc => G6P + ADP, Rate Law: cytosol*V3m*ATP*Glc/(K3DGlc*K3ATP+K3Glc*ATP+K3ATP*Glc+Glc*ATP)
Yvol=59.0 dimensionless; k18=24.7 minute inverseReaction: ACA => ACAX, Rate Law: k18/Yvol*(cytosol*ACA-extracellular*ACAX)

States:

NameDescription
ATP[ATP; ATP]
PAMP
FBP[keto-D-fructose 1,6-bisphosphate; D-Fructose 1,6-bisphosphate]
CNX[cyanide; Cyanide ion]
ACA[acetaldehyde; Acetaldehyde]
AMP[AMP; AMP]
DHAP[dihydroxyacetone phosphate; Glycerone phosphate]
CNX0[cyanide; Cyanide ion]
GlcX0[glucose; C00293]
NADH[NADH; NADH]
GlycX[glycerol; Glycerol]
EtOH[ethanol; Ethanol]
Glyc[glycerol; Glycerol]
Pyr[pyruvate; Pyruvate; pyruvic acid]
F6P[CHEBI_20935; D-Fructose 6-phosphate]
BPG[3-phospho-D-glyceroyl dihydrogen phosphate; 3-Phospho-D-glyceroyl phosphate]
G6P[aldehydo-D-glucose 6-phosphate; D-Glucose 6-phosphate]
GAP[D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]
GlcX[glucose; C00293]
Glc[glucose; C00293]
PEP[Phosphoenolpyruvate; phosphoenolpyruvate; phosphoenolpyruvate]
NAD[NAD(+); NAD+]
EtOHX[ethanol; C000469]
ACAX[acetaldehyde; Acetaldehyde]
ADP[ADP; ADP]

I


Iancu2007_CardiacMyoscytes_cAMPsignaling: MODEL1006230117v0.0.1

This a model from the article: Compartmentation of cAMP signaling in cardiac myocytes: a computational study. Iancu…

Details

Receptor-mediated changes in cAMP production play an essential role in sympathetic and parasympathetic regulation of the electrical, mechanical, and metabolic activity of cardiac myocytes. However, responses to receptor activation cannot be easily ascribed to a uniform increase or decrease in cAMP activity throughout the entire cell. In this study, we used a computational approach to test the hypothesis that in cardiac ventricular myocytes the effects of beta(1)-adrenergic receptor (beta(1)AR) and M(2) muscarinic receptor (M(2)R) activation involve compartmentation of cAMP. A model consisting of two submembrane (caveolar and extracaveolar) microdomains and one bulk cytosolic domain was created using published information on the location of beta(1)ARs and M(2)Rs, as well as the location of stimulatory (G(s)) and inhibitory (G(i)) G-proteins, adenylyl cyclase isoforms inhibited (AC5/6) and stimulated (AC4/7) by G(i), and multiple phosphodiesterase isoforms (PDE2, PDE3, and PDE4). Results obtained with the model indicate that: 1), bulk basal cAMP can be high ( approximately 1 microM) and only modestly stimulated by beta(1)AR activation ( approximately 2 microM), but caveolar cAMP varies in a range more appropriate for regulation of protein kinase A ( approximately 100 nM to approximately 2 microM); 2), M(2)R activation strongly reduces the beta(1)AR-induced increases in caveolar cAMP, with less effect on bulk cAMP; and 3), during weak beta(1)AR stimulation, M(2)R activation not only reduces caveolar cAMP, but also produces a rebound increase in caveolar cAMP following termination of M(2)R activity. We conclude that compartmentation of cAMP can provide a quantitative explanation for several aspects of cardiac signaling. link: http://identifiers.org/pubmed/17293406

Iarosz2015 - brain tumor: BIOMD0000000775v0.0.1

The paper describes a model of brain tumor. Created by COPASI 4.25 (Build 207) This model is described in the arti…

Details

In recent years, it became clear that a better understanding of the interactions among the main elements involved in the cancer network is necessary for the treatment of cancer and the suppression of cancer growth. In this work we propose a system of coupled differential equations that model brain tumour under treatment by chemotherapy, which considers interactions among the glial cells, the glioma, the neurons, and the chemotherapeutic agents. We study the conditions for the glioma growth to be eliminated, and identify values of the parameters for which the inhibition of the glioma growth is obtained with a minimal loss of healthy cells. link: http://identifiers.org/pubmed/25596516

Parameters:

NameDescription
fi = 100.0 1/dReaction: => Q, Rate Law: tme*fi
b2 = 0.0018 1/dReaction: C => ; G, Rate Law: tme*b2*C*G
a = 2.0 1; gg = -1.1088E-4 1/d; H = 1.0 1Reaction: => N; G, Rate Law: tme*a*gg*H*N
o1 = 0.0068 1/dReaction: => G, Rate Law: tme*o1*G*(1-G)
b1 = 0.018 1/dReaction: G => ; C, Rate Law: tme*b1*G*C
p3 = 4.7E-8 1/d; a3 = 1.0 1Reaction: N => ; Q, Rate Law: tme*p3*N*Q/(a3+N)
a1 = 1.0 1; p1 = 4.7E-8 1/dReaction: G => ; Q, Rate Law: tme*p1*G*Q/(a1+G)
p2 = 4.7E-5 1/d; a2 = 1.0 1Reaction: C => ; Q, Rate Law: tme*p2*C*Q/(a2+C)
zeta = 0.2 1/dReaction: Q =>, Rate Law: tme*zeta*Q
o2 = 0.012 1/dReaction: => C, Rate Law: tme*o2*C*(1-C)

States:

NameDescription
Q[Chemotherapy]
C[glioma cell]
N[neuron]
G[glial cell]

Ibrahim2008 - Mitotic Spindle Assembly Checkpoint - Convey variant: BIOMD0000000187v0.0.1

Ibrahim2008 - Mitotic Spindle Assembly Checkpoint - Convey variantThe Mitotic Spindle Assembly Checkpoint ((M)SAC) is an…

Details

The Mitotic Spindle Assembly Checkpoint ((M)SAC) is an evolutionary conserved mechanism that ensures the correct segregation of chromosomes by restraining cell cycle progression from entering anaphase until all chromosomes have made proper bipolar attachments to the mitotic spindle. Its malfunction can lead to cancer.We have constructed and validated for the human (M)SAC mechanism an in silico dynamical model, integrating 11 proteins and complexes. The model incorporates the perspectives of three central control pathways, namely Mad1/Mad2 induced Cdc20 sequestering based on the Template Model, MCC formation, and APC inhibition. Originating from the biochemical reactions for the underlying molecular processes, non-linear ordinary differential equations for the concentrations of 11 proteins and complexes of the (M)SAC are derived. Most of the kinetic constants are taken from literature, the remaining four unknown parameters are derived by an evolutionary optimization procedure for an objective function describing the dynamics of the APC:Cdc20 complex. MCC:APC dissociation is described by two alternatives, namely the "Dissociation" and the "Convey" model variants. The attachment of the kinetochore to microtubuli is simulated by a switching parameter silencing those reactions which are stopped by the attachment. For both, the Dissociation and the Convey variants, we compare two different scenarios concerning the microtubule attachment dependent control of the dissociation reaction. Our model is validated by simulation of ten perturbation experiments.Only in the controlled case, our models show (M)SAC behaviour at meta- to anaphase transition in agreement with experimental observations. Our simulations revealed that for (M)SAC activation, Cdc20 is not fully sequestered; instead APC is inhibited by MCC binding. link: http://identifiers.org/pubmed/18253502

Parameters:

NameDescription
k5r = 0.2 per second; k5f = 10000.0 liter per mole per second; u = 1.0 dimensionlessReaction: Bub3_BubR1 + Cdc20 => Bub3_BubR1_Cdc20, Rate Law: Cytoplasm*(u*k5f*Bub3_BubR1*Cdc20-k5r*Bub3_BubR1_Cdc20)
k1f = 200000.0 liter per mole per second; k1r = 0.2 per second; u = 1.0 dimensionlessReaction: Mad1_CMad2 + OMad2 => Mad1_CMad2_OMad2, Rate Law: Cytoplasm*(u*k1f*Mad1_CMad2*OMad2-k1r*Mad1_CMad2_OMad2)
k3f = 0.01 per secondReaction: Cdc20_CMad2 => Cdc20 + OMad2, Rate Law: k3f*Cdc20_CMad2*Cytoplasm
kf6 = 1000.0 liter per mole per secondReaction: OMad2 + Cdc20 => Cdc20_CMad2, Rate Law: kf6*OMad2*Cdc20*Cytoplasm
k8r = 0.08 per second; k8f = 5000000.0 liter per mole per secondReaction: APC + Cdc20 => APC_Cdc20, Rate Law: Cytoplasm*(k8f*APC*Cdc20-k8r*APC_Cdc20)
k4r = 0.02 per second; u = 1.0 dimensionless; k4f = 1.0E7 liter per mole per secondReaction: Cdc20_CMad2 + Bub3_BubR1 => MCC, Rate Law: Cytoplasm*(u*k4f*Cdc20_CMad2*Bub3_BubR1-k4r*MCC)
k2f = 1.0E8 liter per mole per second; u = 1.0 dimensionlessReaction: Mad1_CMad2_OMad2 + Cdc20 => Mad1_CMad2 + Cdc20_CMad2, Rate Law: u*k2f*Mad1_CMad2_OMad2*Cdc20*Cytoplasm
u_prime = 0.0 dimensionless; k7bf = 0.08 per secondReaction: MCC_APC => OMad2 + Bub3_BubR1 + APC_Cdc20, Rate Law: u_prime*k7bf*MCC_APC*Cytoplasm
k7f = 1.0E8 liter per mole per second; u = 1.0 dimensionlessReaction: MCC + APC => MCC_APC, Rate Law: u*k7f*MCC*APC*Cytoplasm

States:

NameDescription
Bub3 BubR1 Cdc20[Cell division cycle protein 20 homolog; Vdx protein; Mitotic checkpoint protein BUB3]
Mad1 CMad2[Mitotic spindle assembly checkpoint protein MAD2A; Mitotic spindle assembly checkpoint protein MAD1; protein complex]
Bub3 BubR1[Vdx protein; Mitotic checkpoint protein BUB3]
APC Cdc20[Cell division cycle protein 20 homolog; Anaphase-promoting complex subunit CDC26; Anaphase-promoting complex subunit 13; Anaphase-promoting complex subunit 11; Anaphase-promoting complex subunit 10; Cell division cycle protein 23 homolog; Anaphase-promoting complex subunit 7; Cell division cycle protein 16 homolog; Anaphase-promoting complex subunit 5; Anaphase-promoting complex subunit 4; Cell division cycle protein 27 homolog; Anaphase-promoting complex subunit 2; Anaphase-promoting complex subunit 1]
MCC[Cell division cycle protein 20 homolog; Mitotic spindle assembly checkpoint protein MAD2A; Vdx protein; Mitotic checkpoint protein BUB3; mitotic checkpoint complex]
OMad2[Mitotic spindle assembly checkpoint protein MAD2A]
APC[anaphase-promoting complex; Anaphase-promoting complex subunit CDC26; Anaphase-promoting complex subunit 13; Anaphase-promoting complex subunit 11; Anaphase-promoting complex subunit 10; Cell division cycle protein 23 homolog; Anaphase-promoting complex subunit 7; Cell division cycle protein 16 homolog; Anaphase-promoting complex subunit 5; Anaphase-promoting complex subunit 4; Cell division cycle protein 27 homolog; Anaphase-promoting complex subunit 2; Anaphase-promoting complex subunit 1]
MCC APC[Cell division cycle protein 20 homolog; Mitotic spindle assembly checkpoint protein MAD2A; Vdx protein; Mitotic checkpoint protein BUB3; Anaphase-promoting complex subunit CDC26; Anaphase-promoting complex subunit 13; Anaphase-promoting complex subunit 11; Anaphase-promoting complex subunit 10; Cell division cycle protein 23 homolog; Anaphase-promoting complex subunit 7; Cell division cycle protein 16 homolog; Anaphase-promoting complex subunit 5; Anaphase-promoting complex subunit 4; Cell division cycle protein 27 homolog; Anaphase-promoting complex subunit 2; Anaphase-promoting complex subunit 1; anaphase-promoting complex; mitotic checkpoint complex]
Mad1 CMad2 OMad2[protein complex; Mitotic spindle assembly checkpoint protein MAD2A; Mitotic spindle assembly checkpoint protein MAD1]
Cdc20[Cell division cycle protein 20 homolog]
Cdc20 CMad2[Mitotic spindle assembly checkpoint protein MAD2A; Cell division cycle protein 20 homolog]

Ibrahim2008 - Mitotic Spindle Assembly Checkpoint - Dissociation variant: BIOMD0000000186v0.0.1

Ibrahim2008 - Mitotic Spindle Assembly Checkpoint - Dissociation variantThe Mitotic Spindle Assembly Checkpoint ((M)SAC)…

Details

The Mitotic Spindle Assembly Checkpoint ((M)SAC) is an evolutionary conserved mechanism that ensures the correct segregation of chromosomes by restraining cell cycle progression from entering anaphase until all chromosomes have made proper bipolar attachments to the mitotic spindle. Its malfunction can lead to cancer.We have constructed and validated for the human (M)SAC mechanism an in silico dynamical model, integrating 11 proteins and complexes. The model incorporates the perspectives of three central control pathways, namely Mad1/Mad2 induced Cdc20 sequestering based on the Template Model, MCC formation, and APC inhibition. Originating from the biochemical reactions for the underlying molecular processes, non-linear ordinary differential equations for the concentrations of 11 proteins and complexes of the (M)SAC are derived. Most of the kinetic constants are taken from literature, the remaining four unknown parameters are derived by an evolutionary optimization procedure for an objective function describing the dynamics of the APC:Cdc20 complex. MCC:APC dissociation is described by two alternatives, namely the "Dissociation" and the "Convey" model variants. The attachment of the kinetochore to microtubuli is simulated by a switching parameter silencing those reactions which are stopped by the attachment. For both, the Dissociation and the Convey variants, we compare two different scenarios concerning the microtubule attachment dependent control of the dissociation reaction. Our model is validated by simulation of ten perturbation experiments.Only in the controlled case, our models show (M)SAC behaviour at meta- to anaphase transition in agreement with experimental observations. Our simulations revealed that for (M)SAC activation, Cdc20 is not fully sequestered; instead APC is inhibited by MCC binding. link: http://identifiers.org/pubmed/18253502

Parameters:

NameDescription
k7f = 1.0E8 liter per mole per second; u = 1.0 dimensionlessReaction: MCC + APC => MCC_APC, Rate Law: u*k7f*MCC*APC*Cytoplasm
k1f = 200000.0 liter per mole per second; k1r = 0.2 per second; u = 1.0 dimensionlessReaction: Mad1_CMad2 + OMad2 => Mad1_CMad2_OMad2, Rate Law: Cytoplasm*(u*k1f*Mad1_CMad2*OMad2-k1r*Mad1_CMad2_OMad2)
k3f = 0.01 per secondReaction: Cdc20_CMad2 => Cdc20 + OMad2, Rate Law: k3f*Cdc20_CMad2*Cytoplasm
kf6 = 1000.0 liter per mole per secondReaction: OMad2 + Cdc20 => Cdc20_CMad2, Rate Law: kf6*OMad2*Cdc20*Cytoplasm
u_prime = 0.0 dimensionless; k7r = 0.08 per secondReaction: MCC_APC => MCC + APC, Rate Law: u_prime*k7r*MCC_APC*Cytoplasm
k8r = 0.08 per second; k8f = 5000000.0 liter per mole per secondReaction: APC + Cdc20 => APC_Cdc20, Rate Law: Cytoplasm*(k8f*APC*Cdc20-k8r*APC_Cdc20)
k2f = 1.0E8 liter per mole per second; u = 1.0 dimensionlessReaction: Mad1_CMad2_OMad2 + Cdc20 => Mad1_CMad2 + Cdc20_CMad2, Rate Law: u*k2f*Mad1_CMad2_OMad2*Cytoplasm*Cdc20
k4r = 0.02 per second; u = 1.0 dimensionless; k4f = 1.0E7 liter per mole per secondReaction: Cdc20_CMad2 + Bub3_BubR1 => MCC, Rate Law: Cytoplasm*(u*k4f*Cdc20_CMad2*Bub3_BubR1-k4r*MCC)
k5r = 0.2 per second; k5f = 10000.0 liter per mole per second; u = 1.0 dimensionlessReaction: Bub3_BubR1 + Cdc20 => Bub3_BubR1_Cdc20, Rate Law: Cytoplasm*(u*k5f*Bub3_BubR1*Cdc20-k5r*Bub3_BubR1_Cdc20)

States:

NameDescription
Bub3 BubR1 Cdc20[Cell division cycle protein 20 homolog; Vdx protein; Mitotic checkpoint protein BUB3]
Mad1 CMad2[protein complex; Mitotic spindle assembly checkpoint protein MAD2A; Mitotic spindle assembly checkpoint protein MAD1]
Bub3 BubR1[Vdx protein; Mitotic checkpoint protein BUB3]
APC Cdc20[Cell division cycle protein 20 homolog; Anaphase-promoting complex subunit CDC26; Anaphase-promoting complex subunit 13; Anaphase-promoting complex subunit 11; Anaphase-promoting complex subunit 10; Cell division cycle protein 23 homolog; Anaphase-promoting complex subunit 7; Cell division cycle protein 16 homolog; Anaphase-promoting complex subunit 5; Anaphase-promoting complex subunit 4; Cell division cycle protein 27 homolog; Anaphase-promoting complex subunit 2; Anaphase-promoting complex subunit 1]
MCC[Cell division cycle protein 20 homolog; Mitotic spindle assembly checkpoint protein MAD2A; Vdx protein; Mitotic checkpoint protein BUB3; mitotic checkpoint complex]
OMad2[Mitotic spindle assembly checkpoint protein MAD2A]
APC[anaphase-promoting complex; Anaphase-promoting complex subunit CDC26; Anaphase-promoting complex subunit 13; Anaphase-promoting complex subunit 11; Anaphase-promoting complex subunit 10; Cell division cycle protein 23 homolog; Anaphase-promoting complex subunit 7; Cell division cycle protein 16 homolog; Anaphase-promoting complex subunit 5; Anaphase-promoting complex subunit 4; Cell division cycle protein 27 homolog; Anaphase-promoting complex subunit 2; Anaphase-promoting complex subunit 1]
MCC APC[Cell division cycle protein 20 homolog; Mitotic spindle assembly checkpoint protein MAD2A; Vdx protein; Mitotic checkpoint protein BUB3; Anaphase-promoting complex subunit CDC26; Anaphase-promoting complex subunit 13; Anaphase-promoting complex subunit 11; Anaphase-promoting complex subunit 10; Cell division cycle protein 23 homolog; Anaphase-promoting complex subunit 7; Cell division cycle protein 16 homolog; Anaphase-promoting complex subunit 5; Anaphase-promoting complex subunit 4; Cell division cycle protein 27 homolog; Anaphase-promoting complex subunit 2; Anaphase-promoting complex subunit 1; anaphase-promoting complex; mitotic checkpoint complex]
Mad1 CMad2 OMad2[Mitotic spindle assembly checkpoint protein MAD2A; Mitotic spindle assembly checkpoint protein MAD1; protein complex]
Cdc20[Cell division cycle protein 20 homolog]
Cdc20 CMad2[Mitotic spindle assembly checkpoint protein MAD2A; Cell division cycle protein 20 homolog]

Ibrahim2008_Cdc20_Sequestring_Template_Model: BIOMD0000000194v0.0.1

*Biophysical Chemistry 134 (2008) 93-100*# Mad2 binding is not sufficient for complete Cdc20 sequestering in mitotic tra…

Details

For successful mitosis, metaphase has to be arrested until all centromeres are properly attached. The onset of anaphase, which is initiated by activating the APC, is controlled by the spindle assembly checkpoint (M)SAC. Mad2, which is a constitutive member of the (M)SAC, is supposed to inhibit the activity of the APC by sequestering away its co-activator Cdc20. Mad1 recruits Mad2 to unattached kinetochores and is compulsory for the establishment of the Mad2 and Cdc20 complexes. Recently, based on results from in vivo and in vitro studies, two biochemical models were proposed: the Template and the Exchange model. Here, we derive a mathematical description to compare the dynamical behaviour of the two models. Our simulation analysis supports the Template model. Using experimentally determined values for the model parameters, the Cdc20 concentration is reduced down to only about half. Thus, although the Template model displays good metaphase-to-anaphase switching behaviour, it is not able to completely describe (M)SAC regulation. This situation is neither improved by amplification nor by p31(comet) inhibition. We speculate that either additional reaction partners are required for total inhibition of Cdc20 or an extended mechanism has to be introduced for (M)SAC regulation. link: http://identifiers.org/pubmed/18295960

Parameters:

NameDescription
eta_T = 0.01 per secondReaction: Cdc20_CMad2 => Cdc20 + OMad2, Rate Law: Cytoplasm*eta_T*Cdc20_CMad2
beta_T = 0.2 per second; alpha_T = 200000.0 liter per mole per second; u = 1.0 dimensionlessReaction: Mad1_CMad2 + OMad2 => Mad1_CMad2_OMad2, Rate Law: Cytoplasm*(u*alpha_T*Mad1_CMad2*OMad2-beta_T*Mad1_CMad2_OMad2)
gamma_T = 1.0E9 liter per mole per second; u = 1.0 dimensionlessReaction: Mad1_CMad2_OMad2 + Cdc20 => Mad1_CMad2 + Cdc20_CMad2, Rate Law: Cytoplasm*u*gamma_T*Mad1_CMad2_OMad2*Cdc20

States:

NameDescription
Mad1 CMad2[Mitotic spindle assembly checkpoint protein MAD1; Mitotic spindle assembly checkpoint protein MAD2A]
OMad2[Mitotic spindle assembly checkpoint protein MAD2A]
Cdc20[Cell division cycle protein 20 homolog]
Mad1 CMad2 OMad2[Mitotic spindle assembly checkpoint protein MAD1; Mitotic spindle assembly checkpoint protein MAD2A]
Cdc20 CMad2[Cell division cycle protein 20 homolog; Mitotic spindle assembly checkpoint protein MAD2A]

Ibrahim2008_MCC_assembly_model_KDM: BIOMD0000000193v0.0.1

*BioSystems (2007), doi:10.1016/j.biosystems.2008.06.007*# *In-silico* study of kinetochore control, amplification, and…

Details

Eukaryotic cells rely on a surveillance mechanism, the "Spindle Assembly Checkpoint"SACM in order to ensure accurate chromosome segregation by preventing anaphase initiation until all chromosomes are correctly attached to the mitotic spindle. In different organisms, a mitotic checkpoint complex (MCC) composed of Mad2, Bub3, BubR1/Mad3, and Cdc20 inhibits the anaphase promoting complex (APC/C) to initiate promotion into anaphase. The mechanism of MCC formation and its regulation by the kinetochore are unclear. Here, we constructed dynamical models of MCC formation involving different kinetochore control mechanisms including amplification as well as inhibition effects, and analysed their quantitative properties. In particular, in this system, fast and stable metaphase to anaphase transition can only be triggered when the kinetochore controls the Bub3:BubR1-related reactions; signal amplification and inhibition play a subordinate role. Furthermore, when introducing experimentally determined parameter values into the models analysed here, we found that effective MCC formation is not combined with complete Cdc20 sequestering. Instead, the MCC might bind and completely block the APC/C. The SACM might function by an MCC:APC/C complex rearrangement. link: http://identifiers.org/pubmed/18675311

Parameters:

NameDescription
k1f = 200000.0 liter per mole per second; k1r = 0.2 per second; u = 1.0 dimensionlessReaction: Mad1_CMad2 + OMad2 => Mad1_CMad2_OMad2, Rate Law: Cytoplasm*(u*k1f*Mad1_CMad2*OMad2-k1r*Mad1_CMad2_OMad2)
k3f = 0.01 per secondReaction: Cdc20_CMad2 => Cdc20 + OMad2, Rate Law: Cytoplasm*k3f*Cdc20_CMad2
k2f = 1.0E7 liter per mole per second; u = 1.0 dimensionlessReaction: Mad1_CMad2_OMad2 + Cdc20 => Mad1_CMad2 + Cdc20_CMad2, Rate Law: Cytoplasm*u*k2f*Mad1_CMad2_OMad2*Cdc20
kf6 = 1000.0 liter per mole per secondReaction: OMad2 + Cdc20 => Cdc20_CMad2, Rate Law: Cytoplasm*kf6*OMad2*Cdc20
k4r = 0.02 per second; u = 1.0 dimensionless; k4f = 1.0E7 liter per mole per secondReaction: Cdc20_CMad2 + Bub3_BubR1 => MCC, Rate Law: Cytoplasm*(u*k4f*Cdc20_CMad2*Bub3_BubR1-k4r*MCC)
k5r = 0.2 per second; k5f = 10000.0 liter per mole per second; u = 1.0 dimensionlessReaction: Bub3_BubR1 + Cdc20 => Bub3_BubR1_Cdc20, Rate Law: Cytoplasm*(u*k5f*Bub3_BubR1*Cdc20-k5r*Bub3_BubR1_Cdc20)

States:

NameDescription
Cdc20 CMad2[Mitotic spindle assembly checkpoint protein MAD2A; Cell division cycle protein 20 homolog]
Bub3 BubR1 Cdc20[Cell division cycle protein 20 homolog; Mitotic checkpoint serine/threonine-protein kinase BUB1 beta; Mitotic checkpoint protein BUB3]
Mad1 CMad2[Mitotic spindle assembly checkpoint protein MAD2A; Mitotic spindle assembly checkpoint protein MAD1]
OMad2[Mitotic spindle assembly checkpoint protein MAD2A]
Bub3 BubR1[Mitotic checkpoint protein BUB3; Mitotic checkpoint serine/threonine-protein kinase BUB1 beta]
Cdc20[Cell division cycle protein 20 homolog]
Mad1 CMad2 OMad2[Mitotic spindle assembly checkpoint protein MAD2A; Mitotic spindle assembly checkpoint protein MAD1]
MCC[mitotic checkpoint complex; Mitotic checkpoint serine/threonine-protein kinase BUB1 beta; Mitotic spindle assembly checkpoint protein MAD2A; Cell division cycle protein 20 homolog; Mitotic checkpoint protein BUB3]

Ihekwaba2004_NFkB_Sensitivity: BIOMD0000000230v0.0.1

This a model from the article: Sensitivity analysis of parameters controlling oscillatory signalling in the NF-kappaB…

Details

Analysis of cellular signalling interactions is expected to create an enormous informatics challenge, perhaps even greater than that of analysing the genome. A key step in the evolution towards a more quantitative understanding of signalling is to specify explicitly the kinetics of all chemical reaction steps in a pathway. We have reconstructed a model of the nuclear factor, kappaB (NF-kappaB) signalling pathway, containing 64 parameters and 26 variables, including steps in which the activation of the NF-kappaB transcription factor is intimately associated with the phosphorylation and ubiquitination of its inhibitor kappaB by a membrane-associated kinase, and its translocation from the cytoplasm to the nucleus. We apply sensitivity analysis to the model. This identifies those parameters in this (IkappaB)/NF-kappaB signalling system (containing only induced IkappaBalpha isoform) that most affect the oscillatory concentration of nuclear NF-kappaB (in terms of both period and amplitude). The intention is to provide guidance on which proteins are likely to be most significant as drug targets or should be exploited for further, more detailed experiments. The sensitivity coefficients were found to be strongly dependent upon the magnitude of the parameter change studied, indicating the highly non-linear nature of the system. Of the 64 parameters in the model, only eight to nine exerted a major control on nuclear NF-kappaB oscillations, and each of these involved as reaction participants either the IkappaB kinase (IKK) or IkappaBalpha, directly. This means that the dominant dynamics of the pathway can be reflected, in addition to that of nuclear NF-kappaB itself, by just two of the other pathway variables. This is conveniently observed in a phase-plane plot. link: http://identifiers.org/pubmed/17052119

Parameters:

NameDescription
k1 = 0.5Reaction: IkBa + NFkB => IkBaNFkB, Rate Law: k1*IkBa*NFkB
k29 = 2.8E-4Reaction: IkBat => sink, Rate Law: k29*IkBat
k16 = 2.25E-5Reaction: IkBaNFkB => NFkB, Rate Law: k16*IkBaNFkB
k7 = 0.5Reaction: NFkB + IKKIkBa => IKKIkBaNFkB, Rate Law: k7*IKKIkBa*NFkB
k39 = 2.0E-4Reaction: IkBan => IkBa, Rate Law: k39*IkBan
k14 = 5.0E-4Reaction: IKKIkBeNFkB => NFkB + IKKIkBe, Rate Law: k14*IKKIkBeNFkB
k52 = 0.185Reaction: IkBaNFkB + IKK => IKKIkBaNFkB, Rate Law: k52*IKK*IkBaNFkB
k6 = 5.0E-4Reaction: IkBeNFkB => NFkB + IkBe, Rate Law: k6*IkBeNFkB
k55 = 0.048Reaction: IkBbNFkB + IKK => IKKIkBbNFkB, Rate Law: k55*IKK*IkBbNFkB
k57 = 0.0052Reaction: IkBbnNFkBn => IkBbNFkB, Rate Law: k57*IkBbnNFkBn
k2 = 5.0E-4Reaction: IkBaNFkB => IkBa + NFkB, Rate Law: k2*IkBaNFkB
k48 = 0.00408Reaction: source => IkBe + sink; IkBet, Rate Law: k48*IkBet
k61 = 1.2E-4Reaction: IKK => sink, Rate Law: k61*IKK
k53 = 0.00125Reaction: IKKIkBaNFkB => IkBaNFkB + IKK, Rate Law: k53*IKKIkBaNFkB
k41 = 0.00175Reaction: IKKIkBb => IKK + IkBb, Rate Law: k41*IKKIkBb
k4 = 5.0E-4Reaction: IkBbNFkB => IkBb + NFkB, Rate Law: k4*IkBbNFkB
k47 = 0.00175Reaction: IKKIkBe => IkBe + IKK, Rate Law: k47*IKKIkBe
k63 = 0.0015Reaction: IKKIkBb => IKK, Rate Law: k63*IKKIkBb
k60 = 0.0052Reaction: IkBenNFkBn => IkBeNFkB, Rate Law: k60*IkBenNFkBn
k50 = 1.5E-4Reaction: IkBe => IkBen, Rate Law: k50*IkBe
k64 = 0.0022Reaction: IKKIkBe => IKK, Rate Law: k64*IKKIkBe
k54 = 0.0138Reaction: IkBanNFkBn => IkBaNFkB, Rate Law: k54*IkBanNFkBn
k12 = 0.0075Reaction: IKKIkBbNFkB => IKK + NFkB, Rate Law: k12*IKKIkBbNFkB
k49 = 1.13E-4Reaction: IkBe => sink, Rate Law: k49*IkBe
k30 = 1.78E-7Reaction: source => IkBbt, Rate Law: k30*source
k38 = 3.0E-4Reaction: IkBa => IkBan, Rate Law: k38*IkBa
k51 = 1.0E-4Reaction: IkBen => IkBe, Rate Law: k51*IkBen
k20 = 8.0E-5Reaction: NFkBn => NFkB, Rate Law: k20*NFkBn
k36 = 0.00408Reaction: source => IkBa + sink; IkBat, Rate Law: k36*IkBat
k28 = 0.0165Reaction: source => IkBat + sink; NFkBn, Rate Law: k28*NFkBn*NFkBn
k40 = 0.006Reaction: IkBb + IKK => IKKIkBb, Rate Law: k40*IKK*IkBb
k33 = 2.8E-4Reaction: IkBet => sink, Rate Law: k33*IkBet
k35 = 0.00125Reaction: IKKIkBa => IKK + IkBa, Rate Law: k35*IKKIkBa
k9 = 0.0204Reaction: IKKIkBaNFkB => NFkB + IKK, Rate Law: k9*IKKIkBaNFkB
k37 = 1.13E-4Reaction: IkBa => sink, Rate Law: k37*IkBa
k56 = 0.00175Reaction: IKKIkBbNFkB => IkBbNFkB + IKK, Rate Law: k56*IKKIkBbNFkB
k34 = 0.0225Reaction: IKK + IkBa => IKKIkBa, Rate Law: k34*IKK*IkBa
k58 = 0.07Reaction: IKK + IkBeNFkB => IKKIkBeNFkB, Rate Law: k58*IKK*IkBeNFkB
k32 = 1.27E-7Reaction: source => IkBet, Rate Law: k32*source
k23 = 0.5Reaction: IkBbn + NFkBn => IkBbnNFkBn, Rate Law: k23*IkBbn*NFkBn
k59 = 0.00175Reaction: IKKIkBeNFkB => IKK + IkBeNFkB, Rate Law: k59*IKKIkBeNFkB
k18 = 2.25E-5Reaction: IkBeNFkB => NFkB, Rate Law: k18*IkBeNFkB
k31 = 2.8E-4Reaction: IkBbt => sink, Rate Law: k31*IkBbt
k27 = 1.54E-6Reaction: source => IkBat, Rate Law: k27*source
k17 = 2.25E-5Reaction: IkBbNFkB => NFkB, Rate Law: k17*IkBbNFkB
k46 = 0.009Reaction: IkBe + IKK => IKKIkBe, Rate Law: k46*IKK*IkBe
k3 = 0.5Reaction: IkBb + NFkB => IkBbNFkB, Rate Law: k3*IkBb*NFkB
k25 = 0.5Reaction: NFkBn + IkBen => IkBenNFkBn, Rate Law: k25*IkBen*NFkBn
k42 = 0.00408Reaction: source => IkBb + sink; IkBbt, Rate Law: k42*IkBbt
k8 = 5.0E-4Reaction: IKKIkBaNFkB => NFkB + IKKIkBa, Rate Law: k8*IKKIkBaNFkB
k13 = 0.5Reaction: NFkB + IKKIkBe => IKKIkBeNFkB, Rate Law: k13*IKKIkBe*NFkB
k10 = 0.5Reaction: IKKIkBb + NFkB => IKKIkBbNFkB, Rate Law: k10*IKKIkBb*NFkB
k21 = 0.5Reaction: NFkBn + IkBan => IkBanNFkBn, Rate Law: k21*IkBan*NFkBn
k44 = 1.5E-4Reaction: IkBb => IkBbn, Rate Law: k44*IkBb
k11 = 5.0E-4Reaction: IKKIkBbNFkB => IKKIkBb + NFkB, Rate Law: k11*IKKIkBbNFkB
k15 = 0.011Reaction: IKKIkBeNFkB => NFkB + IKK, Rate Law: k15*IKKIkBeNFkB
k45 = 1.0E-4Reaction: IkBbn => IkBb, Rate Law: k45*IkBbn
k5 = 0.5Reaction: NFkB + IkBe => IkBeNFkB, Rate Law: k5*IkBe*NFkB
k62 = 0.00407Reaction: IKKIkBa => IKK, Rate Law: k62*IKKIkBa

States:

NameDescription
NFkBn[Nuclear factor NF-kappa-B p105 subunit]
sinksink
IkBa[NF-kappa-B inhibitor alpha]
IkBeNFkB[NF-kappa-B inhibitor epsilon; Nuclear factor NF-kappa-B p105 subunit]
IkBb[NF-kappa-B inhibitor beta]
IKKIkBaNFkB[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor alpha; Nuclear factor NF-kappa-B p105 subunit]
IkBaNFkB[NF-kappa-B inhibitor alpha; Nuclear factor NF-kappa-B p105 subunit]
IkBbNFkB[NF-kappa-B inhibitor beta; Nuclear factor NF-kappa-B p105 subunit]
IKK[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha]
IKKIkBb[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor beta]
IKKIkBa[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor alpha]
IKKIkBeNFkB[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor epsilon; Nuclear factor NF-kappa-B p105 subunit]
sourcesource
NFkB[Nuclear factor NF-kappa-B p105 subunit]
IkBe[NF-kappa-B inhibitor epsilon]
IKKIkBbNFkB[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor beta; Nuclear factor NF-kappa-B p105 subunit]
IkBatIkBat

Imam2011_RhodobacterSphaeroides_MetabolicNetwork: MODEL1106220000v0.0.1

This model is from the article: iRsp1095: A genome-scale reconstruction of the Rhodobacter sphaeroides metabolic netwo…

Details

BACKGROUND: Rhodobacter sphaeroides is one of the best studied purple non-sulfur photosynthetic bacteria and serves as an excellent model for the study of photosynthesis and the metabolic capabilities of this and related facultative organisms. The ability of R. sphaeroides to produce hydrogen (H₂), polyhydroxybutyrate (PHB) or other hydrocarbons, as well as its ability to utilize atmospheric carbon dioxide (CO₂) as a carbon source under defined conditions, make it an excellent candidate for use in a wide variety of biotechnological applications. A genome-level understanding of its metabolic capabilities should help realize this biotechnological potential. RESULTS: Here we present a genome-scale metabolic network model for R. sphaeroides strain 2.4.1, designated iRsp1095, consisting of 1,095 genes, 796 metabolites and 1158 reactions, including R. sphaeroides-specific biomass reactions developed in this study. Constraint-based analysis showed that iRsp1095 agreed well with experimental observations when modeling growth under respiratory and phototrophic conditions. Genes essential for phototrophic growth were predicted by single gene deletion analysis. During pathway-level analyses of R. sphaeroides metabolism, an alternative route for CO₂ assimilation was identified. Evaluation of photoheterotrophic H2 production using iRsp1095 indicated that maximal yield would be obtained from growing cells, with this predicted maximum ~50% higher than that observed experimentally from wild type cells. Competing pathways that might prevent the achievement of this theoretical maximum were identified to guide future genetic studies. CONCLUSIONS: iRsp1095 provides a robust framework for future metabolic engineering efforts to optimize the solar- and nutrient-powered production of biofuels and other valuable products by R. sphaeroides and closely related organisms. link: http://identifiers.org/pubmed/21777427

Imam2013 - Metabolic network in Rhodobacter sphaeroides (iRsp1140): MODEL1304240000v0.0.1

Rhodobacter sphaeroides model (Version 2 - iRsp1140)

Details

BACKGROUND: Improving our understanding of processes at the core of cellular lifestyles can be aided by combining information from genetic analyses, high-throughput experiments and computational predictions. RESULTS: We combined data and predictions derived from phenotypic, physiological, genetic and computational analyses to dissect the metabolic and energetic networks of the facultative photosynthetic bacterium Rhodobacter sphaeroides. We focused our analysis on pathways crucial to the production and recycling of pyridine nucleotides during aerobic respiratory and anaerobic photosynthetic growth in the presence of an organic electron donor. In particular, we assessed the requirement for NADH/NADPH transhydrogenase enzyme, PntAB during respiratory and photosynthetic growth. Using high-throughput phenotype microarrays (PMs), we found that PntAB is essential for photosynthetic growth in the presence of many organic electron donors, particularly those predicted to require its activity to produce NADPH. Utilizing the genome-scale metabolic model iRsp1095, we predicted alternative routes of NADPH synthesis and used gene expression analyses to show that transcripts from a subset of the corresponding genes were conditionally increased in a ΔpntAB mutant. We then used a combination of metabolic flux predictions and mutational analysis to identify flux redistribution patterns utilized in the ΔpntAB mutant to compensate for the loss of this enzyme. Data generated from metabolic and phenotypic analyses of wild type and mutant cells were used to develop iRsp1140, an expanded genome-scale metabolic reconstruction for R. sphaeroides with improved ability to analyze and predict pathways associated with photosynthesis and other metabolic processes. CONCLUSIONS: These analyses increased our understanding of key aspects of the photosynthetic lifestyle, highlighting the added importance of NADPH production under these conditions. It also led to a significant improvement in the predictive capabilities of a metabolic model for the different energetic lifestyles of a facultative organism. link: http://identifiers.org/pubmed/24034347

Inada2009_AtrioventricularNode_AtrioNodalCell: MODEL1006230082v0.0.1

This a model from the article: One-dimensional mathematical model of the atrioventricular node including atrio-nodal,…

Details

Mathematical models are a repository of knowledge as well as research and teaching tools. Although action potential models have been developed for most regions of the heart, there is no model for the atrioventricular node (AVN). We have developed action potential models for single atrio-nodal, nodal, and nodal-His cells. The models have the same action potential shapes and refractoriness as observed in experiments. Using these models, together with models for the sinoatrial node (SAN) and atrial muscle, we have developed a one-dimensional (1D) multicellular model including the SAN and AVN. The multicellular model has slow and fast pathways into the AVN and using it we have analyzed the rich behavior of the AVN. Under normal conditions, action potentials were initiated in the SAN center and then propagated through the atrium and AVN. The relationship between the AVN conduction time and the timing of a premature stimulus (conduction curve) is consistent with experimental data. After premature stimulation, atrioventricular nodal reentry could occur. After slow pathway ablation or block of the L-type Ca(2+) current, atrioventricular nodal reentry was abolished. During atrial fibrillation, the AVN limited the number of action potentials transmitted to the ventricle. In the absence of SAN pacemaking, the inferior nodal extension acted as the pacemaker. In conclusion, we have developed what we believe is the first detailed mathematical model of the AVN and it shows the typical physiological and pathophysiological characteristics of the tissue. The model can be used as a tool to analyze the complex structure and behavior of the AVN. link: http://identifiers.org/pubmed/19843444

Inada2009_AtrioventricularNode_NodalCell: MODEL1006230088v0.0.1

This a model from the article: One-dimensional mathematical model of the atrioventricular node including atrio-nodal,…

Details

Mathematical models are a repository of knowledge as well as research and teaching tools. Although action potential models have been developed for most regions of the heart, there is no model for the atrioventricular node (AVN). We have developed action potential models for single atrio-nodal, nodal, and nodal-His cells. The models have the same action potential shapes and refractoriness as observed in experiments. Using these models, together with models for the sinoatrial node (SAN) and atrial muscle, we have developed a one-dimensional (1D) multicellular model including the SAN and AVN. The multicellular model has slow and fast pathways into the AVN and using it we have analyzed the rich behavior of the AVN. Under normal conditions, action potentials were initiated in the SAN center and then propagated through the atrium and AVN. The relationship between the AVN conduction time and the timing of a premature stimulus (conduction curve) is consistent with experimental data. After premature stimulation, atrioventricular nodal reentry could occur. After slow pathway ablation or block of the L-type Ca(2+) current, atrioventricular nodal reentry was abolished. During atrial fibrillation, the AVN limited the number of action potentials transmitted to the ventricle. In the absence of SAN pacemaking, the inferior nodal extension acted as the pacemaker. In conclusion, we have developed what we believe is the first detailed mathematical model of the AVN and it shows the typical physiological and pathophysiological characteristics of the tissue. The model can be used as a tool to analyze the complex structure and behavior of the AVN. link: http://identifiers.org/pubmed/19843444

Inada2009_AtrioventricularNode_NodelHisCell: MODEL1006230008v0.0.1

This a model from the article: One-dimensional mathematical model of the atrioventricular node including atrio-nodal,…

Details

Mathematical models are a repository of knowledge as well as research and teaching tools. Although action potential models have been developed for most regions of the heart, there is no model for the atrioventricular node (AVN). We have developed action potential models for single atrio-nodal, nodal, and nodal-His cells. The models have the same action potential shapes and refractoriness as observed in experiments. Using these models, together with models for the sinoatrial node (SAN) and atrial muscle, we have developed a one-dimensional (1D) multicellular model including the SAN and AVN. The multicellular model has slow and fast pathways into the AVN and using it we have analyzed the rich behavior of the AVN. Under normal conditions, action potentials were initiated in the SAN center and then propagated through the atrium and AVN. The relationship between the AVN conduction time and the timing of a premature stimulus (conduction curve) is consistent with experimental data. After premature stimulation, atrioventricular nodal reentry could occur. After slow pathway ablation or block of the L-type Ca(2+) current, atrioventricular nodal reentry was abolished. During atrial fibrillation, the AVN limited the number of action potentials transmitted to the ventricle. In the absence of SAN pacemaking, the inferior nodal extension acted as the pacemaker. In conclusion, we have developed what we believe is the first detailed mathematical model of the AVN and it shows the typical physiological and pathophysiological characteristics of the tissue. The model can be used as a tool to analyze the complex structure and behavior of the AVN. link: http://identifiers.org/pubmed/19843444

Intosalmi2015 - Th17 core network model: BIOMD0000001004v0.0.1

a dynamic description for the core molecular mechanisms steering Th17 cell differentiation and use mathematical modeling…

Details

Background

The differentiation of naive CD 4(+) helper T (Th) cells into effector Th17 cells is steered by extracellular cytokines that activate and control the lineage specific transcriptional program. While the inducing cytokine signals and core transcription factors driving the differentiation towards Th17 lineage are well known, detailed mechanistic interactions between the key components are poorly understood.#### Results We develop an integrative modeling framework which combines RNA sequencing data with mathematical modeling and enables us to construct a mechanistic model for the core Th17 regulatory network in a data-driven manner.#### Conclusions Our results show significant evidence, for instance, for inhibitory mechanisms between the transcription factors and reveal a previously unknown dependency between the dosage of the inducing cytokine TGF b and the expression of the master regulator of competing (induced) regulatory T cell lineage. Further, our experimental validation approves this dependency in Th17 polarizing conditions. link: http://identifiers.org/pubmed/26578352

Invergo2014 - Phototransduction cascade in mouse rod cells: BIOMD0000000578v0.0.1

Invergo2014 - Phototransduction cascade in mouse rod cellsThis model is described in the article: [A comprehensive mode…

Details

Vertebrate visual phototransduction is perhaps the most well-studied G-protein signaling pathway. A wealth of available biochemical and electrophysiological data has resulted in a rich history of mathematical modeling of the system. However, while the most comprehensive models have relied upon amphibian biochemical and electrophysiological data, modern research typically employs mammalian species, particularly mice, which exhibit significantly faster signaling dynamics. In this work, we present an adaptation of a previously published, comprehensive model of amphibian phototransduction that can produce quantitatively accurate simulations of the murine photoresponse. We demonstrate the ability of the model to predict responses to a wide range of stimuli and under a variety of mutant conditions. Finally, we employ the model to highlight a likely unknown mechanism related to the interaction between rhodopsin and rhodopsin kinase. link: http://identifiers.org/pubmed/24675755

Parameters:

NameDescription
kG2 = 2200.0; kG1_3 = 0.0Reaction: Gt + R3 => R3_Gt, Rate Law: kG1_3*Gt*R3-kG2*R3_Gt
kRK3_ATP = 4000.0Reaction: R1_RKpre => R2_RKpost, Rate Law: kRK3_ATP*R1_RKpre
kRK2 = 250.0; kRK1_5 = 0.0Reaction: R5 + RK => R5_RKpre, Rate Law: kRK1_5*RK*R5-kRK2*R5_RKpre
E = 0.0; betadark = 3.19; betasub = 0.0021826Reaction: cGMP => ; Ga_GTP_PDE_a_Ga_GTP, Ga_GTP_a_PDE_a_Ga_GTP, PDE_a_Ga_GTP, Rate Law: (betadark+betasub*E)*cGMP
kA1_3 = 0.0; kA2 = 0.026Reaction: Arr + R3 => R3_Arr, Rate Law: kA1_3*Arr*R3-kA2*R3_Arr
kP1_rev = 0.0; kP1 = 0.05497Reaction: Ga_GTP + PDE => PDE_Ga_GTP, Rate Law: kP1*PDE*Ga_GTP-kP1_rev*PDE_Ga_GTP
kRGS1 = 4.8182E-5Reaction: Ga_GTP_a_PDE_a_Ga_GTP + RGS => RGS_Ga_GTP_a_PDE_a_Ga_GTP, Rate Law: kRGS1*RGS*Ga_GTP_a_PDE_a_Ga_GTP
kA3 = 1.1651Reaction: R1_Arr => Arr + Ops, Rate Law: kA3*R1_Arr
kA1_4 = 0.0; kA2 = 0.026Reaction: Arr + R4 => R4_Arr, Rate Law: kA1_4*Arr*R4-kA2*R4_Arr
kRK4 = 250.0Reaction: R1_RKpost => R1 + RK, Rate Law: kRK4*R1_RKpost
kG2 = 2200.0; kG1_2 = 0.0Reaction: Gt + R2 => R2_Gt, Rate Law: kG1_2*Gt*R2-kG2*R2_Gt
kA1_1 = 0.0; kA2 = 0.026Reaction: Arr + R1 => R1_Arr, Rate Law: kA1_1*Arr*R1-kA2*R1_Arr
kRec4 = 0.610084; kRec3 = 4.1081E-4Reaction: RK + RecR_Ca => RecR_Ca_RK, Rate Law: kRec3*RecR_Ca*RK-kRec4*RecR_Ca_RK
kA1_5 = 0.0; kA2 = 0.026Reaction: Arr + R5 => R5_Arr, Rate Law: kA1_5*Arr*R5-kA2*R5_Arr
kRK2 = 250.0; kRK1_4 = 0.0Reaction: R4 + RK => R4_RKpre, Rate Law: kRK1_4*RK*R4-kRK2*R4_RKpre
kG1_1 = 0.0; kG2 = 2200.0Reaction: Gt + R1 => R1_Gt, Rate Law: kG1_1*Gt*R1-kG2*R1_Gt
kRGS2 = 98.0Reaction: RGS_Ga_GTP_a_PDE_a_Ga_GTP => Ga_GDP + PDE_a_Ga_GTP + RGS, Rate Law: kRGS2*RGS_Ga_GTP_a_PDE_a_Ga_GTP
kG6 = 8500.0Reaction: R2_G_GTP => G_GTP + R2, Rate Law: kG6*R2_G_GTP
kRK1_3 = 0.0; kRK2 = 250.0Reaction: R3 + RK => R3_RKpre, Rate Law: kRK1_3*RK*R3-kRK2*R3_RKpre
kG3 = 8500.0; kG4_GDP = 400.0Reaction: R1_Gt => R1_G, Rate Law: kG3*R1_Gt-kG4_GDP*R1_G
kRK2 = 250.0; kRK1_0 = 0.1724Reaction: R0 + RK => R0_RKpre, Rate Law: kRK1_0*RK*R0-kRK2*R0_RKpre
kG5_GTP = 3500.0Reaction: R1_G => R1_G_GTP, Rate Law: kG5_GTP*R1_G
kRec1 = 0.011; kRec2 = 0.05Reaction: RecT + Ca2_free => RecR_Ca, Rate Law: kRec1*RecT*Ca2_free-kRec2*RecR_Ca
m1 = 3.0; Kc1 = 0.171; Kc2 = 0.059; m2 = 1.5; alfamax = 60.0Reaction: => cGMP; Ca2_free, Rate Law: alfamax/(1+(Ca2_free/Kc1)^m1)+alfamax/(1+(Ca2_free/Kc2)^m2)
kP2 = 940.7Reaction: PDE_Ga_GTP => PDE_a_Ga_GTP, Rate Law: kP2*PDE_Ga_GTP
ktherm = 0.0238Reaction: R1 => Ops, Rate Law: ktherm*R1
kRK2 = 250.0; kRK1_1 = 0.0Reaction: R1 + RK => R1_RKpre, Rate Law: kRK1_1*RK*R1-kRK2*R1_RKpre
kA1_6 = 0.0; kA2 = 0.026Reaction: Arr + R6 => R6_Arr, Rate Law: kA1_6*Arr*R6-kA2*R6_Arr
kRK2 = 250.0; kRK1_6 = 0.0Reaction: R6 + RK => R6_RKpre, Rate Law: kRK1_6*RK*R6-kRK2*R6_RKpre
kRK2 = 250.0; kRK1_2 = 0.0Reaction: R2 + RK => R2_RKpre, Rate Law: kRK1_2*RK*R2-kRK2*R2_RKpre
kA1_2 = 0.0; kA2 = 0.026Reaction: Arr + R2 => R2_Arr, Rate Law: kA1_2*Arr*R2-kA2*R2_Arr
kG2 = 2200.0; kG1_6 = 0.0Reaction: Gt + R6 => R6_Gt, Rate Law: kG1_6*Gt*R6-kG2*R6_Gt
stimulus = 0.0; Rtot = 1.0E8Reaction: R_Gt => R0_Gt, Rate Law: stimulus*R_Gt/Rtot

States:

NameDescription
RGS Ga GTP a PDE a Ga GTPRGS_Ga_GTP_a_PDE_a_Ga_GTP
R0 RKpreR0_RKpre
R5 RKpreR5_RKpre
R1 GR1_G
R5 RKpostR5_RKpost
RGSRGS
cGMPcGMP
R3 GR3_G
R6 GR6_G
PDE a Ga GTPPDE_a_Ga_GTP
R1 G GTPR1_G_GTP
R6 ArrR6_Arr
R6 GtR6_Gt
R3 RKpreR3_RKpre
R2 ArrR2_Arr
R6 RKpostR6_RKpost
R1 RKpreR1_RKpre
R2 RKpostR2_RKpost
R1 RKpostR1_RKpost
R4R4
RKRK
R1R1
R1 ArrR1_Arr
R6 G GTPR6_G_GTP
R6 RKpreR6_RKpre
R GtR_Gt
R3 G GTPR3_G_GTP
RecTRecT
RGS PDE a Ga GTPRGS_PDE_a_Ga_GTP
ArrArr
PDE Ga GTPPDE_Ga_GTP
R1 GtR1_Gt
R3 RKpostR3_RKpost
R3R3
R6R6
R2 GtR2_Gt
R3 ArrR3_Arr
R3 GtR3_Gt
R2R2
R2 G GTPR2_G_GTP

Irani2015 - Genome-scale metabolic model of P.pastoris N-glycosylation: MODEL1510220000v0.0.1

Irani2015 - Genome-scale metabolic model of P.pastoris N-glycosylationThis model is described in the article: [Genome-s…

Details

Pichia pastoris is used for commercial production of human therapeutic proteins, and genome-scale models of P. pastoris metabolism have been generated in the past to study the metabolism and associated protein production by this yeast. A major challenge with clinical usage of recombinant proteins produced by P. pastoris is the difference in N-glycosylation of proteins produced by humans and this yeast. However, through metabolic engineering, a P. pastoris strain capable of producing humanized N-glycosylated proteins was constructed. The current genome-scale models of P. pastoris do not address native nor humanized N-glycosylation, and we therefore developed ihGlycopastoris, an extension to the iLC915 model with both native and humanized N-glycosylation for recombinant protein production, but also an estimation of N-glycosylation of P. pastoris native proteins. This new model gives a better prediction of protein yield, demonstrates the effect of the different types of N-glycosylation of protein yield, and can be used to predict potential targets for strain improvement. The model represents a step towards a more complete description of protein production in P. pastoris, which is required for using these models to understand and optimize protein production processes. link: http://identifiers.org/pubmed/26480251

Iribe2006_CaMKIIkineticsModel: MODEL1006230085v0.0.1

This a model from the article: Modulatory effect of calmodulin-dependent kinase II (CaMKII) on sarcoplasmic reticulum…

Details

We hypothesize that slow inactivation of Ca2+/calmodulin-dependent kinase II (CaMKII) and its modulatory effect on sarcoplasmic reticulum (SR) Ca2+ handling are important for various interval-force (I-F) relations, in particular for the beat interval dependency in transient alternans during the decay of post-extrasystolic potentiation. We have developed a mathematical model of a single cardiomyocyte to integrate various I-F relations, including alternans, by incorporating a conceptual CaMKII kinetics model into the SR Ca2+ handling model. Our model integrates I-F relations, such as the beat interval-dependent twitch force duration, restitution and potentiation, positive staircase phenomenon and alternans. We found that CaMKII affects more or less all I-F relations, and it is a key factor for integration of the various I-F relations in our model. Alternans arises, in the model, out of a steep relation between SR Ca2+ load and release, owing to SR load-dependent changes in the releasability of Ca2+ via the ryanodine receptor. Beat interval-dependent CaMKII activity, owing to its kinetic properties and amplifying effect on SR Ca2+ load dependency of Ca2+ release, replicated the beat interval dependency of alternans, as observed experimentally. Additionally, our model enabled reproduction of the effects of various interventions on alternans, such as the slowing or accelerating of Ca2+ release and/or uptake. We conclude that a slow time-dependent factor, represented in the model by CaMKII, is important for the integration of I-F relations, including alternans, and that our model offers a useful tool for further analysis of the roles of integrative Ca2+ handling in myocardial I-F relations. link: http://identifiers.org/pubmed/16608699

Irvine1999_CardiacSodiumChannel: MODEL0848062679v0.0.1

This a model from the article: Cardiac sodium channel Markov model with temperature dependence and recovery from inact…

Details

A Markov model of the cardiac sodium channel is presented. The model is similar to the CA1 hippocampal neuron sodium channel model developed by Kuo and Bean (1994. Neuron. 12:819-829) with the following modifications: 1) an additional open state is added; 2) open-inactivated transitions are made voltage-dependent; and 3) channel rate constants are exponential functions of enthalpy, entropy, and voltage and have explicit temperature dependence. Model parameters are determined using a simulated annealing algorithm to minimize the error between model responses and various experimental data sets. The model reproduces a wide range of experimental data including ionic currents, gating currents, tail currents, steady-state inactivation, recovery from inactivation, and open time distributions over a temperature range of 10 degrees C to 25 degrees C. The model also predicts measures of single channel activity such as first latency, probability of a null sweep, and probability of reopening. link: http://identifiers.org/pubmed/10096885

Isaeva2008 - Modelling of Anti-Tumour Immune Response Immunocorrective Effect of Weak Centimetre Electromagnetic Waves: BIOMD0000000910v0.0.1

Modelling of anti-tumour immune response: Immunocorrective effect of weak centimetre electromagnetic waves O.G. Isaeva*…

Details

Abstract

We formulate the dynamical model for the anti-tumour immune response based on intercellular cytokine-mediated interactions with the interleukin-2 (IL-2) taken into account. The analysis shows that the expression level of tumour antigens on antigen presenting cells has a distinct influence on the tumour dynamics. At low antigen presentation, a progressive tumour growth takes place to the highest possible value. At high antigen presentation, there is a decrease in tumour size to some value when the dynamical equilibrium between the tumour and the immune system is reached. In the case of the medium antigen presentation, both these regimes can be realized depending on the initial tumour size and the condition of the immune system. A pronounced immunomodulating effect (the suppression of tumour growth and the normalization of IL-2 concentration) is established by considering the influence of low-intensity electromagnetic microwaves as a parametric perturbation of the dynamical system. This finding is in qualitative agreement with the recent experimental results on immunocorrective effects of centimetre electromagnetic waves in tumour-bearing mice.

Volume 10, Issue 3, Pages 185-201 link: http://identifiers.org/doi/10.1080/17486700802373540

Parameters:

NameDescription
beta_T = 8.4E-8; alpha_T = 0.22; gama_prime_L = 4.0E-7Reaction: T => ; L, Rate Law: compartment*(alpha_T*T*ln(beta_T*T/alpha_T)+gama_prime_L*L*T)
beta_L = 0.33Reaction: L =>, Rate Law: compartment*beta_L*L
alpha_bar_L = 6.6E-8; gama_T = 6.6E-7Reaction: I2 => ; L, T, Rate Law: compartment*(alpha_bar_L*L*I2+gama_T*T*I2)
VL = 79000.0; alpha_L = 9.9E-9Reaction: => L; I2, Rate Law: compartment*(VL+alpha_L*L*I2)
alpha_I2 = 1.25E7; K_T = 52000.0Reaction: => I2; T, Rate Law: compartment*alpha_I2*T/(T+K_T)

States:

NameDescription
I2[Interleukin-2]
T[Neoplastic Cell]
L[C12543]

Isaeva2009 - Different strategies for cancer treatment: MODEL2001140002v0.0.1

We formulate and analyse a mathematical model describing immune response to avasculartumour under the influence of immun…

Details

We formulate and analyse a mathematical model describing immune response to avascular tumour under the influence of immunotherapy and chemotherapy and their combinations as well as vaccine treatments. The effect of vaccine therapy is considered as a parametric perturbation of the model. In the case of a weak immune response, neither immunotherapy nor chemotherapy is found to cause tumour regression to a small size, which would be below the clinically detectable threshold. Numerical simulations show that the efficiency of vaccine therapy depends on both the tumour size and the condition of immune system as well as on the response of the organism to vaccination. In particular, we found that vaccine therapy becomes more effective when used without time delay from a prescribed date of vaccination after surgery and is ineffective without preliminary treatment. For a strong immune response, our model predicts the tumour remission under vaccine therapy. Our study of successive chemo/immuno, immuno/chemo and concurrent chemoimmunotherapy shows that the chemo/immuno sequence is more effective while concurrent chemoimmunotherapy is more sparing.

Vol. 10, No. 4, December 2009, 253–272 link: http://identifiers.org/doi/10.1080/17486700802536054

Islam2010_Dehalococcoides_Metabolism: MODEL1011080003v0.0.1

This is metabolic network reconstruction of Dehalococcoides , iAI549, described in the article Characterizing the met…

Details

Dehalococcoides strains respire a wide variety of chloro-organic compounds and are important for the bioremediation of toxic, persistent, carcinogenic, and ubiquitous ground water pollutants. In order to better understand metabolism and optimize their application, we have developed a pan-genome-scale metabolic network and constraint-based metabolic model of Dehalococcoides. The pan-genome was constructed from publicly available complete genome sequences of Dehalococcoides sp. strain CBDB1, strain 195, strain BAV1, and strain VS. We found that Dehalococcoides pan-genome consisted of 1118 core genes (shared by all), 457 dispensable genes (shared by some), and 486 unique genes (found in only one genome). The model included 549 metabolic genes that encoded 356 proteins catalyzing 497 gene-associated model reactions. Of these 497 reactions, 477 were associated with core metabolic genes, 18 with dispensable genes, and 2 with unique genes. This study, in addition to analyzing the metabolism of an environmentally important phylogenetic group on a pan-genome scale, provides valuable insights into Dehalococcoides metabolic limitations, low growth yields, and energy conservation. The model also provides a framework to anchor and compare disparate experimental data, as well as to give insights on the physiological impact of "incomplete" pathways, such as the TCA-cycle, CO(2) fixation, and cobalamin biosynthesis pathways. The model, referred to as iAI549, highlights the specialized and highly conserved nature of Dehalococcoides metabolism, and suggests that evolution of Dehalococcoides species is driven by the electron acceptor availability. link: http://identifiers.org/pubmed/20811585

Ito2019 - gefitnib resistance of lung adenocarcinoma caused by MET amplification: BIOMD0000000827v0.0.1

The model is based on publication: Mathematical analysis of gefitinib resistance of lung adenocarcinoma caused by MET a…

Details

Gefitinib, one of the tyrosine kinase inhibitors of epidermal growth factor receptor (EGFR), is effective for treating lung adenocarcinoma harboring EGFR mutation; but later, most cases acquire a resistance to gefitinib. One of the mechanisms conferring gefitinib resistance to lung adenocarcinoma is the amplification of the MET gene, which is observed in 5-22% of gefitinib-resistant tumors. A previous study suggested that MET amplification could cause gefitinib resistance by driving ErbB3-dependent activation of the PI3K pathway. In this study, we built a mathematical model of gefitinib resistance caused by MET amplification using lung adenocarcinoma HCC827-GR (gefitinib resistant) cells. The molecular reactions involved in gefitinib resistance consisted of dimerization and phosphorylation of three molecules, EGFR, ErbB3, and MET were described by a series of ordinary differential equations. To perform a computer simulation, we quantified each molecule on the cell surface using flow cytometry and estimated unknown parameters by dimensional analysis. Our simulation showed that the number of active ErbB3 molecules is around a hundred-fold smaller than that of active MET molecules. Limited contribution of ErbB3 in gefitinib resistance by MET amplification is also demonstrated using HCC827-GR cells in culture experiments. Our mathematical model provides a quantitative understanding of the molecular reactions underlying drug resistance. link: http://identifiers.org/pubmed/30824185

Parameters:

NameDescription
k_p2 = 1.0E10Reaction: X_10_p_MET_MET + X_5_EGFR_ErbB3 => X_10_p_MET_MET + X_8_p_EGFR_ErbB3, Rate Law: compartment*k_p2*X_5_EGFR_ErbB3*X_10_p_MET_MET
k_4 = 2.6E12; l_4 = 1.0Reaction: X_3_MET => X_7_MET_MET, Rate Law: compartment*(0.5*k_4*X_3_MET^2-l_4*X_7_MET_MET)
k_3 = 2.4E11; l_3 = 1.0Reaction: X_1_EGFR + X_2_ErbB3 => X_5_EGFR_ErbB3, Rate Law: compartment*(k_3*X_1_EGFR*X_2_ErbB3-l_3*X_5_EGFR_ErbB3)
k_2 = 1.9E12; l_2 = 1.08Reaction: X_2_ErbB3 => X_6_ErbB3_ErbB3, Rate Law: compartment*(0.5*k_2*X_2_ErbB3^2-l_2*X_6_ErbB3_ErbB3)
l_p3 = 0.028Reaction: X_9_p_ErbB3_ErbB3 => X_6_ErbB3_ErbB3, Rate Law: compartment*l_p3*X_9_p_ErbB3_ErbB3
k_p1 = 0.045; l_p1 = 0.028Reaction: X_7_MET_MET => X_10_p_MET_MET, Rate Law: compartment*(k_p1*X_7_MET_MET-l_p1*X_10_p_MET_MET)
l_p2 = 0.028Reaction: X_8_p_EGFR_ErbB3 => X_5_EGFR_ErbB3, Rate Law: compartment*l_p2*X_8_p_EGFR_ErbB3
l_1 = 1.24; k_1 = 2.3E10Reaction: X_1_EGFR => X_4_EGFR_EGFR, Rate Law: compartment*(0.5*k_1*X_1_EGFR^2-l_1*X_4_EGFR_EGFR)
k_p3 = 1.0E10Reaction: X_10_p_MET_MET + X_6_ErbB3_ErbB3 => X_10_p_MET_MET + X_9_p_ErbB3_ErbB3, Rate Law: compartment*k_p3*X_6_ErbB3_ErbB3*X_10_p_MET_MET

States:

NameDescription
X 9 p ErbB3 ErbB3[CCO:13867; Combination]
X 3 MET[CCO:2065]
X 1 EGFR[CCO:1956]
X 8 p EGFR ErbB3[CCO:13867; CCO:1956; Combination]
X 5 EGFR ErbB3[Combination; CCO:1956; CCO:13867]
X 6 ErbB3 ErbB3[CCO:13867; Combination]
X 7 MET MET[Met-Met; Combination]
X 4 EGFR EGFR[Combination; CCO:1956]
X 10 p MET MET[Combination; Met-Met]
X 2 ErbB3[CCO:13867]

Iwamoto2010 - Cell cycle reponse to DNA damage: BIOMD0000000939v0.0.1

After DNA damage, cells activate p53, a tumor suppressor gene, and select a cell fate (e.g., DNA repair, cell cycle arre…

Details

After DNA damage, cells activate p53, a tumor suppressor gene, and select a cell fate (e.g., DNA repair, cell cycle arrest, or apoptosis). Recently, a p53 oscillatory behavior was observed following DNA damage. However, the relationship between this p53 oscillation and cell-fate selection is unclear. Here, we present a novel model of the DNA damage signaling pathway that includes p53 and whole cell cycle regulation and explore the relationship between p53 oscillation and cell fate selection. The simulation run without DNA damage qualitatively realized experimentally observed data from several cell cycle regulators, indicating that our model was biologically appropriate. Moreover, the comprehensive sensitivity analysis for the proposed model was implemented by changing the values of all kinetic parameters, which revealed that the cell cycle regulation system based on the proposed model has robustness on a fluctuation of reaction rate in each process. Simulations run with four different intensities of DNA damage, i.e. Low-damage, Medium-damage, High-damage, and Excess-damage, realized cell cycle arrest in all cases. Low-damage, Medium-damage, High-damage, and Excess-damage corresponded to the DNA damage caused by 100, 200, 400, and 800 J/m(2) doses of UV-irradiation, respectively, based on expression of p21, which plays a crucial role in cell cycle arrest. In simulations run with High-damage and Excess-damage, the length of the cell cycle arrest was shortened despite the severe DNA damage, and p53 began to oscillate. Cells initiated apoptosis and were killed at 400 and 800 J/m(2) doses of UV-irradiation, corresponding to High-damage and Excess-damage, respectively. Therefore, our model indicated that the oscillatory mode of p53 profoundly affects cell fate selection. link: http://identifiers.org/pubmed/21095219

Parameters:

NameDescription
k24 = 0.0225Reaction: Cyclin_E_Cdk2_active_unphosphorylated + p27 => p27_CyclinE_Cdk2, Rate Law: nuclear*k24*Cyclin_E_Cdk2_active_unphosphorylated*p27
k37 = 5.0E-5Reaction: => p21, Rate Law: nuclear*k37
k33 = 1.75E-4Reaction: p21_CyclinA_Cdk2 => Cyclin_A_Cdk2_active + p21, Rate Law: nuclear*k33*p21_CyclinA_Cdk2
k14 = 0.0075Reaction: Cyclin_A_Cdk2_active => Cdk2; APC_Ccdc20_active, APC_Ccdh1_active, Rate Law: nuclear*k14*Cyclin_A_Cdk2_active*(APC_Ccdc20_active+APC_Ccdh1_active)
k50 = 0.0025Reaction: Rb_PP_E2F => E2F + Rb_PPP; Cyclin_A_Cdk2_active, Rate Law: nuclear*k50*Rb_PP_E2F*Cyclin_A_Cdk2_active
k32 = 0.0025Reaction: Cyclin_A_Cdk2_active + p21 => p21_CyclinA_Cdk2, Rate Law: nuclear*k32*Cyclin_A_Cdk2_active*p21
k104 = 1.75E-4Reaction: p21_CyclinB_Cdk1 => p21 + Cyclin_B_Cdk1_nuclear, Rate Law: nuclear*k104*p21_CyclinB_Cdk1
k133 = 5.0E-4Reaction: Cyclin_B_Cdk1_nuclear => Cyclin_B_Cdk1_nuclear_inactive; Wee1, Rate Law: nuclear*k133*Wee1*Cyclin_B_Cdk1_nuclear
k99 = 2.0E-4Reaction: => Wee1, Rate Law: nuclear*k99
k46 = 0.0025Reaction: Rb_E2F => Rb_PP_E2F; Cyclin_D_Cdk4, Rate Law: nuclear*k46*Cyclin_D_Cdk4*Rb_E2F
k15 = 0.005Reaction: Cyclin_A_Cdk2_inactive => Cdk2; APC_Ccdc20_active, APC_Ccdh1_active, Rate Law: nuclear*k15*Cyclin_A_Cdk2_inactive*(APC_Ccdc20_active+APC_Ccdh1_active)
k131 = 0.01Reaction: Cyclin_B_Cdk1_active_phosphorylated_cytoplasm => Cyclin_B_Cdk1_nuclear, Rate Law: k131*Cyclin_B_Cdk1_active_phosphorylated_cytoplasm*Cyclin_B_Cdk1_active_phosphorylated_cytoplasm
k125 = 0.005Reaction: APC_Ccdh1_inactive => APC_Ccdh1_active, Rate Law: nuclear*k125*APC_Ccdh1_inactive
k130 = 3.0E-6Reaction: => Cyclin_A; E2F, Rate Law: nuclear*k130*E2F
k85 = 0.005Reaction: Cdc25A_active => Cdc25A_inactive, Rate Law: nuclear*k85*Cdc25A_active
k35 = 0.05Reaction: p27 => ; Cyclin_E_Cdk2_active_unphosphorylated, Rate Law: nuclear*k35*p27*Cyclin_E_Cdk2_active_unphosphorylated
k22 = 0.006Reaction: Cyclin_E_Cdk2_inactive_phosphorylated => Cyclin_E_Cdk2_active_unphosphorylated; Cdc25A_active, Rate Law: nuclear*k22*Cdc25A_active*Cyclin_E_Cdk2_inactive_phosphorylated
k111 = 0.001Reaction: Cdc25C_inactive => Cdc25C_Ps216_phosphorylated_inactive; Chk1_active, Rate Law: nuclear*k111*Chk1_active*Cdc25C_inactive
k60 = 1.0E-4Reaction: => p53, Rate Law: nuclear*k60
k38 = 0.001Reaction: => p21; p53, Rate Law: nuclear*k38*p53
k49 = 0.04Reaction: Rb_PP_E2F => E2F + Rb_PPP; Cyclin_E_Cdk2_active_unphosphorylated, Rate Law: nuclear*k49*Cyclin_E_Cdk2_active_unphosphorylated*Rb_PP_E2F
k59 = 5.0E-4; k58 = 5.0E-5Reaction: => Rb; p16, Rate Law: nuclear*k58/(1+k59*p16)
k42 = 1.0E-4; k41 = 5.0E-5Reaction: => p16; Rb, Rate Law: nuclear*k41/(1+k42*Rb)
k12 = 2.0E-4Reaction: Cyclin_A_Cdk2_inactive => Cyclin_A + Cdk2, Rate Law: nuclear*k12*Cyclin_A_Cdk2_inactive
k36 = 0.0015Reaction: p27 => ; Cyclin_A_Cdk2_active, Rate Law: nuclear*k36*p27*Cyclin_A_Cdk2_active
k44 = 5.0E-4Reaction: Cyclin_D_Cdk4 + p16 =>, Rate Law: nuclear*k44*Cyclin_D_Cdk4*p16
k20 = 5.0E-4Reaction: Cyclin_D_Cdk4 + p27 => p27_CyclinD_Cdk4, Rate Law: nuclear*k20*Cyclin_D_Cdk4*p27
k107 = 0.002Reaction: B_Myb_active =>, Rate Law: nuclear*k107*B_Myb_active
k62 = 0.001Reaction: p53 => ; deg, Mdm2, Rate Law: nuclear*k62*p53
k21 = 5.0E-5Reaction: p27_CyclinD_Cdk4 => p27 + Cyclin_D_Cdk4, Rate Law: nuclear*k21*p27_CyclinD_Cdk4
k54 = 0.01Reaction: E2F => ; Cyclin_A_Cdk2_active, Rate Law: nuclear*k54*E2F*Cyclin_A_Cdk2_active
k48 = 0.0025Reaction: Rb_E2F => Rb_PP_E2F; p21_CyclinD_Cdk4, Rate Law: nuclear*k48*p21_CyclinD_Cdk4*Rb_E2F
k61 = 0.07Reaction: => p53; ATM_ATR, Rate Law: nuclear*k61*ATM_ATR
k16 = 0.005Reaction: Cyclin_E_Cdk2_inactive_phosphorylated => Cdk2, Rate Law: nuclear*k16*Cyclin_E_Cdk2_inactive_phosphorylated
k103 = 0.0225Reaction: Cyclin_B_Cdk1_nuclear + p21 => p21_CyclinB_Cdk1, Rate Law: nuclear*k103*Cyclin_B_Cdk1_nuclear*p21
k18 = 5.0E-4Reaction: p21 + Cyclin_D_Cdk4 => p21_CyclinD_Cdk4, Rate Law: nuclear*k18*p21*Cyclin_D_Cdk4
k8 = 2.5E-5Reaction: Cyclin_E_Cdk2_inactive_phosphorylated => Cyclin_E + Cdk2, Rate Law: nuclear*k8*Cyclin_E_Cdk2_inactive_phosphorylated
k30 = 0.0025Reaction: Cyclin_A_Cdk2_active + p27 => p27_CyclinA_Cdk2, Rate Law: nuclear*k30*Cyclin_A_Cdk2_active*p27
k27 = 1.75E-4Reaction: p21_CyclinE_Cdk2 => Cyclin_E_Cdk2_active_unphosphorylated + p21, Rate Law: nuclear*k27*p21_CyclinE_Cdk2
k31 = 1.75E-4Reaction: p27_CyclinA_Cdk2 => Cyclin_A_Cdk2_active + p27, Rate Law: nuclear*k31*p27_CyclinA_Cdk2
k108 = 1.0E-5Reaction: => Cdc25C_inactive, Rate Law: nuclear*k108
k25 = 1.75E-4Reaction: p27_CyclinE_Cdk2 => Cyclin_E_Cdk2_active_unphosphorylated + p27, Rate Law: nuclear*k25*p27_CyclinE_Cdk2
k106 = 0.05Reaction: B_Myb_inactive => B_Myb_active; Cyclin_A_Cdk2_active, Rate Law: nuclear*k106*B_Myb_inactive*Cyclin_A_Cdk2_active
k23 = 0.00175Reaction: Cyclin_E_Cdk2_active_unphosphorylated => Cyclin_E_Cdk2_inactive_phosphorylated, Rate Law: nuclear*k23*Cyclin_E_Cdk2_active_unphosphorylated
k43 = 5.0E-4Reaction: p16 =>, Rate Law: nuclear*k43*p16
k109 = 0.01Reaction: Cdc25C_active => Cdc25C_inactive, Rate Law: nuclear*k109*Cdc25C_active
k47 = 0.0025Reaction: Rb_E2F => Rb_PP_E2F; p27_CyclinD_Cdk4, Rate Law: nuclear*k47*p27_CyclinD_Cdk4*Rb_E2F
k137 = 0.03Reaction: Cyclin_B_Cdk1_nuclear => ; APC_Ccdh1_active, Rate Law: nuclear*k137*Cyclin_B_Cdk1_nuclear*APC_Ccdh1_active
k40 = 0.002Reaction: => p16, Rate Law: nuclear*k40
k53 = 5.0E-5Reaction: E2F =>, Rate Law: nuclear*k53*E2F
k17 = 0.05Reaction: Cyclin_E_Cdk2_active_unphosphorylated => Cdk2, Rate Law: nuclear*k17*Cyclin_E_Cdk2_active_unphosphorylated*Cyclin_E_Cdk2_active_unphosphorylated
k19 = 0.005Reaction: p21_CyclinD_Cdk4 => p21 + Cyclin_D_Cdk4, Rate Law: nuclear*k19*p21_CyclinD_Cdk4
k45 = 5.0E-5Reaction: E2F + Rb => Rb_E2F, Rate Law: nuclear*k45*E2F*Rb
k134 = 0.01Reaction: Cyclin_B_Cdk1_nuclear_inactive => Cyclin_B_Cdk1_nuclear; Cdc25C_active, Cdc25C_Ps216_phosphorylated_active, Rate Law: nuclear*k134*Cyclin_B_Cdk1_nuclear_inactive*(Cdc25C_active+Cdc25C_Ps216_phosphorylated_active)
k122 = 0.005Reaction: APC_Ccdc20_active => APC_Ccdc20_inactive; APC_Ccdh1_active, Rate Law: nuclear*k122*APC_Ccdh1_active*APC_Ccdc20_active
k57 = 0.005Reaction: Rb =>, Rate Law: nuclear*k57*Rb
k86 = 5.0E-4Reaction: Cdc25A_active =>, Rate Law: nuclear*k86*Cdc25A_active
k34 = 5.0E-8Reaction: => p27, Rate Law: nuclear*k34
k26 = 0.0225Reaction: Cyclin_E_Cdk2_active_unphosphorylated + p21 => p21_CyclinE_Cdk2, Rate Law: nuclear*k26*Cyclin_E_Cdk2_active_unphosphorylated*p21
k39 = 0.005Reaction: p21 =>, Rate Law: nuclear*k39*p21
k11 = 5.0E-4Reaction: Cyclin_A + Cdk2 => Cyclin_A_Cdk2_inactive, Rate Law: nuclear*k11*Cyclin_A*Cdk2
k110 = 1.0Reaction: Cdc25C_inactive => Cdc25C_active; Cyclin_B_Cdk1_active_phosphorylated_cytoplasm, Cyclin_B_Cdk1_nuclear, Rate Law: k110*Cdc25C_inactive*(Cyclin_B_Cdk1_active_phosphorylated_cytoplasm+Cyclin_B_Cdk1_nuclear)
k105 = 0.05Reaction: => B_Myb_inactive; E2F, Rate Law: nuclear*k105*E2F

States:

NameDescription
Cyclin B Cdk1 nuclear[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin-B1; protein-containing complex]
p21 CyclinE Cdk2[G1/S-specific cyclin-E1; Cyclin-dependent kinase inhibitor 1; Cyclin-dependent kinase 2; protein-containing complex]
p27 CyclinE Cdk2[G1/S-specific cyclin-E1; Cyclin-dependent kinase inhibitor 1B; Cyclin-dependent kinase 2; protein-containing complex]
Rb E2F[Transcription factor E2F1; Retinoblastoma-like protein 2; protein-containing complex]
Cyclin A total[Cyclin-A2]
B Myb inactive[Myb-related protein B; inactive]
Cdc25A active[M-phase inducer phosphatase 1; active]
p53[Cellular tumor antigen p53]
E2F[Transcription factor E2F1]
APC Ccdc20 active[anaphase-promoting complex; Cell division cycle protein 20 homolog; protein-containing complex; active]
B Myb active[Myb-related protein B; active]
Cdc25C Ps216 phosphorylated inactive[M-phase inducer phosphatase 3; phosphorylated; inactive]
Cyclin B total[G2/mitotic-specific cyclin-B1]
p16[Cyclin-dependent kinase inhibitor 2A]
Cdk2[Cyclin-dependent kinase 2]
Cyclin A Cdk2 inactive[Cyclin-dependent kinase 2; Cyclin-A2; inactive; protein-containing complex]
Cyclin A Cdk2 active[Cyclin-dependent kinase 2; Cyclin-A2; protein-containing complex; active]
p21 CyclinB Cdk1[Cyclin-dependent kinase 1; Cyclin-dependent kinase inhibitor 1; G2/mitotic-specific cyclin-B1; protein-containing complex]
Wee1[Wee1-like protein kinase]
Rb[Retinoblastoma-like protein 2]
Rb PP E2F[Retinoblastoma-like protein 2; Transcription factor E2F1; phosphorylated; protein-containing complex]
p21 CyclinA Cdk2[Cyclin-A2; Cyclin-dependent kinase inhibitor 1; Cyclin-dependent kinase 2; protein-containing complex]
p27 CyclinD Cdk4[G1/S-specific cyclin-D2; Cyclin-dependent kinase 4; Cyclin-dependent kinase inhibitor 1B; protein-containing complex]
p27 CyclinA Cdk2[Cyclin-dependent kinase 2; Cyclin-A2; Cyclin-dependent kinase inhibitor 1B; protein-containing complex]
Rb PPP[Retinoblastoma-like protein 2; phosphorylated]
Cyclin A[Cyclin-A2]
p21 CyclinD Cdk4[Cyclin-dependent kinase 4; G1/S-specific cyclin-D2; Cyclin-dependent kinase inhibitor 1; protein-containing complex]
Cyclin B Cdk1 nuclear inactive[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin-B1; protein-containing complex; inactive]
p21[Cyclin-dependent kinase inhibitor 1]
p27[Cyclin-dependent kinase inhibitor 1B]
APC C Cdc20 active x20[Cell division cycle protein 20 homolog; anaphase-promoting complex; active; protein-containing complex]
APC Ccdh1 active[anaphase-promoting complex; Cadherin-1; protein-containing complex; active]
Cyclin E Cdk2 active unphosphorylated[G1/S-specific cyclin-E1; Cyclin-dependent kinase 2; protein-containing complex; active]
Cdc25C inactive[M-phase inducer phosphatase 3; inactive]

Iyer2004_VentricularMyocyte: MODEL0847999575v0.0.1

This a model from the article: A computational model of the human left-ventricular epicardial myocyte. Iyer V, Mazha…

Details

A computational model of the human left-ventricular epicardial myocyte is presented. Models of each of the major ionic currents present in these cells are formulated and validated using experimental data obtained from studies of recombinant human ion channels and/or whole-cell recording from single myocytes isolated from human left-ventricular subepicardium. Continuous-time Markov chain models for the gating of the fast Na(+) current, transient outward current, rapid component of the delayed rectifier current, and the L-type calcium current are modified to represent human data at physiological temperature. A new model for the gating of the slow component of the delayed rectifier current is formulated and validated against experimental data. Properties of calcium handling and exchanger currents are altered to appropriately represent the dynamics of intracellular ion concentrations. The model is able to both reproduce and predict a wide range of behaviors observed experimentally including action potential morphology, ionic currents, intracellular calcium transients, frequency dependence of action-potential duration, Ca(2+)-frequency relations, and extrasystolic restitution/post-extrasystolic potentiation. The model therefore serves as a useful tool for investigating mechanisms of arrhythmia and consequences of drug-channel interactions in the human left-ventricular myocyte. link: http://identifiers.org/pubmed/15345532

Iyer2007_Arrhythmia_CardiacDeath: MODEL1006230030v0.0.1

This a model from the article: Mechanisms of abnormal calcium homeostasis in mutations responsible for catecholaminerg…

Details

Catecholaminergic polymorphic ventricular tachycardia is a heritable arrhythmia unmasked by exertion or stress and is characterized by triggered activity and sudden cardiac death. In this study, we simulated mutations in 2 genes linked to catecholaminergic polymorphic ventricular tachycardia, the first located in calsequestrin (CSQN2) and the second in the ryanodine receptor (RyR2). The aim of the study was to investigate the mechanistic basis for spontaneous Ca2+ release events that lead to delayed afterdepolarizations in affected patients. Sarcoplasmic reticulum (SR) luminal Ca2+ sensing was incorporated into a model of the human ventricular myocyte, and CSQN2 mutations were modeled by simulating disrupted RyR2 luminal Ca2+ sensing. In voltage-clamp mode, the mutant CSQN2 model recapitulated the smaller calcium transients, smaller time to peak calcium transient, and accelerated recovery from inactivation seen in experiments. In current clamp mode, in the presence of beta stimulation, we observed delayed afterdepolarizations, suggesting that accelerated recovery of RyR2 induced by impaired luminal Ca2+ sensing underlies the triggered activity observed in mutant CSQN2-expressing myocytes. In current-clamp mode, in a model of mutant RyR2 that is characterized by reduced FKBP12.6 binding to the RyR2 on beta stimulation, the impaired coupled gating characteristic of these mutations was modeled by reducing cooperativity of RyR2 activation. In current-clamp mode, the mutant RyR2 model exhibited increased diastolic RyR2 open probability that resulted in formation of delayed afterdepolarizations. In conclusion, these minimal order models of mutant CSQN2 and RyR2 provide plausible mechanisms by which defects in RyR2 gating may lead to the cellular triggers for arrhythmia, with implications for the development of targeted therapy. link: http://identifiers.org/pubmed/17234962

Izhikevich2003_SpikingNeuron: BIOMD0000000127v0.0.1

The model is according to the paper *Simple Model of Spiking Neurons* In this paper, a simple spiking model is presente…

Details

A model is presented that reproduces spiking and bursting behavior of known types of cortical neurons. The model combines the biologically plausibility of Hodgkin-Huxley-type dynamics and the computational efficiency of integrate-and-fire neurons. Using this model, one can simulate tens of thousands of spiking cortical neurons in real time (1 ms resolution) using a desktop PC. link: http://identifiers.org/pubmed/18244602

Izhikevich2004_SpikingNeurons_Class1Excitable: BIOMD0000000141v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich EM. IEEE Trans Neural N…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

Izhikevich2004_SpikingNeurons_Class2Excitable: BIOMD0000000142v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich EM. IEEE Trans Neural N…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

Izhikevich2004_SpikingNeurons_inhibitionInducedSpiking: BIOMD0000000129v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich EM. IEEE Trans Neural N…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

Izhikevich2004_SpikingNeurons_integrator: BIOMD0000000130v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich, EM Neural Networks, IE…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

Izhikevich2004_SpikingNeurons_reboundBurst: BIOMD0000000131v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich EM. IEEE Trans Neural Net…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

Izhikevich2004_SpikingNeurons_reboundSpike: BIOMD0000000132v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich EM. IEEE Trans Neural Net…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

Izhikevich2004_SpikingNeurons_resonator: BIOMD0000000133v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich EM. IEEE Trans Neural Net…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

Izhikevich2004_SpikingNeurons_SpikeLatency: BIOMD0000000134v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich EM. IEEE Trans Neural N…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

Izhikevich2004_SpikingNeurons_subthresholdOscillations: BIOMD0000000135v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich EM. IEEE Trans Neural N…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

Izhikevich2004_SpikingNeurons_thresholdVariability: BIOMD0000000136v0.0.1

This a model from the article: Which model to use for cortical spiking neurons? Izhikevich EM. IEEE Trans Neural N…

Details

We discuss the biological plausibility and computational efficiency of some of the most useful models of spiking and bursting neurons. We compare their applicability to large-scale simulations of cortical neural networks. link: http://identifiers.org/pubmed/15484883

J


Jafarnejad2019 - Mechanistically detailed systems biology modeling of the HGF/Met pathway in hepatocellular carcinoma: MODEL2003200001v0.0.1

Hepatocyte growth factor (HGF) signaling through its receptor Met has been implicated in hepatocellular carcinoma tumori…

Details

Hepatocyte growth factor (HGF) signaling through its receptor Met has been implicated in hepatocellular carcinoma tumorigenesis and progression. Met interaction with integrins is shown to modulate the downstream signaling to Akt and ERK (extracellular-regulated kinase). In this study, we developed a mechanistically detailed systems biology model of HGF/Met signaling pathway that incorporated specific interactions with integrins to investigate the efficacy of integrin-binding peptide, AXT050, as monotherapy and in combination with other therapeutics targeting this pathway. Here we report that the modeled dynamics of the response to AXT050 revealed that receptor trafficking is sufficient to explain the effect of Met-integrin interactions on HGF signaling. Furthermore, the model predicted patient-specific synergy and antagonism of efficacy and potency for combination of AXT050 with sorafenib, cabozantinib, and rilotumumab. Overall, the model provides a valuable framework for studying the efficacy of drugs targeting receptor tyrosine kinase interaction with integrins, and identification of synergistic drug combinations for the patients. link: http://identifiers.org/pubmed/31452933

Jafri1998_VentricularMyocyte: MODEL0847869198v0.0.1

This a model from the article: Cardiac Ca2+ dynamics: the roles of ryanodine receptor adaptation and sarcoplasmic reti…

Details

We construct a detailed mathematical model for Ca2+ regulation in the ventricular myocyte that includes novel descriptions of subcellular mechanisms based on recent experimental findings: 1) the Keizer-Levine model for the ryanodine receptor (RyR), which displays adaptation at elevated Ca2+; 2) a model for the L-type Ca2+ channel that inactivates by mode switching; and 3) a restricted subspace into which the RyRs and L-type Ca2+ channels empty and interact via Ca2+. We add membrane currents from the Luo-Rudy Phase II ventricular cell model to our description of Ca2+ handling to formulate a new model for ventricular action potentials and Ca2+ regulation. The model can simulate Ca2+ transients during an action potential similar to those seen experimentally. The subspace [Ca2+] rises more rapidly and reaches a higher level (10-30 microM) than the bulk myoplasmic Ca2+ (peak [Ca2+]i approximately 1 microM). Termination of sarcoplasmic reticulum (SR) Ca2+ release is predominately due to emptying of the SR, but is influenced by RyR adaptation. Because force generation is roughly proportional to peak myoplasmic Ca2+, we use [Ca2+]i in the model to explore the effects of pacing rate on force generation. The model reproduces transitions seen in force generation due to changes in pacing that cannot be simulated by previous models. Simulation of such complex phenomena requires an interplay of both RyR adaptation and the degree of SR Ca2+ loading. This model, therefore, shows improved behavior over existing models that lack detailed descriptions of subcellular Ca2+ regulatory mechanisms. link: http://identifiers.org/pubmed/9512016

Jaiswal2017 - Cell cycle arrest: BIOMD0000000641v0.0.1

Jaiswal2017 - Cell cycle arrestThis model is described in the article: [ATM/Wip1 activities at chromatin control Plk1 r…

Details

After DNA damage, the cell cycle is arrested to avoid propagation of mutations. Arrest in G2 phase is initiated by ATM-/ATR-dependent signaling that inhibits mitosis-promoting kinases such as Plk1. At the same time, Plk1 can counteract ATR-dependent signaling and is required for eventual resumption of the cell cycle. However, what determines when Plk1 activity can resume remains unclear. Here, we use FRET-based reporters to show that a global spread of ATM activity on chromatin and phosphorylation of ATM targets including KAP1 control Plk1 re-activation. These phosphorylations are rapidly counteracted by the chromatin-bound phosphatase Wip1, allowing cell cycle restart despite persistent ATM activity present at DNA lesions. Combining experimental data and mathematical modeling, we propose a model for how the minimal duration of cell cycle arrest is controlled. Our model shows how cell cycle restart can occur before completion of DNA repair and suggests a mechanism for checkpoint adaptation in human cells. link: http://identifiers.org/pubmed/28607002

Parameters:

NameDescription
Kcc2a = 1.0; Kch2cc = 1.0; Km10 = 10.0; Kt2cc = 10.0Reaction: CellCact = ((Kcc2a+CellCact)*CellCina/(Km10+CellCina)-Kt2cc*Timeract*CellCact/(Km10+CellCact))-Kch2cc*CellCact*Effectoract/(Km10+CellCact), Rate Law: ((Kcc2a+CellCact)*CellCina/(Km10+CellCina)-Kt2cc*Timeract*CellCact/(Km10+CellCact))-Kch2cc*CellCact*Effectoract/(Km10+CellCact)
Kti2t = 10.0; Km1 = 1.0; Kd2t = 2.0Reaction: Timeract = Kd2t*Damage*Timerinact/(Km1+Timerinact)-Kti2t*Timeract/(Km1+Timeract), Rate Law: Kd2t*Damage*Timerinact/(Km1+Timerinact)-Kti2t*Timeract/(Km1+Timeract)
Km10 = 10.0; Kd2ch = 1.0; Kcc2ch = 1.0Reaction: Effectoract = Kd2ch*Damage*Effectorina/(Km10+Effectorina)-Kcc2ch*CellCact*Effectoract/(Km10+Effectoract), Rate Law: Kd2ch*Damage*Effectorina/(Km10+Effectorina)-Kcc2ch*CellCact*Effectoract/(Km10+Effectoract)

States:

NameDescription
NHEJ[double-strand break repair via nonhomologous end joining]
HR[double-strand break repair via homologous recombination]
CellCact[Serine/threonine-protein kinase PLK1]
Effectoract[Serine/threonine-protein kinase ATR]
Effectorina[Serine/threonine-protein kinase ATR]
Timeract[Serine-protein kinase ATM]
CellCina[Serine/threonine-protein kinase PLK1]
Damage[DNA damage response, detection of DNA damage]
Timerinact[Serine-protein kinase ATM]

Jamshidi01_RBC_MetabolicNetwork: MODEL1103210001v0.0.1

**Dynamic simulation of the human red blood cell metabolic network** Neema Jamshidi, Jeremy S. Edwards, Tom Fahland, G…

Details

We have developed a Mathematica application package to perform dynamic simulations of the red blood cell (RBC) metabolic network. The package relies on, and integrates, many years of mathematical modeling and biochemical work on red blood cell metabolism. The extensive data regarding the red blood cell metabolic network and the previous kinetic analysis of all the individual components makes the human RBC an ideal 'model' system for mathematical metabolic models. The Mathematica package can be used to understand the dynamics and regulatory characteristics of the red blood cell. link: http://identifiers.org/pubmed/11294796

Jamshidi2007 - Genome-scale metabolic network of Mycobacterium tuberculosis (iNJ661): MODEL1507180001v0.0.1

Jamshidi2007 - Genome-scale metabolic network of Mycobacterium tuberculosis (iNJ661)This model is described in the artic…

Details

BACKGROUND: Mycobacterium tuberculosis continues to be a major pathogen in the third world, killing almost 2 million people a year by the most recent estimates. Even in industrialized countries, the emergence of multi-drug resistant (MDR) strains of tuberculosis hails the need to develop additional medications for treatment. Many of the drugs used for treatment of tuberculosis target metabolic enzymes. Genome-scale models can be used for analysis, discovery, and as hypothesis generating tools, which will hopefully assist the rational drug development process. These models need to be able to assimilate data from large datasets and analyze them. RESULTS: We completed a bottom up reconstruction of the metabolic network of Mycobacterium tuberculosis H37Rv. This functional in silico bacterium, iNJ661, contains 661 genes and 939 reactions and can produce many of the complex compounds characteristic to tuberculosis, such as mycolic acids and mycocerosates. We grew this bacterium in silico on various media, analyzed the model in the context of multiple high-throughput data sets, and finally we analyzed the network in an 'unbiased' manner by calculating the Hard Coupled Reaction (HCR) sets, groups of reactions that are forced to operate in unison due to mass conservation and connectivity constraints. CONCLUSION: Although we observed growth rates comparable to experimental observations (doubling times ranging from about 12 to 24 hours) in different media, comparisons of gene essentiality with experimental data were less encouraging (generally about 55%). The reasons for the often conflicting results were multi-fold, including gene expression variability under different conditions and lack of complete biological knowledge. Some of the inconsistencies between in vitro and in silico or in vivo and in silico results highlight specific loci that are worth further experimental investigations. Finally, by considering the HCR sets in the context of known drug targets for tuberculosis treatment we proposed new alternative, but equivalent drug targets. link: http://identifiers.org/pubmed/17555602

Jarrah2014 - mathematical model of the immune response in muscle degeneration and subsequent regeneration in Duchenne muscular dystrophy in mdx mice: BIOMD0000001015v0.0.1

Duchenne muscular dystrophy (DMD) is a genetic disease that results in the death of affected boys by early adulthood.The…

Details

Duchenne muscular dystrophy (DMD) is a genetic disease that results in the death of affected boys by early adulthood. The genetic defect responsible for DMD has been known for over 25 years, yet at present there is neither cure nor effective treatment for DMD. During early disease onset, the mdx mouse has been validated as an animal model for DMD and use of this model has led to valuable but incomplete insights into the disease process. For example, immune cells are thought to be responsible for a significant portion of muscle cell death in the mdx mouse; however, the role and time course of the immune response in the dystrophic process have not been well described. In this paper we constructed a simple mathematical model to investigate the role of the immune response in muscle degeneration and subsequent regeneration in the mdx mouse model of Duchenne muscular dystrophy. Our model suggests that the immune response contributes substantially to the muscle degeneration and regeneration processes. Furthermore, the analysis of the model predicts that the immune system response oscillates throughout the life of the mice, and the damaged fibers are never completely cleared. link: http://identifiers.org/pubmed/25013809

Jarrett2015 - Modelling the interaction between immune response, bacterial dynamics and inflammatory damage: BIOMD0000000920v0.0.1

Mathematical model of pro- and anti-inflammatory response, inflammation/damage and infection dynamics in BALB/c mouse wi…

Details

The immune system is a complex system of chemical and cellular interactions that responds quickly to queues that signal infection and then reverts to a basal level once the challenge is eliminated. Here, we present a general, four-component model of the immune system's response to a Staphylococcal aureus (S. aureus) infection, using ordinary differential equations. To incorporate both the infection and the immune system, we adopt the style of compartmenting the system to include bacterial dynamics, damage and inflammation to the host, and the host response. We incorporate interactions not previously represented including cross-talk between inflammation/damage and the infection and the suppression of the anti-inflammatory pathway in response to inflammation/damage. As a result, the most relevant equilibrium of the system, representing the health state, is an all-positive basal level. The model is able to capture eight different experimental outcomes for mice challenged with intratibial osteomyelitis due to S. aureus, primarily involving immunomodulation and vaccine therapies. For further validation and parameter exploration, we perform a parameter sensitivity analysis which suggests that the model is very stable with respect to variations in parameters, indicates potential immunomodulation strategies and provides a possible explanation for the difference in immune potential for different mouse strains. link: http://identifiers.org/pubmed/24814512

Parameters:

NameDescription
mu_1 = 0.12; K_B = 1.0; alpha_1 = 0.27; beta_1 = 0.01; rho_1 = 0.2Reaction: => pro_inflammatory__P; inflammation__I, bacterial_infection__B, anti_inflammatory__A, Rate Law: BALB_c_Mouse*((alpha_1*inflammation__I+rho_1*bacterial_infection__B)*(1-pro_inflammatory__P)-(beta_1*anti_inflammatory__A+mu_1*(1-bacterial_infection__B/K_B))*pro_inflammatory__P)
K_B = 1.0; beta_4 = 5.0; g = 0.9; alpha_4 = 1.5; gamma = 0.01Reaction: => bacterial_infection__B; inflammation__I, pro_inflammatory__P, Rate Law: BALB_c_Mouse*(((g*(1-bacterial_infection__B/K_B)+alpha_4*inflammation__I)-beta_4*pro_inflammatory__P)*bacterial_infection__B+exp((-1)*gamma*time))
mu_2 = 0.25; beta_2 = 0.135; K_B = 1.0; alpha_2 = 0.11Reaction: => anti_inflammatory__A; pro_inflammatory__P, inflammation__I, bacterial_infection__B, Rate Law: BALB_c_Mouse*(alpha_2*pro_inflammatory__P-(beta_2*inflammation__I+mu_2*(1-bacterial_infection__B/K_B))*anti_inflammatory__A)
beta_3 = 2.0; rho_2 = 0.45; alpha_3 = 1.05; mu_3 = 0.0174Reaction: => inflammation__I; pro_inflammatory__P, bacterial_infection__B, anti_inflammatory__A, Rate Law: BALB_c_Mouse*((alpha_3*pro_inflammatory__P+rho_2*bacterial_infection__B)-(beta_3*anti_inflammatory__A+mu_3)*inflammation__I)

States:

NameDescription
inflammation I[Inflammation]
bacterial infection B[Staphylococcus aureus infection]
pro inflammatory P[inflammatory response; Th17 cell; Th1 cell]
anti inflammatory A[regulatory T-lymphocyte]

Jarrett2018 - trastuzumab-induced immune response in murine HER2+ breast cancer model: BIOMD0000000745v0.0.1

The paper describes a model on the trastuzumab-induced immune response in murine(mouse) HER2+ breast cancer. Created by…

Details

The goal of this study is to develop an integrated, mathematical–experimental approach for understanding the interactions between the immune system and the effects of trastuzumab on breast cancer that overexpresses the human epidermal growth factor receptor 2 (HER2+). A system of coupled, ordinary differential equations was constructed to describe the temporal changes in tumour growth, along with intratumoural changes in the immune response, vascularity, necrosis and hypoxia. The mathematical model is calibrated with serially acquired experimental data of tumour volume, vascularity, necrosis and hypoxia obtained from either imaging or histology from a murine model of HER2+ breast cancer. Sensitivity analysis shows that model components are sensitive for 12 of 13 parameters, but accounting for uncertainty in the parameter values, model simulations still agree with the experimental data. Given theinitial conditions, the mathematical model predicts an increase in the immune infiltrates over time in the treated animals. Immunofluorescent staining results are presented that validate this prediction by showing an increased co-staining of CD11c and F4/80 (proteins expressed by dendritic cells and/or macrophages) in the total tissue for the treated tumours compared to the controls. We posit that the proposed mathematical–experimental approach can be used to elucidate driving interactions between the trastuzumab-induced responses in the tumour and the immune system that drive the stabilization of vasculature while simultaneously decreasing tumour growth—conclusions revealed by the mathematical model that were not deducible from the experimental data alone. link: http://identifiers.org/doi/10.1093/imammb/dqy014

Parameters:

NameDescription
ut = 0.187 1/msReaction: T => ; I, Rate Law: tumor*ut*T*I
beta = 0.027 1/msReaction: => N; V, Rate Law: tumor*(beta+beta*V*N)
beta = 0.027 1/ms; un = 0.911 1/msReaction: N => ; V, I, Rate Law: tumor*(beta*V+beta*N+un*N*I)
an = 0.2 1/ms; av = 0.199 1/msReaction: => I; V, N, Rate Law: tumor*(av*V+an*N)
ai = 0.045 1/ms; at = 0.101 1/msReaction: => V; T, I, Rate Law: tumor*(at*T+ai*I)
rho = 1.523 1; g = 0.044 1/msReaction: => T; H, Rate Law: tumor*g*T*(rho*H+1)
gamma = 0.743 1/ms; delta = 0.284 1Reaction: H => ; V, Rate Law: tumor*(gamma*delta*H*H+gamma*V*H)
ai = 0.045 1/ms; uv = 1.723 1/ms; at = 0.101 1/msReaction: V => ; T, I, Rate Law: tumor*(at*T*V+ai*I*V+uv*V*T)
an = 0.2 1/ms; av = 0.199 1/ms; ui = 0.722 1/msReaction: I => ; V, N, T, Rate Law: tumor*(av*V*I+an*N*I+ui*I*T)

States:

NameDescription
I[immune response to tumor cell]
T[Tumor Volume]
N[necrotic cell death]
VV
H[Hypoxia]

Jelic2005_HypothalamicPituitaryAdrenal: MODEL1006230013v0.0.1

This a model from the article: Mathematical modeling of the hypothalamic-pituitary-adrenal system activity. Jelic S,…

Details

Mathematical modeling has proven to be valuable in understanding of the complex biological systems dynamics. In the present report we have developed an initial model of the hypothalamic-pituitary-adrenal system self-regulatory activity. A four-dimensional non-linear differential equation model of the hormone secretion was formulated and used to analyze plasma cortisol levels in humans. The aim of this work was to explore in greater detail the role of this system in normal, homeostatic, conditions, since it is the first and unavoidable step in further understanding of the role of this complex neuroendocrine system in pathophysiological conditions. Neither the underlying mechanisms nor the physiological significance of this system are fully understood yet. link: http://identifiers.org/pubmed/16112688

Jenkinson2011_EGF_MAPK: BIOMD0000000399v0.0.1

This is a model described in the article: Thermodynamically Consistent Model Calibration in Chemical Kinetics. Garrett…

Details

BACKGROUND: The dynamics of biochemical reaction systems are constrained by the fundamental laws of thermodynamics, which impose well-defined relationships among the reaction rate constants characterizing these systems. Constructing biochemical reaction systems from experimental observations often leads to parameter values that do not satisfy the necessary thermodynamic constraints. This can result in models that are not physically realizable and may lead to inaccurate, or even erroneous, descriptions of cellular function. RESULTS: We introduce a thermodynamically consistent model calibration (TCMC) method that can be effectively used to provide thermodynamically feasible values for the parameters of an open biochemical reaction system. The proposed method formulates the model calibration problem as a constrained optimization problem that takes thermodynamic constraints (and, if desired, additional non-thermodynamic constraints) into account. By calculating thermodynamically feasible values for the kinetic parameters of a well-known model of the EGF/ERK signaling cascade, we demonstrate the qualitative and quantitative significance of imposing thermodynamic constraints on these parameters and the effectiveness of our method for accomplishing this important task. MATLAB software, using the Systems Biology Toolbox 2.1, can be accessed from http://www.cis.jhu.edu/~goutsias/CSS lab/software.html. An SBML file containing the thermodynamically feasible EGF/ERK signaling cascade model can be found in the BioModels database. CONCLUSIONS: TCMC is a simple and flexible method for obtaining physically plausible values for the kinetic parameters of open biochemical reaction systems. It can be effectively used to recalculate a thermodynamically consistent set of parameter values for existing thermodynamically infeasible biochemical reaction models of cellular function as well as to estimate thermodynamically feasible values for the parameters of new models. Furthermore, TCMC can provide dimensionality reduction, better estimation performance, and lower computational complexity, and can help to alleviate the problem of data overfitting. link: http://identifiers.org/pubmed/21548948

Parameters:

NameDescription
k20 = 5.17656E-5 peritempermin; kr20 = 12.816 perminReaction: x35 + x43 => x37, Rate Law: k20*x35*x43-kr20*x37
kr41 = 44.60169 permin; k41 = 0.001522817 peritemperminReaction: x30 + x33 => x35, Rate Law: k41*x30*x33-kr41*x35
kr56 = 1.229629 permin; k56 = 0.004700229 peritemperminReaction: x83 + x60 => x84, Rate Law: k56*x83*x60-kr56*x84
kr42 = 1.870396 permin; k42 = 0.009688174 peritemperminReaction: x44 + x45 => x46, Rate Law: k42*x44*x45-kr42*x46
k25 = 6.871213E-4 peritempermin; kr25 = 1.218132 perminReaction: x24 + x34 => x35, Rate Law: k25*x24*x34-kr25*x35
k52 = 0.003826571 peritempermin; kr52 = 19.85279 perminReaction: x51 + x57 => x58, Rate Law: k52*x51*x57-kr52*x58
k6 = 4.123214E-4 permin; kr6 = 0.294324 perminReaction: x34 => x65, Rate Law: k6*x34-kr6*x65
kr50 = 9.954943 permin; k50 = 5.464454E-4 peritemperminReaction: x53 + x49 => x54, Rate Law: k50*x53*x49-kr50*x54
kr14 = 196.6479 permin; k14 = 6.370566E-7 peritemperminReaction: x8 + x14 => x17, Rate Law: k14*x8*x14-kr14*x17
k49 = 10.73099 perminReaction: x79 => x47 + x53, Rate Law: k49*x79
k8 = 5.174108E-4 peritempermin; kr8 = 0.9058936 perminReaction: x5 + x14 => x15, Rate Law: k8*x5*x14-kr8*x15
k60 = 0.08693199 perminReaction: x8 => x87, Rate Law: k60*x8
k5 = NaN perminReaction: x93 => x9 + x67, Rate Law: k5*x93
k58 = 1.714511E-4 peritempermin; kr58 = 0.1138168 perminReaction: x60 + x57 => x62, Rate Law: k58*x60*x57-kr58*x62
k3 = 31.71871 permin; kr3 = 2.220991 perminReaction: x4 => x5, Rate Law: k3*x4-kr3*x5
k33 = 10.96212 permin; kr33 = 1.788597E-5 peritemperminReaction: x38 => x40 + x30, Rate Law: k33*x38-kr33*x40*x30
k4 = 3.047285E-5 peritempermin; kr4 = 0.1230832 perminReaction: x34 + x12 => x91, Rate Law: k4*x34*x12-kr4*x91
kr37 = 5.477036E-6 peritempermin; k37 = 29.34687 perminReaction: x34 => x15 + x39, Rate Law: k37*x34-kr37*x15*x39
k45 = 6340.081 perminReaction: x48 => x49 + x45, Rate Law: k45*x48
k59 = 6.409354 perminReaction: x62 => x55 + x60, Rate Law: k59*x62
k22 = 1.445554E-4 peritempermin; kr22 = 0.6220457 perminReaction: x31 + x15 => x32, Rate Law: k22*x31*x15-kr22*x32
k47 = 1632.425 perminReaction: x50 => x51 + x45, Rate Law: k47*x50
k34 = 0.2467995 permin; kr34 = 1.283286E-4 peritemperminReaction: x25 => x15 + x30, Rate Law: k34*x25-kr34*x15*x30
k7 = 0.003011324 perminReaction: x5 => x8, Rate Law: k7*x5
Km36 = 7.719778E14 items; Vm36 = 615.0325 itemsperminReaction: x40 => x31, Rate Law: Vm36*x40/(Km36+x40)
k19 = 349.772 permin; kr19 = 5.84737E-6 peritemperminReaction: x36 => x35 + x28, Rate Law: k19*x36-kr19*x35*x28
k21 = 0.4722901 permin; kr21 = 1.714441E-5 peritemperminReaction: x37 => x35 + x26, Rate Law: k21*x37-kr21*x35*x26
kr24 = 563.2135 permin; k24 = 0.007178843 peritemperminReaction: x22 + x33 => x34, Rate Law: k24*x22*x33-kr24*x34
k18 = 0.004463938 peritempermin; kr18 = 11.1361 perminReaction: x26 + x66 => x67, Rate Law: k18*x26*x66-kr18*x67
k48 = 6.874119E-4 peritempermin; kr48 = 1489.015 perminReaction: x51 + x53 => x52, Rate Law: k48*x51*x53-kr48*x52
k32 = 14.19908 permin; kr32 = 5.54527E-5 peritemperminReaction: x35 => x15 + x38, Rate Law: k32*x35-kr32*x15*x38
kr23 = 17.39321 permin; k23 = 420.3359 perminReaction: x32 => x33, Rate Law: k23*x32-kr23*x33
kr44 = 0.5985189 permin; k44 = 0.001406622 peritemperminReaction: x47 + x45 => x48, Rate Law: k44*x47*x45-kr44*x48
k55 = 1120.398 perminReaction: x82 => x83 + x77, Rate Law: k55*x82
k57 = 19.75184 perminReaction: x84 => x81 + x60, Rate Law: k57*x84
kr35 = 3.866434E-4 peritempermin; k35 = 1.836058 perminReaction: x30 => x24 + x22, Rate Law: k35*x30-kr35*x24*x22

States:

NameDescription
x85[Phosphoprotein; Mitogen-activated protein kinase 1; MI:0501]
x89[GDP; Pro-epidermal growth factor; Epidermal growth factor receptor; Ras GTPase-activating protein 1; Growth factor receptor-bound protein 2; Son of sevenless homolog 1; GTPase HRas; AP-type membrane coat adaptor complex]
x93[GDP; Pro-epidermal growth factor; Epidermal growth factor receptor; Ras GTPase-activating protein 1; SHC-transforming protein 2; Growth factor receptor-bound protein 2; Son of sevenless homolog 1; GTPase HRas; AP-type membrane coat adaptor complex]
x62[Phosphoprotein; Mitogen-activated protein kinase 1; MI:0501]
x26[GDP; GTPase HRas]
x45[RAF proto-oncogene serine/threonine-protein kinase]
x66[Son of sevenless homolog 1; Growth factor receptor-bound protein 2; SHC-transforming protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x91[Pro-epidermal growth factor; Epidermal growth factor receptor; Ras GTPase-activating protein 1; SHC-transforming protein 2; Growth factor receptor-bound protein 2; AP-type membrane coat adaptor complex]
x59[Mitogen-activated protein kinase 1; Phosphoprotein]
x44[MI:0501]
x61[Phosphoprotein; Mitogen-activated protein kinase 1; MI:0501]
x50[Phosphoprotein; RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
x31[SHC-transforming protein 2; 605217]
x33[SHC-transforming protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x47[Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase kinase 1Mitogen-activated protein kinase kinase 1, isoform CRA_acDNA FLJ76051, highly similar to Homo sapiens mitogen-activated protein kinase kinase 1 (MAP2K1), mRNA; 176872]
x92[Pro-epidermal growth factor; Epidermal growth factor receptor; Ras GTPase-activating protein 1; SHC-transforming protein 2; Growth factor receptor-bound protein 2; Son of sevenless homolog 1; AP-type membrane coat adaptor complex]
x8[Epidermal growth factor receptor; Pro-epidermal growth factor]
x28[GTP; GTPase HRas]
x35[Son of sevenless homolog 1; Growth factor receptor-bound protein 2; SHC-transforming protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x43[GTP; GTPase HRas]
x64[SHC-transforming protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x36[GDP; GTPase HRas; Son of sevenless homolog 1; Growth factor receptor-bound protein 2; SHC-transforming protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x4[EGF:EGFR dimer [plasma membrane]; Pro-epidermal growth factor; Epidermal growth factor receptor]
x32[SHC-transforming protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x27[GDP; GTPase HRas; Son of sevenless homolog 1; Growth factor receptor-bound protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x60[MI:0501]
x30[Son of sevenless homolog 1; Growth factor receptor-bound protein 2]
x58[Phosphoprotein; Mitogen-activated protein kinase 1; Dual specificity mitogen-activated protein kinase kinase 1]
x84[Phosphoprotein; Mitogen-activated protein kinase 1; MI:0501]
x88[Pro-epidermal growth factor; Epidermal growth factor receptor; Growth factor receptor-bound protein 2; Ras GTPase-activating protein 1; Son of sevenless homolog 1; AP-type membrane coat adaptor complex]
x34[Growth factor receptor-bound protein 2; SHC-transforming protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x51[Dual specificity mitogen-activated protein kinase kinase 1; Phosphoprotein]
x94[GTP; Pro-epidermal growth factor; Epidermal growth factor receptor; Ras GTPase-activating protein 1; SHC-transforming protein 2; Growth factor receptor-bound protein 2; Son of sevenless homolog 1; GTPase HRas; AP-type membrane coat adaptor complex]
x29[GTP; GTPase HRas; Son of sevenless homolog 1; Growth factor receptor-bound protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x53[MI:0501]
x37[GTP; GTPase HRas; Son of sevenless homolog 1; Growth factor receptor-bound protein 2; SHC-transforming protein 2; Ras GTPase-activating protein 1; Epidermal growth factor receptor; Pro-epidermal growth factor]
x5[Phosphoprotein; Pro-epidermal growth factor; Epidermal growth factor receptor]
x63[SHC-transforming protein 2; Ras GTPase-activating protein 1; Pro-epidermal growth factor; Epidermal growth factor receptor]
x48[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
x52[Phosphoprotein; Protein phosphatase 1 regulatory subunit 12C; Dual specificity mitogen-activated protein kinase kinase 1]
x83[Mitogen-activated protein kinase 1; Phosphoprotein]
x49[Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase kinase 1Mitogen-activated protein kinase kinase 1, isoform CRA_acDNA FLJ76051, highly similar to Homo sapiens mitogen-activated protein kinase kinase 1 (MAP2K1), mRNA; Phosphoprotein; 176872]
x65[Growth factor receptor-bound protein 2; SHC-transforming protein 2; Ras GTPase-activating protein 1; Pro-epidermal growth factor; Epidermal growth factor receptor]

Jenner2018 - treatment of oncolytic virus: BIOMD0000000789v0.0.1

The paper describes a model of oncolytic virotherapy. Created by COPASI 4.26 (Build 213) This model is described in t…

Details

Oncolytic virotherapy is an experimental cancer treatment that uses genetically engineered viruses to target and kill cancer cells. One major limitation of this treatment is that virus particles are rapidly cleared by the immune system, preventing them from arriving at the tumour site. To improve virus survival and infectivity Kim et al. (Biomaterials 32(9):2314-2326, 2011) modified virus particles with the polymer polyethylene glycol (PEG) and the monoclonal antibody herceptin. Whilst PEG modification appeared to improve plasma retention and initial infectivity, it also increased the virus particle arrival time. We derive a mathematical model that describes the interaction between tumour cells and an oncolytic virus. We tune our model to represent the experimental data by Kim et al. (2011) and obtain optimised parameters. Our model provides a platform from which predictions may be made about the response of cancer growth to other treatment protocols beyond those in the experiments. Through model simulations, we find that the treatment protocol affects the outcome dramatically. We quantify the effects of dosage strategy as a function of tumour cell replication and tumour carrying capacity on the outcome of oncolytic virotherapy as a treatment. The relative significance of the modification of the virus and the crucial role it plays in optimising treatment efficacy are explored. link: http://identifiers.org/pubmed/29644518

Parameters:

NameDescription
a = 0.0 1; di = 0.0 1/dReaction: => V; I, Rate Law: tme*burst(a, di, I)
di = 0.0 1/dReaction: I =>, Rate Law: tme*di*I
r = 0.037 1/d; L = 3.49E9 1Reaction: => S, Rate Law: tme*tg(r, L, S)
b = 0.0 1/dReaction: S => I; V, T, Rate Law: tme*inf(b, S, V, T)
dv = 0.0 1/dReaction: V =>, Rate Law: tme*dv*V

States:

NameDescription
S[neoplastic cell]
I[neoplastic cell]
T[neoplastic cell]
V[Oncolytic Virus]

Jenner2019 - Oncolytic virotherapy for tumours following a Gompertz growth law: BIOMD0000000850v0.0.1

This is a mathematical model using a Gompertz growth law to describe the in vivo dynamics of a cancer under treatment wi…

Details

Oncolytic viruses are genetically engineered to treat growing tumours and represent a very promising therapeutic strategy. Using a Gompertz growth law, we discuss a model that captures the in vivo dynamics of a cancer under treatment with an oncolytic virus. With the aid of local stability analysis and bifurcation plots, the typical interactions between virus and tumour are investigated. The system shows a singular equilibrium and a number of nonlinear behaviours that have interesting biological consequences, such as long-period oscillations and bistable states where two different outcomes can occur depending on the initial conditions. Complete tumour eradication appears to be possible only for parameter combinations where viral characteristics match well with the tumour growth rate. Interestingly, the model shows that therapies with a high initial injection or involving a highly effective virus do not universally result in successful strategies for eradication. Further, the use of additional, "boosting" injection schedules does not always lead to complete eradication. Our framework, instead, suggests that low viral loads can be in some cases more effective than high loads, and that a less resilient virus can help avoid high amplitude oscillations between tumours and virus. Finally, the model points to a number of interesting findings regarding the role of oscillations and bistable states between a tumour and an oncolytic virus. Strategies for the elimination of such fluctuations depend strongly on the initial viral load and the combination of parameters describing the features of the tumour and virus. link: http://identifiers.org/pubmed/31400344

Parameters:

NameDescription
m = 0.1; K = 100.0Reaction: => U, Rate Law: compartment*m*ln(K/U)*U
xi = 0.01Reaction: I => V, Rate Law: compartment*xi*I
gamma = 0.1Reaction: V =>, Rate Law: compartment*gamma*V

States:

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

Jerby2010_Liver_Metabolism: MODEL1009150002v0.0.1

This is the genome-scale metabolic network described in the article: Computational reconstruction of tissue-specific m…

Details

The computational study of human metabolism has been advanced with the advent of the first generic (non-tissue specific) stoichiometric model of human metabolism. In this study, we present a new algorithm for rapid reconstruction of tissue-specific genome-scale models of human metabolism. The algorithm generates a tissue-specific model from the generic human model by integrating a variety of tissue-specific molecular data sources, including literature-based knowledge, transcriptomic, proteomic, metabolomic and phenotypic data. Applying the algorithm, we constructed the first genome-scale stoichiometric model of hepatic metabolism. The model is verified using standard cross-validation procedures, and through its ability to carry out hepatic metabolic functions. The model's flux predictions correlate with flux measurements across a variety of hormonal and dietary conditions, and improve upon the predictive performance obtained using the original, generic human model (prediction accuracy of 0.67 versus 0.46). Finally, the model better predicts biomarker changes in genetic metabolic disorders than the generic human model (accuracy of 0.67 versus 0.59). The approach presented can be used to construct other human tissue-specific models, and be applied to other organisms. link: http://identifiers.org/pubmed/20823844

Jesty1993_ProteolyticPositiveFeedback: MODEL1108260010v0.0.1

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

Details

A model of a proteolytic positive-feedback loop, similar in general terms to feedback loops that occur in blood coagulation and other systems, has been examined by both explicit and numerical analysis. In this loop, modeled as a closed system, each enzyme (E1, E2) catalyzes the formation of the other from its respective zymogen (Z1, Z2), and both enzymes are subject to irreversible inhibition. The system shows three major characteristics. (1) No significant Z1 or Z2 activation occurs unless the combination of initial conditions and kinetic parameters is above a threshold level. This threshold occurs when the product of the enzyme generation rates equals the product of their inhibition rates. When the formation-rate product is less than the inhibition-rate product, there is no response: E1 and E2 generation is minimal and the lag time is effectively infinite. Conversely, when the generation-rate product exceeds the inhibition-rate product, explosive formation of both E1 and E2 is seen. For responses exceeding the threshold, the following obtain. (2) The lag time in E1 and E2 generation is a highly nonlinear function of the zymogen concentrations and the enzyme generation and inhibition rates. In contrast, there is a simple logarithmic relationship between the lag time and the initial trace concentration of the enzyme that is responsible for initiating the system; in this model, E1. (3) The extent of Z1 and Z2 activation is similarly a nonlinear function of the conditions and parameters but is independent of the initiating trace level of E1.(ABSTRACT TRUNCATED AT 250 WORDS) link: http://identifiers.org/pubmed/8512937

Jiang2007 - GSIS system, Pancreatic Beta Cells: BIOMD0000000239v0.0.1

Jiang2007 - GSIS system, Pancreatic Beta CellsDescription of a core kinetic model of the glucose-stimulated insulin secr…

Details

The construction and characterization of a core kinetic model of the glucose-stimulated insulin secretion system (GSIS) in pancreatic beta cells is described. The model consists of 44 enzymatic reactions, 59 metabolic state variables, and 272 parameters. It integrates five subsystems: glycolysis, the TCA cycle, the respiratory chain, NADH shuttles, and the pyruvate cycle. It also takes into account compartmentalization of the reactions in the cytoplasm and mitochondrial matrix. The model shows expected behavior in its outputs, including the response of ATP production to starting glucose concentration and the induction of oscillations of metabolite concentrations in the glycolytic pathway and in ATP and ADP concentrations. Identification of choke points and parameter sensitivity analysis indicate that the glycolytic pathway, and to a lesser extent the TCA cycle, are critical to the proper behavior of the system, while parameters in other components such as the respiratory chain are less critical. Notably, however, sensitivity analysis identifies the first reactions of nonglycolytic pathways as being important for the behavior of the system. The model is robust to deletion of malic enzyme activity, which is absent in mouse pancreatic beta cells. The model represents a step toward the construction of a model with species-specific parameters that can be used to understand mouse models of diabetes and the relationship of these mouse models to the human disease state. link: http://identifiers.org/pubmed/17514510

Parameters:

NameDescription
K1ATP=6.3E-5; V1=5.0E-4; K1GLC=1.0E-4Reaction: GLC + ATP_cyt => F6P + ADP_cyt, Rate Law: CYTOPLASM*V1*ATP_cyt*GLC/((K1GLC+GLC)*(K1ATP+ATP_cyt))
kminus2=1400.0; k4=214.0; v31_MDH=3.8617E-7; k3=4650.0; k2=3.5E7; kminus1=26.0; kminus4=260000.0; kminus3=570000.0; k1=3.4E7Reaction: NADH_cyt + OXA_cyt => Mal_cyt + NAD, Rate Law: CYTOPLASM*v31_MDH*(k1*k2*k3*k4*NADH_cyt*OXA_cyt-kminus1*kminus2*kminus3*kminus4*Mal_cyt*NAD)/(kminus1*(kminus2+k3)*k4+k1*(kminus2+k3)*k4*NADH_cyt+kminus1*(kminus2+k3)*kminus4*NAD+k2*k3*k4*OXA_cyt+kminus1*kminus2*kminus3*Mal_cyt+k1*k2*(k3+k4)*NADH_cyt*OXA_cyt+(kminus1+kminus2)*kminus3*kminus4*Mal_cyt*NAD+k1+kminus2+kminus3*NADH_cyt*Mal_cyt+k1*k2*kminus3*NADH_cyt*OXA_cyt*Mal_cyt+k2*k3*kminus4*OXA_cyt*NAD+k2*kminus3*kminus4*OXA_cyt*Mal_cyt*NAD)
KcR=31.44; Ks=5.0E-4; v11_ACO=3.8617E-7; KcF=20.47; Kp=1.1E-4Reaction: Cit => IsoCit, Rate Law: MATRIX*(KcF*Kp*Cit-KcR*Ks*IsoCit)*v11_ACO/(Ks*IsoCit+Kp*Cit+Ks*Kp)
KmQ=7.5E-6; KmC=4.5E-4; Kib=2.0E-5; KmP=6.0E-4; Kip=0.07; Kiq=5.0E-6; Kir=6.7E-6; v15_SCS=3.8617E-7; KmB=3.5E-5; Kc2=100.0; KmA=5.0E-6; KmP2=6.0E-4; Kia=4.0E-4; Keq=8.375; Kic=3.0E-5; Kc1=100.0; KmC2=4.5E-4Reaction: GDP + SCoA + Pi => Suc + GTP + CoA, Rate Law: MATRIX*(GDP*SCoA*pi-Suc*GTP*CoA/Keq)*(Kc1*v15_SCS+Kc2*v15_SCS*(KmC*Suc/KmC2*Kip+pi/KmC2))/(Kia*KmB*pi+KmB*GDP*pi+KmA*SCoA*pi+KmC*GDP*SCoA+GDP*SCoA*pi+GDP*SCoA*pi*pi/KmC2+Kia*KmB*KmC*Suc/Kip+Kia*KmB*KmC*Suc*GTP/Kip/Kiq+Kia*KmB*KmC*Suc*CoA/Kip/Kir+Kia*KmB*Kic*GTP*CoA/KmQ/Kir+Kia*KmB*KmC*Suc*GTP*CoA/Kip/KmQ/Kir+Kia*KmB*KmC*Suc*Suc*GTP*CoA/Kip/KmP2/KmQ/Kir+Kia*KmB*pi*GTP/Kiq+Kia*KmB*pi*CoA/Kir+Kia*KmB*pi*GTP*CoA/KmQ/Kir+Kia*KmB*pi*Suc*GTP*CoA/KmP2/KmQ/Kir+KmB*KmC*GDP*Suc/Kip+KmA*KmC*SCoA*Suc/Kip+KmC*GDP*SCoA*Suc/Kip+KmC*GDP*SCoA*pi*Suc/KmC2/Kip+KmA*SCoA*pi*GTP/Kiq+KmB*GDP*pi*CoA/Kir+KmA*KmC*SCoA*Suc*GTP/Kip/Kiq+KmB*KmC*GDP*Suc*CoA/Kip/Kir)
KmS1=9.0E-4; v32_AspTA=3.8617E-7; KmP2=0.004; Keq=6.2; KiP2=0.0083; KcF=300.0; KmP1=4.0E-5; KiS1=0.002; KcR=1000.0; KmS2=1.0E-4Reaction: Asp_cyt + OG_cyt => OXA_cyt + Glu_cyt, Rate Law: CYTOPLASM*KcF*KcR*v32_AspTA*(Asp_cyt*OG_cyt-OXA_cyt*Glu_cyt/Keq)/(KcR*KmS2*Asp_cyt+KcR*KmS1*OG_cyt+KcF*KmP2*OXA_cyt/Keq+KcF*KmP1*Glu_cyt/Keq+KcR*Asp_cyt*OG_cyt+KcF*KmP2*Asp_cyt*OXA_cyt/(Keq*KiS1)+KcF*OXA_cyt*Glu_cyt/Keq+KcR*KmS1*OG_cyt*Glu_cyt/KiP2)
KiP2=0.0028; v22_AGC=3.3211E-4; KiS2=0.0032; KcR=10.0; KcF=10.0; alpha=1.0; delta=1.0; KiS1=8.0E-5; KiP1=1.8E-4; beta=1.0; gamma=1.0Reaction: Glu_cyt + Asp => Asp_cyt + Glu, Rate Law: MATRIX*(Asp*Glu_cyt/alpha/KiS1/KiS2*KcF-Glu*Asp_cyt/beta/KiP1/KiP2*KcR)*v22_AGC/(1+Asp/KiS1+Glu_cyt/KiS2+Glu/KiP1+Asp_cyt/KiP2+Asp*Glu_cyt/alpha/KiS1/KiS2+Glu*Asp_cyt/beta/KiP1/KiP2+Glu_cyt*Asp_cyt/gamma/KiS2/KiP2+Asp*Glu/delta/KiS1/KiP1)
e=6.4E-4; v12_IDHa=3.8617E-7; b=29.6; f=3.6E-4; d=7.8E-5; KcF=105.0; c=2.3E-4Reaction: IsoCit + NAD_p => OG + NADH; ADP, Rate Law: MATRIX*KcF*v12_IDHa*(IsoCit*IsoCit+b*ADP*IsoCit)/(IsoCit*IsoCit+c*IsoCit+d*ADP+e*ADP*IsoCit+f)
Kiq=3.5E-5; KmP=5.9E-7; v9_PDC=3.8617E-7; Kic=1.8E-4; KmC=5.0E-5; KmA=2.5E-5; KcF=856.0; KmR=6.9E-7; KmB=1.3E-5; Kip=6.0E-5; Kib=3.0E-4; Kir=3.6E-5; Kia=5.5E-4Reaction: Pyr + CoA + NAD_p => CO2 + Acetyl_CoA + NADH, Rate Law: MATRIX*KcF*v9_PDC*Pyr*CoA*NAD_p/(KmC*Pyr*CoA+KmB*Pyr*NAD_p+KmA*CoA*NAD_p+Pyr*CoA*NAD_p+KmA*KmP*Kib*Kic/KmR/Kip/Kiq*Acetyl_CoA*NADH+KmC/Kir*Pyr*CoA*NADH+KmB/Kiq*Pyr*NAD_p*Acetyl_CoA+KmA*KmP*Kib*Kic/KmR/Kip/Kia/Kiq*Pyr*Acetyl_CoA*NADH)
V=3.99E-8; K=3.4E-5; v37_GUT2P=0.001Reaction: G3P + FAD => FADH2 + DHAP, Rate Law: CYTOPLASM*V*v37_GUT2P*G3P/(K+G3P)
KiP1=0.0014; KiS1=3.0E-4; KiS2=7.0E-4; KiP2=1.7E-4; alpha=1.0; delta=1.0; beta=1.0; gamma=1.0; v30_OGC=3.3211E-4; KcR=4.83; KcF=3.675Reaction: Mal_cyt + OG => OG_cyt + Mal, Rate Law: MATRIX*(OG*Mal_cyt/alpha/KiS1/KiS2*KcF-Mal*OG_cyt/beta/KiP1/KiP2*KcR)*v30_OGC/(1+OG/KiS1+Mal_cyt/KiS2+Mal/KiP1+OG_cyt/KiP2+OG*Mal_cyt/alpha/KiS1/KiS2+Mal*OG_cyt/beta/KiP1/KiP2+Mal_cyt*OG_cyt/gamma/KiS2/KiP2+OG*Mal/delta/KiS1/KiP1)
KmP1=1.08E-6; KiP2=1.19E-5; Keq=8.99; KiS1=7.6E-5; KiS2=2.4E-7; KcR=0.3; KmP2=2.42E-5; KcF=2.18; KmS1=3.9E-5; v35_ACD=3.3211E-5; KmS2=1.2E-7; KiP1=7.53E-5Reaction: FADH2 + ETFox => ETFred + FAD, Rate Law: MATRIX*KcF*KcR*v35_ACD*(FADH2*ETFox-ETFred*FAD/Keq)/(KcR*KiS1*KmS2+KcR*KmS2*FADH2+KcR*KmS1*ETFox+KcF*KmP2*ETFred/Keq+KcF*KmP1*FAD/Keq+KcR*FADH2*ETFox+KcF*KmP2*FADH2*ETFred/(Keq*KiS1)+KcF*ETFred*FAD/Keq+KcR*KmS1*ETFox*FAD/KiP2+KcR*FADH2*ETFox*ETFred/KiP1+KcF*ETFox*ETFred*FAD/(KiS2*Keq))
KiS2=4.4E-4; v42_CIC=3.3211E-4; KiS1=1.3E-4; KcF=5.6; KcR=3.5; alpha=1.0; delta=1.0; beta=1.0; gamma=1.0; KiP1=3.3E-4; KiP2=4.18E-5Reaction: IsoCitcyt + Mal => Mal_cyt + IsoCit, Rate Law: MATRIX*(IsoCitcyt*Mal/alpha/KiS1/KiS2*KcF-Mal_cyt*IsoCit/beta/KiP1/KiP2*KcR)*v42_CIC/(1+IsoCitcyt/KiS1+Mal/KiS2+Mal_cyt/KiP1+IsoCit/KiP2+IsoCitcyt*Mal/alpha/KiS1/KiS2+Mal_cyt*IsoCit/beta/KiP1/KiP2+Mal*IsoCit/gamma/KiS2/KiP2+IsoCitcyt*Mal_cyt/delta/KiS1/KiP1)
Kb=4.5E-6; v10_CS=3.8617E-7; Kia=5.0E-6; Kc=3.9E-5; Ka=5.0E-6; Keq=1.8E7; Kid=0.0043; V=0.004833; Kib=4.5E-6Reaction: Cit_cyt + CoA_cyt => OXA_cyt + Acetyl_CoA_cyt, Rate Law: CYTOPLASM*Kid*Kc*V*Acetyl_CoA_cyt*OXA_cyt*v10_CS/(Acetyl_CoA_cyt*OXA_cyt+Ka*OXA_cyt+Kb*Acetyl_CoA_cyt+Kia*Kib)/(Keq*Kia*Kb)
K4NAD=0.001; K4GAP=0.001; V4=0.01Reaction: GAP + NAD => DPG + NADH_cyt, Rate Law: CYTOPLASM*V4*NAD*GAP/((K4GAP+GAP)*(K4NAD+NAD))
KmP=3.0E-4; Kib=7.4E-4; KmR=6.0E-4; KmC=5.0E-5; Kic=1.0E-4; KmB=2.5E-5; Kia=7.2E-4; KmA=2.2E-4; Kir=2.5E-5; Kip=1.1E-6; Kiq=8.1E-5; v14_OGDC=3.8617E-7; KcF=177.0Reaction: OG + CoA + NAD_p => CO2 + SCoA + NADH, Rate Law: MATRIX*KcF*v14_OGDC*OG*CoA*NAD_p/(KmC*OG*CoA+KmB*OG*NAD_p+KmA*CoA*NAD_p+OG*CoA*NAD_p+KmA*KmP*Kib*Kic/KmR/Kip/Kiq*SCoA*NADH+KmC/Kir*OG*CoA*NADH+KmB/Kiq*OG*NAD_p*SCoA+KmA*KmP*Kib*Kic/KmR/Kip/Kia/Kiq*OG*SCoA*NADH)
v34_ETF_QO=3.3211E-5; KcR=101.0; KiS1=3.1E-7; KmS1=3.1E-7; KmP1=3.2E-7; Keq=0.66; KiP2=3.0E-7; KmP2=4.2E-9; KmS2=3.9E-7; KcF=78.0Reaction: ETFred + Q => ETFox + QH2, Rate Law: MATRIX*KcF*KcR*v34_ETF_QO*(ETFred*Q-ETFox*QH2/Keq)/(KcR*KmS2*ETFred+KcR*KmS1*Q+KcF*KmP2*ETFox/Keq+KcF*KmP1*QH2/Keq+KcR*ETFred*Q+KcF*KmP2*ETFred*ETFox/(Keq*KiS1)+KcF*ETFox*QH2/Keq+KcR*KmS1*Q*QH2/KiP2)
KmS1=9.0E-4; KmP2=0.004; Keq=6.2; KiP2=0.0083; KcF=300.0; KmP1=4.0E-5; KiS1=0.002; KcR=1000.0; KmS2=1.0E-4; v21_AspTA=3.8617E-7Reaction: OXA + Glu => Asp + OG, Rate Law: MATRIX*KcF*KcR*v21_AspTA*(OXA*Glu-Asp*OG/Keq)/(KcR*KmS2*OXA+KcR*KmS1*Glu+KcF*KmP2*Asp/Keq+KcF*KmP1*OG/Keq+KcR*OXA*Glu+KcF*KmP2*OXA*Asp/(Keq*KiS1)+KcF*Asp*OG/Keq+KcR*KmS1*Glu*OG/KiP2)
KmP1=3.0E-7; KmP2=1.5E-6; KmS1=3.0E-5; KiP2=5.6E-6; Keq=0.037; KiS1=4.1E-6; v16_SDH=9.9211E-5; KmS2=6.9E-5; KcR=1.73; KcF=69.3Reaction: Suc + Q => Fum + QH2, Rate Law: MATRIX*KcF*KcR*v16_SDH*(Suc*Q-Fum*QH2/Keq)/(KcR*KmS2*Suc+KcR*KmS1*Q+KcF*KmP2*Fum/Keq+KcF*KmP1*QH2/Keq+KcR*Suc*Q+KcF*KmP2*Suc*Fum/(Keq*KiS1)+KcF*Fum*QH2/Keq+KcR*KmS1*Q*QH2/KiP2)
KmS1=9.2E-6; KmP1=9.9E-6; KcF=498.0; v24_Complex_I=3.3211E-4; KmP2=5.9E-5; KcR=229.0; KiP2=9.8E-8; KiS1=2.1E-8; KmS2=2.6E-4; Keq=407.9Reaction: NADH + Q => NAD_p + QH2, Rate Law: MATRIX*KcF*KcR*v24_Complex_I*(NADH*Q-NAD_p*QH2/Keq)/(KcR*KmS2*NADH+KcR*KmS1*Q+KcF*KmP2*NAD_p/Keq+KcF*KmP1*QH2/Keq+KcR*NADH*Q+KcF*KmP2*NADH*NAD_p/(Keq*KiS1)+KcF*NAD_p*QH2/Keq+KcR*KmS1*Q*QH2/KiP2)
Knadp=0.011; Kcat=0.333; Kmal=1.25E-4; v39_MDH=3.8617E-7Reaction: Mal_cyt + NADP_cyt => NADPH_cyt + PYR_cyt, Rate Law: CYTOPLASM*v39_MDH*Kcat*Mal_cyt*NADP_cyt/((Kmal+Mal_cyt)*(Knadp+NADP_cyt))
KiS1=0.0087; Keq=0.69; KmS2=4.0E-4; KmP2=4.0E-4; KcR=0.15; KcF=337.0; KiP2=0.012; v20_AlaTA=3.8617E-7; KmS1=0.002; KmP1=0.032Reaction: Ala + OG => Glu + Pyr, Rate Law: MATRIX*KcF*KcR*v20_AlaTA*(Ala*OG-Glu*Pyr/Keq)/(KcR*KmS2*Ala+KcR*KmS1*OG+KcF*KmP2*Glu/Keq+KcF*KmP1*Pyr/Keq+KcR*Ala*OG+KcF*KmP2*Ala*Glu/(Keq*KiS1)+KcF*Glu*Pyr/Keq+KcR*KmS1*OG*Pyr/KiP2)
k3b=0.05; k3f=1.0Reaction: FBP => GAP, Rate Law: CYTOPLASM*(k3f*FBP-k3b*GAP^2)
Kib=4.0E-6; v10_CS=3.8617E-7; V=0.005267; Kia=1.0E-5; Ka=1.18E-5; Kb=4.8E-6Reaction: OXA + Acetyl_CoA => Cit + CoA, Rate Law: MATRIX*V*Acetyl_CoA*OXA*v10_CS/(Acetyl_CoA*OXA+Ka*OXA+Kb*Acetyl_CoA+Kia*Kib)
KiS2=4.4E-4; KiS1=1.3E-4; KcF=5.6; KcR=3.5; alpha=1.0; delta=1.0; v33_CIC=3.3211E-4; beta=1.0; gamma=1.0; KiP1=3.3E-4; KiP2=4.18E-5Reaction: Cit_cyt + Mal => Mal_cyt + Cit, Rate Law: MATRIX*(Cit_cyt*Mal/alpha/KiS1/KiS2*KcF-Mal_cyt*Cit/beta/KiP1/KiP2*KcR)*v33_CIC/(1+Cit_cyt/KiS1+Mal/KiS2+Mal_cyt/KiP1+Cit/KiP2+Cit_cyt*Mal/alpha/KiS1/KiS2+Mal_cyt*Cit/beta/KiP1/KiP2+Mal*Cit/gamma/KiS2/KiP2+Cit_cyt*Mal_cyt/delta/KiS1/KiP1)
k9b=10000.0; k9f=10000.0Reaction: AMP + ATP_cyt => ADP_cyt, Rate Law: CYTOPLASM*(k9f*AMP*ATP_cyt-k9b*ADP_cyt^2)
V=0.0399; K=34.0; v38_GUT2P=0.001Reaction: NADH_cyt + DHAP => G3P + NAD, Rate Law: CYTOPLASM*V*v38_GUT2P*NADH_cyt/(K+NADH_cyt)
k8f=1000.0; k8b=0.143Reaction: PYR_cyt + NADH_cyt => LAC + NAD, Rate Law: CYTOPLASM*(k8f*NADH_cyt*PYR_cyt-k8b*NAD*LAC)
k2=0.017; K2ATP=1.0E-5; V2=0.0015; K2=1.6E-9Reaction: F6P + ATP_cyt => FBP + ADP_cyt; AMP, Rate Law: CYTOPLASM*V2*ATP_cyt*F6P^2/((K2*(1+k2*(ATP_cyt/AMP)^2)+F6P^2)*(K2ATP+ATP_cyt))
KcR=20.0; KmB=0.00163; KcF=200.0; Kic=1.3E-4; Kia=1.5E-4; v36_PC=3.8617E-7; Kiq=1.9E-4; Kib=0.0016; Keq=9.0; KmR=5.1E-5; KmC=3.7E-4; KmQ=2.4E-4; Kip=0.0079; Kir=2.4E-4; KmP=0.016; KmA=1.1E-4Reaction: ATP + CO2 + Pyr => Pi + ADP + OXA, Rate Law: MATRIX*KcF*KcR*v36_PC*(ATP*CO2*Pyr-pi*ADP*OXA/Keq)/(Kia*KmB*KcR*Pyr+KmC*KcR*ATP*CO2+KmA*KcR*CO2*Pyr+KmB*KcR*ATP*Pyr+KcR*ATP*CO2*Pyr+Kip*KmQ*KcF*OXA/Keq+KmQ*KcF*pi*OXA/Keq+KmP*KcF*ADP*OXA/Keq+KmR*KcF*pi*ADP/Keq+KcF*pi*ADP*OXA/Keq+Kia*KmB*KcR*Pyr*pi/Kip+Kia*KmB*KcR*Pyr*ADP/Kia+Kiq*KmP*KcF*CO2*OXA/Kib/Keq+Kia*KmP*KcF*ATP*OXA/Kia/Keq+KmA*KcR*ATP*CO2*OXA/Kir+KmR*KcF*Pyr*pi*ADP/Kic/Keq+KmA*KcR*CO2*Pyr*ADP/Kiq+KmA*KcR*CO2*Pyr*pi/Kip+KmP*KcF*CO2*ADP*OXA/Kib/Keq+KmQ*KcF*CO2*pi*OXA/Kib/Keq)
Kp=2.5E-5; Ks=5.0E-6; KcF=800.0; v17_FM=3.8617E-7; KcR=900.0Reaction: Fum => Mal, Rate Law: MATRIX*(KcF*Kp*Fum-KcR*Ks*Mal)*v17_FM/(Ks*Mal+Kp*Fum+Ks*Kp)
Km=0.01295; Kcat=130.5; v44_MDH=3.8617E-7Reaction: Mal + NADP_p => NADPH + Pyr, Rate Law: MATRIX*v44_MDH*Kcat*Mal/(Km+Mal)
KcR=31.44; v29_ACO=3.8617E-7; Ks=5.0E-4; KcF=20.47; Kp=1.1E-4Reaction: Cit_cyt => IsoCitcyt, Rate Law: CYTOPLASM*(KcF*Kp*Cit_cyt-KcR*Ks*IsoCitcyt)*v29_ACO/(Ks*IsoCitcyt+Kp*Cit_cyt+Ks*Kp)
v40_AAC=3.3211E-4; V=0.1667; K=0.012Reaction: ADP_cyt => ADP, Rate Law: MATRIX*V*v40_AAC*ADP_cyt/(K+ADP_cyt)
KiP2=0.0019; KmP2=1.7E-4; KcF=0.39; KcR=0.04; v18_MDH=3.8617E-7; KmP1=0.0016; KiS1=1.1E-5; KiP1=0.0071; KmS1=7.2E-5; KiS2=1.0E-4; KmS2=1.1E-4Reaction: Mal + NAD_p => NADH + OXA, Rate Law: MATRIX*(KcF*Mal*NAD_p/KiS1/KmS2-KcR*OXA*NADH/KmP1/KiP2)*v18_MDH/(1+Mal/KiS1+KmS1*NAD_p/KiS1/KmS2+KmP2*OXA/KmP1/KiP2+NADH/KiP2+Mal*NAD_p/KiS1/KmS2+KmP2*Mal*OXA/KiS1/KmP1/KiP2+KmS1*NAD_p*NADH/KiS1/KmS2/KiP2+OXA*NADH/KmP1*KiP2+Mal*NAD_p*OXA/KiS1/KmS2/KiP1+NAD_p*OXA*NADH/KiS2/KmP1/KiP2)
k5b=500.0; k5f=1000.0Reaction: DPG + ADP_cyt => PEP + ATP_cyt, Rate Law: CYTOPLASM*(k5f*DPG*ADP_cyt-k5b*PEP*ATP_cyt)
flow = 0.011Reaction: LAC =>, Rate Law: CYTOPLASM*LAC*flow
v41_IDHc=3.8617E-7; phir13=1.3E-10; phir123=4.6E-14; phi12=9.0E-8; phi0=0.051; phir23=9.4E-8; phi2=9.6E-7; phir1=3.7E-7; phir12=6.0E-12; phir2=2.9E-5; phir3=2.5E-4; phir0=0.066; phi1=9.5E-8Reaction: IsoCitcyt + NADP_cyt => OG_cyt + NADPH_cyt; CO2, Rate Law: CYTOPLASM*v41_IDHc*(IsoCitcyt*NADP_cyt/(phi0*IsoCitcyt*NADP_cyt+phi1*NADP_cyt+phi2*IsoCitcyt+phi12)-OG_cyt*NADPH_cyt*CO2/(phir0*OG_cyt*NADPH_cyt*CO2+phir1*NADPH_cyt*CO2+phir2*OG_cyt*CO2+phir3*OG_cyt*NADPH_cyt+phir12*CO2+phir13*NADPH_cyt+phir23*OG_cyt+phir123))
V=0.075; v28_Complex_V=0.0033211; Km=0.0045; Ki=0.047Reaction: ADP + Pi => ATP + H2O, Rate Law: MATRIX*v28_Complex_V*V*ADP/(Km+ADP+ADP*ADP/Ki)
KmB=3.0E-6; Kb2=5.7E-6; KcF=426.8; Kq2=1.9E-6; v25_Complex_III=9.963E-9; Kq1=2.8E-6; Kb1=5.4E-6; KmA=2.8E-5; k8=622.1Reaction: QH2 + Cytc3p => Q + Cytc2p, Rate Law: MT_IMS*KcF*v25_Complex_III*QH2*Cytc3p/((KmA*Kq2*Kb2+KmA*Kq2*Cytc3p+KcF/k8*Kq1*QH2*Kb1+KcF/k8*Kq1*QH2*Cytc3p)*Cytc2p+KmA*Cytc3p+KmB*QH2+QH2*Cytc3p)

States:

NameDescription
G3P[sn-Glycerol 3-phosphate; sn-glycerol 3-phosphate]
Cit[Citrate; citric acid]
Ala[L-Alanine; L-alanine]
SCoA[Succinyl-CoA; succinyl-CoA]
Glu cyt[L-Glutamate; L-glutamic acid]
GDP[GDP; GDP]
OXA[Oxaloacetate; oxaloacetic acid]
FADH2[FADH2; FADH2]
IsoCitcyt[Isocitrate; isocitric acid]
Asp[L-Aspartate; L-aspartic acid]
OG cyt[2-Oxoglutarate; 2-oxoglutaric acid]
QH2[ubiquinol; Ubiquinol]
GAP[D-Glyceraldehyde 3-phosphate; D-glyceraldehyde 3-phosphate]
Acetyl CoA cyt[Acetyl-CoA; acetyl-CoA]
ATP cyt[C0002]
ETFox[Electron-transferring flavoprotein]
PEP[Phosphoenolpyruvate; phosphoenolpyruvate]
NAD[NAD(+); NAD+]
H2O[H2O; water]
ADP[C0008]
OG[2-Oxoglutarate; 2-oxoglutaric acid]
Q[ubiquinones; Ubiquinone]
DPG[3-Phospho-D-glyceroyl phosphate; 3-phospho-D-glyceroyl dihydrogen phosphate]
ATP[C0002]
AMP[AMP; AMP]
DHAP[Glycerone phosphate; dihydroxyacetone phosphate]
GTP[GTP; GTP]
Mal[(S)-Malate; (S)-malic acid]
NAD p[NAD+; NAD(+)]
NADH cyt[NADH; NADH]
Mal cyt[(S)-Malate; (S)-malic acid]
Glu[L-Glutamate; L-glutamic acid]
Fum[Fumarate; fumaric acid]
IsoCit[Isocitrate; isocitric acid]
Suc[Succinate; succinic acid]
OXA cyt[Oxaloacetate; oxaloacetic acid]
CoA[CoA; coenzyme A]
NADH[NADH; NADH]
CoA cyt[CoA; coenzyme A]
LAC[(S)-Lactate; (S)-lactic acid]
Pi[Orthophosphate; phosphate(3-)]
Cit cyt[Citrate; citric acid]
FAD[FAD; FAD]
Asp cyt[L-Aspartate; L-aspartic acid]
ETFred[Reduced electron-transferring flavoprotein]
PYR cyt[Pyruvate; pyruvic acid]
Acetyl CoA[Acetyl-CoA; acetyl-CoA]

Jiao2018 - Feedback regulation in a stem cell model with acute myeloid leukaemia: BIOMD0000000898v0.0.1

This is a mathematical model describing the hematopoietic lineages with leukemia lineages, as controlled by end-product…

Details

BACKGROUND:The haematopoietic lineages with leukaemia lineages are considered in this paper. In particular, we mainly consider that haematopoietic lineages are tightly controlled by negative feedback inhibition of end-product. Actually, leukemia has been found 100 years ago. Up to now, the exact mechanism is still unknown, and many factors are thought to be associated with the pathogenesis of leukemia. Nevertheless, it is very necessary to continue the profound study of the pathogenesis of leukemia. Here, we propose a new mathematical model which include some negative feedback inhibition from the terminally differentiated cells of haematopoietic lineages to the haematopoietic stem cells and haematopoietic progenitor cells in order to describe the regulatory mechanisms mentioned above by a set of ordinary differential equations. Afterwards, we carried out detailed dynamical bifurcation analysis of the model, and obtained some meaningful results. RESULTS:In this work, we mainly perform the analysis of the mathematic model by bifurcation theory and numerical simulations. We have not only incorporated some new negative feedback mechanisms to the existing model, but also constructed our own model by using the modeling method of stem cell theory with probability method. Through a series of qualitative analysis and numerical simulations, we obtain that the weak negative feedback for differentiation probability is conducive to the cure of leukemia. However, with the strengthening of negative feedback, leukemia will be more difficult to be cured, and even induce death. In contrast, strong negative feedback for differentiation rate of progenitor cells can promote healthy haematopoiesis and suppress leukaemia. CONCLUSIONS:These results demonstrate that healthy progenitor cells are bestowed a competitive advantage over leukaemia stem cells. Weak g1, g2, and h1 enable the system stays in the healthy state. However, strong h2 can promote healthy haematopoiesis and suppress leukaemia. link: http://identifiers.org/pubmed/29745850

Parameters:

NameDescription
v_1_D = 0.5; Z_1 = 10.0; p_1_D = 0.45; K_1 = 1.0Reaction: => S_HSC, Rate Law: compartment*p_1_D*(K_1-Z_1)*v_1_D*S_HSC
v_1_D = 0.5; p_1_D = 0.45Reaction: S_HSC => A_PC, Rate Law: compartment*(1-p_1_D)*v_1_D*S_HSC
p_2_D = 0.68; K_2 = 1.0; v_2_D = 0.72; Z_2 = 10.0Reaction: => A_PC, Rate Law: compartment*p_2_D*(K_2-Z_2)*v_2_D*A_PC
p_30 = 0.8; v_30 = 0.7Reaction: L_LSC => T_TDLC, Rate Law: compartment*(1-p_30)*v_30*L_LSC
p_2_D = 0.68; v_2_D = 0.72Reaction: A_PC => D_TDSC, Rate Law: compartment*(1-p_2_D)*v_2_D*A_PC
d_2 = 0.3Reaction: T_TDLC =>, Rate Law: compartment*d_2*T_TDLC
p_30 = 0.8; v_30 = 0.7; K_2 = 1.0; Z_2 = 10.0Reaction: => L_LSC, Rate Law: compartment*p_30*(K_2-Z_2)*v_30*L_LSC
d_1 = 0.275Reaction: D_TDSC =>, Rate Law: compartment*d_1*D_TDSC

States:

NameDescription
S HSC[C12551]
A PC[C12662]
T TDLC[EFO:0002954; C41069]
L LSC[C41069]
D TDSC[C12551; EFO:0002954]

Jol2012_YeastMetabolism_EFManalysis: MODEL1201230000v0.0.1

To the extent possible under law, all copyright and related or neighbouring rights to this encoded model have been dedic…

Details

One of the most obvious phenotypes of a cell is its metabolic activity, which is defined by the fluxes in the metabolic network. Although experimental methods to determine intracellular fluxes are well established, only a limited number of fluxes can be resolved. Especially in eukaryotes such as yeast, compartmentalization and the existence of many parallel routes render exact flux analysis impossible using current methods. To gain more insight into the metabolic operation of S. cerevisiae we developed a new computational approach where we characterize the flux solution space by determining elementary flux modes (EFMs) that are subsequently classified as thermodynamically feasible or infeasible on the basis of experimental metabolome data. This allows us to provably rule out the contribution of certain EFMs to the in vivo flux distribution. From the 71 million EFMs in a medium size metabolic network of S. cerevisiae, we classified 54% as thermodynamically feasible. By comparing the thermodynamically feasible and infeasible EFMs, we could identify reaction combinations that span the cytosol and mitochondrion and, as a system, cannot operate under the investigated glucose batch conditions. Besides conclusions on single reactions, we found that thermodynamic constraints prevent the import of redox cofactor equivalents into the mitochondrion due to limits on compartmental cofactor concentrations. Our novel approach of incorporating quantitative metabolite concentrations into the analysis of the space of all stoichiometrically feasible flux distributions allows generating new insights into the system-level operation of the intracellular fluxes without making assumptions on metabolic objectives of the cell. link: http://identifiers.org/pubmed/22416224

Jones1994_BloodCoagulation: BIOMD0000000336v0.0.1

Jones1994_BloodCoagulationThis model is built based on the experimental findings described in Lawson et al., 1994 (PMID:…

Details

A mathematical simulation of the tissue factor pathway to the generation of thrombin has been developed using a combination of empirical, estimated, and deduced rate constants for reactions involving the activation of factor IX, X, V, and VIII, in the formation of thrombin, as well as rate constants for the assembly of the coagulation enzyme complexes which involve factor VIIIa-factor IXa (intrinsic tenase) and factor Va-Xa (prothrombinase) assembled on phospholipid membrane. Differential equations describing the fate of each species in the reaction were developed and solved using an interactive procedure based upon the Runge-Kutta technique. In addition to the theoretical considerations involving the reactions of the tissue factor pathway, a physical constraint associated with the stability of the factor VIIIa-factor IXa complex has been incorporated into the model based upon the empirical observations associated with the stability of this complex. The model system provides a realistic accounting of the fates of each of the proteins in the coagulation reaction through a range of initiator (factor VIIa-tissue factor) concentrations ranging from 5 pM to 5 nM. The model is responsive to alterations in the concentrations of factor VIII, factor V, and their respective activated species, factor VIIIa and factor Va, and overall provides a reasonable approximation of empirical data. The computer model permits the assessment of the reaction over a broad range of conditions and provides a useful tool for the development and management of reaction studies. link: http://identifiers.org/pubmed/8083242

Parameters:

NameDescription
k3 = 1.0E7Reaction: VIII + Xa => Xa + VIIIa, Rate Law: compartment_1*k3*VIII*Xa
k15 = 100000.0Reaction: IX + Xa => Xa + IXa, Rate Law: compartment_1*k15*IX*Xa
k11 = 0.3Reaction: IX_TF_VIIa => TF_VIIa + IXa, Rate Law: compartment_1*k11*IX_TF_VIIa
k2 = 2.0E7Reaction: V + IIa => IIa + Va, Rate Law: compartment_1*k2*V*IIa
k5 = 1.0E7Reaction: mIIa + Va_Xa => Va_Xa + IIa, Rate Law: compartment_1*k5*mIIa*Va_Xa
k19 = 70.0; k6 = 1.0E8Reaction: II + Va_Xa => II_Va_Xa, Rate Law: compartment_1*(k6*II*Va_Xa-k19*II_Va_Xa)
k18 = 0.001; k6 = 1.0E8Reaction: X + VIIIa_IXa => X_VIIIa_IXa, Rate Law: compartment_1*(k6*X*VIIIa_IXa-k18*X_VIIIa_IXa)
k17 = 44.0; k6 = 1.0E8Reaction: X + TF_VIIa => X_TF_VIIa, Rate Law: compartment_1*(k6*X*TF_VIIa-k17*X_TF_VIIa)
k1 = 2.0E7Reaction: V + Xa => Xa + Va, Rate Law: compartment_1*k1*V*Xa
k6 = 1.0E8; k10 = 0.4Reaction: Va + Xa => Va_Xa, Rate Law: compartment_1*(k6*Va*Xa-k10*Va_Xa)
k14 = 32.0Reaction: II_Va_Xa => Va_Xa + mIIa, Rate Law: compartment_1*k14*II_Va_Xa
k12 = 1.15Reaction: X_TF_VIIa => TF_VIIa + Xa, Rate Law: compartment_1*k12*X_TF_VIIa
k16 = 24.0; k6 = 1.0E8Reaction: IX + TF_VIIa => IX_TF_VIIa, Rate Law: compartment_1*(k6*IX*TF_VIIa-k16*IX_TF_VIIa)
k13 = 8.2Reaction: X_VIIIa_IXa => VIIIa_IXa + Xa, Rate Law: compartment_1*k13*X_VIIIa_IXa
k7 = 1.0E7; k9 = 0.005Reaction: VIIIa + IXa => VIIIa_IXa, Rate Law: compartment_1*(k7*VIIIa*IXa-k9*VIIIa_IXa)
I = 0.0Reaction: VIIIa_IXa => ; VIIIa_IXa, Rate Law: compartment_1*(compartment_1*abs(I-VIIIa_IXa)+(I-VIIIa_IXa))/compartment_1
k4 = 2.0E7Reaction: VIII + IIa => IIa + VIIIa, Rate Law: compartment_1*k4*VIII*IIa

States:

NameDescription
IX TF VIIa[Tissue factor; Coagulation factor IX]
VIII[Coagulation factor VIII]
X[Coagulation factor X]
II Va Xa[Prothrombin; Coagulation factor V; Coagulation factor X]
V[Coagulation factor V]
Xa[Coagulation factor X]
TF VIIa[Tissue factor]
VIIIa[Coagulation factor VIII]
X TF VIIa[Tissue factor; Coagulation factor X]
Va[Coagulation factor V]
IIa[Prothrombin]
mIIa[Prothrombin]
Va Xa[Coagulation factor V; Coagulation factor X]
IXa[Coagulation factor IX]
VIIIa IXa[Coagulation factor IX; Coagulation factor VIII]
II[Prothrombin]
IX[Coagulation factor IX]
X VIIIa IXa[Coagulation factor X; Coagulation factor IX; Coagulation factor VIII]

Jung2019 - egulating glioblastoma signaling pathways and anti-invasion therapy cell cycle dynamics model: BIOMD0000000829v0.0.1

This model is based on paper, based on its cell cycle dynamics model: Strategies in regulating glioblastoma signaling p…

Details

Glioblastoma multiforme is one of the most invasive type of glial tumors, which rapidly grows and commonly spreads into nearby brain tissue. It is a devastating brain cancer that often results in death within approximately 12 to 15 months after diagnosis. In this work, optimal control theory was applied to regulate intracellular signaling pathways of miR-451-AMPK-mTOR-cell cycle dynamics via glucose and drug intravenous administration infusions. Glucose level is controlled to activate miR-451 in the up-stream pathway of the model. A potential drug blocking the inhibitory pathway of mTOR by AMPK complex is incorporated to explore regulation of the down-stream pathway to the cell cycle. Both miR-451 and mTOR levels are up-regulated inducing cell proliferation and reducing invasion in the neighboring tissues. Concomitant and alternating glucose and drug infusions are explored under various circumstances to predict best clinical outcomes with least administration costs. link: http://identifiers.org/pubmed/31009513

Parameters:

NameDescription
k_9 = 0.3Reaction: => Plk1; mass_s, CycB, Rate Law: compartment*k_9*mass_s*CycB*(1-Plk1)
S_2 = 1.2; epsilon_2 = 0.02Reaction: => mTOR_R, Rate Law: compartment*S_2/epsilon_2
S_1 = 0.2; epsilon_1 = 0.02Reaction: => AMPK_A, Rate Law: compartment*S_1/epsilon_1
k_7 = 3.0; J_7 = 0.001Reaction: => p55cdc_A; Plk1, p55cdc_T, Rate Law: compartment*k_7*Plk1*(p55cdc_T-p55cdc_A)/((J_7+p55cdc_T)-p55cdc_A)
k_2 = 0.12Reaction: CycB =>, Rate Law: compartment*k_2*CycB
u_1 = 0.0Reaction: => Glucose_G, Rate Law: compartment*u_1
n_1 = 10.0; K_m = 0.5; zeta_1 = 2.5Reaction: mass_s = mass+zeta_1*(1/mTOR_R)^n_1/(K_m^n_1+(1/mTOR_R)^n_1), Rate Law: missing
epsilon_2 = 0.02Reaction: mTOR_R => ; mTOR_R, Rate Law: compartment*mTOR_R/epsilon_2
k_8 = 1.5; J_8 = 0.001; Mad = 1.0Reaction: p55cdc_A =>, Rate Law: compartment*k_8*Mad*p55cdc_A/(J_8+p55cdc_A)
k_10 = 0.06Reaction: Plk1 =>, Rate Law: compartment*k_10*Plk1
k_4 = 105.0; J_4 = 0.04Reaction: Cdh1 => ; mass_s, CycB, Rate Law: compartment*k_4*mass_s*CycB*Cdh1/(J_4+Cdh1)
u_2 = 0.0Reaction: => Drug_D, Rate Law: compartment*u_2
k_6 = 0.3Reaction: p55cdc_T =>, Rate Law: compartment*k_6*p55cdc_T
myu_G = 0.5Reaction: Glucose_G =>, Rate Law: compartment*myu_G*Glucose_G
myu_0 = 0.033; m = 10.0Reaction: => mass, Rate Law: compartment*myu_0*mass*(1-mass/m)
k_5 = 0.015Reaction: => p55cdc_T, Rate Law: compartment*k_5
myu_D = 1.316Reaction: Drug_D =>, Rate Law: compartment*myu_D*Drug_D
l_6 = 1.0; epsilon_2 = 0.02; gamma = 1.0; l_5 = 4.0Reaction: => mTOR_R; deltaD, AMPK_A, Rate Law: compartment*l_5*l_6^2/(epsilon_2*(l_6^2+deltaD*gamma*AMPK_A^2))
HIF = 0.9999999999; p27_p21 = 1.05Reaction: CycB =>, Rate Law: compartment*p27_p21*HIF*CycB
n = 4.0; J_5 = 0.3; k_5_ = 0.6Reaction: => p55cdc_T; CycB, mass_s, Rate Law: compartment*k_5_*(CycB*mass_s)^n/(J_5^n+(CycB*mass_s)^n)
k_1 = 0.12Reaction: => CycB, Rate Law: compartment*k_1
J_3 = 0.04; k_3 = 3.0Reaction: => Cdh1, Rate Law: compartment*k_3*(1-Cdh1)/((J_3+1)-Cdh1)
l_3 = 4.0; beta = 1.0; l_4 = 1.0; epsilon_1 = 0.02Reaction: => AMPK_A; miR_451_M, Rate Law: compartment*l_3*l_4^2/(epsilon_1*(l_4^2+beta*miR_451_M^2))
k_3_ = 30.0; J_3 = 0.04Reaction: => Cdh1; p55cdc_A, Rate Law: compartment*k_3_*p55cdc_A*(1-Cdh1)/((J_3+1)-Cdh1)
l_2 = 1.0; l_1 = 4.0; alpha = 1.6Reaction: => miR_451_M; Glucose_G, AMPK_A, Rate Law: compartment*(Glucose_G+l_1*l_2^2/(l_2^2+alpha*AMPK_A^2))
epsilon_1 = 0.02Reaction: AMPK_A =>, Rate Law: compartment*AMPK_A/epsilon_1
k_2_ = 4.5Reaction: CycB => ; Cdh1, Rate Law: compartment*k_2_*Cdh1*CycB

States:

NameDescription
AMPK A[5'-AMP-activated protein kinase catalytic subunit alpha-2; 5'-AMP-Activated Protein Kinase]
Glucose G[glucose]
p55cdc A[Cell division cycle protein 20 homolog]
deltaDdeltaD
Drug D[drug]
Plk1[Serine/threonine-protein kinase PLK1]
mTOR R[CCO:2475; Serine/threonine-protein kinase mTOR; MTOR Gene]
CycB[G2/mitotic-specific cyclin-B3]
mass[Mass]
Cdh1[Fizzy-related protein homolog]
miR 451 M[cAMP-regulated phosphoprotein 19; MIR451A Pre-miRNA]
p55cdc T[Cell division cycle protein 20 homolog]
mass s[Mass]

Jung2019 - Regulating glioblastoma signaling pathways and anti-invasion therapy - core control model: BIOMD0000000828v0.0.1

This model is based on paper: Strategies in regulating glioblastoma signaling pathways and anti-invasion therapy Abs…

Details

Glioblastoma multiforme is one of the most invasive type of glial tumors, which rapidly grows and commonly spreads into nearby brain tissue. It is a devastating brain cancer that often results in death within approximately 12 to 15 months after diagnosis. In this work, optimal control theory was applied to regulate intracellular signaling pathways of miR-451-AMPK-mTOR-cell cycle dynamics via glucose and drug intravenous administration infusions. Glucose level is controlled to activate miR-451 in the up-stream pathway of the model. A potential drug blocking the inhibitory pathway of mTOR by AMPK complex is incorporated to explore regulation of the down-stream pathway to the cell cycle. Both miR-451 and mTOR levels are up-regulated inducing cell proliferation and reducing invasion in the neighboring tissues. Concomitant and alternating glucose and drug infusions are explored under various circumstances to predict best clinical outcomes with least administration costs. link: http://identifiers.org/pubmed/31009513

Parameters:

NameDescription
myu_D = 1.316Reaction: Drug_D =>, Rate Law: compartment*myu_D*Drug_D
l_6 = 1.0; epsilon_2 = 0.02; gamma = 1.0; l_5 = 4.0Reaction: => mTOR_R; deltaD, AMPK_A, Rate Law: compartment*l_5*l_6^2/(epsilon_2*(l_6^2+deltaD*gamma*AMPK_A^2))
S_2 = 1.2; epsilon_2 = 0.02Reaction: => mTOR_R, Rate Law: compartment*S_2/epsilon_2
S_1 = 0.2; epsilon_1 = 0.02Reaction: => AMPK_A, Rate Law: compartment*S_1/epsilon_1
l_3 = 4.0; beta = 1.0; l_4 = 1.0; epsilon_1 = 0.02Reaction: => AMPK_A; miR_451_M, Rate Law: compartment*l_3*l_4^2/(epsilon_1*(l_4^2+beta*miR_451_M^2))
epsilon_2 = 0.02Reaction: mTOR_R => ; mTOR_R, Rate Law: compartment*mTOR_R/epsilon_2
u_1 = 0.0Reaction: => Glucose_G, Rate Law: compartment*u_1
l_2 = 1.0; l_1 = 4.0; alpha = 1.6Reaction: => miR_451_M; Glucose_G, AMPK_A, Rate Law: compartment*(Glucose_G+l_1*l_2^2/(l_2^2+alpha*AMPK_A^2))
u_2 = 0.0Reaction: => Drug_D, Rate Law: compartment*u_2
epsilon_1 = 0.02Reaction: AMPK_A =>, Rate Law: compartment*AMPK_A/epsilon_1
myu_G = 0.5Reaction: Glucose_G =>, Rate Law: compartment*myu_G*Glucose_G

States:

NameDescription
Drug D[drug]
AMPK A[5'-AMP-activated protein kinase catalytic subunit alpha-2; 5'-AMP-Activated Protein Kinase]
mTOR R[CCO:2475; MTOR Gene; Serine/threonine-protein kinase mTOR]
Glucose G[glucose]
miR 451 M[MIR451A Pre-miRNA; cAMP-regulated phosphoprotein 19]
deltaDdeltaD

Jönsson2005_WUSCHELexpression_ShootApicalMeristem: MODEL1112100000v0.0.1

This is a Systems Biology Markup Language (SBML) file, generated by MathSBML 2.5.13 (4 May 2006) 13-May-2006 20:05:11.20…

Details

The above-ground tissues of higher plants are generated from a small region of cells situated at the plant apex called the shoot apical meristem. An important genetic control circuit modulating the size of the Arabidopsis thaliana meristem is a feed-back network between the CLAVATA3 and WUSCHEL genes. Although the expression patterns for these genes do not overlap, WUSCHEL activity is both necessary and sufficient (when expressed ectopically) for the induction of CLAVATA3 expression. However, upregulation of CLAVATA3 in conjunction with the receptor kinase CLAVATA1 results in the downregulation of WUSCHEL. Despite much work, experimental data for this network are incomplete and additional hypotheses are needed to explain the spatial locations and dynamics of these expression domains. Predictive mathematical models describing the system should provide a useful tool for investigating and discriminating among possible hypotheses, by determining which hypotheses best explain observed gene expression dynamics.We are developing a method using in vivo live confocal microscopy to capture quantitative gene expression data and create templates for computational models. We present two models accounting for the organization of the WUSCHEL expression domain. Our preferred model uses a reaction-diffusion mechanism in which an activator induces WUSCHEL expression. This model is able to organize the WUSCHEL expression domain. In addition, the model predicts the dynamical reorganization seen in experiments where cells, including the WUSCHEL domain, are ablated, and it also predicts the spatial expansion of the WUSCHEL domain resulting from removal of the CLAVATA3 signal.An extended description of the model framework and image processing algorithms can be found at http://www.computableplant.org, together with additional results and simulation movies.http://www.computableplant.org/ and alternatively for a direct link to the page, http://computableplant.ics.uci.edu/bti1036 can be accessed. link: http://identifiers.org/pubmed/15961462

K


Kaiser2014 - Salmonella persistence after ciprofloxacin treatment: BIOMD0000000527v0.0.1

Kaiser2014 - Salmonella persistence after ciprofloxacin treatment The model describes the bacterial tolerance to antibi…

Details

In vivo, antibiotics are often much less efficient than ex vivo and relapses can occur. The reasons for poor in vivo activity are still not completely understood. We have studied the fluoroquinolone antibiotic ciprofloxacin in an animal model for complicated Salmonellosis. High-dose ciprofloxacin treatment efficiently reduced pathogen loads in feces and most organs. However, the cecum draining lymph node (cLN), the gut tissue, and the spleen retained surviving bacteria. In cLN, approximately 10%-20% of the bacteria remained viable. These phenotypically tolerant bacteria lodged mostly within CD103⁺CX₃CR1⁻CD11c⁺ dendritic cells, remained genetically susceptible to ciprofloxacin, were sufficient to reinitiate infection after the end of the therapy, and displayed an extremely slow growth rate, as shown by mathematical analysis of infections with mixed inocula and segregative plasmid experiments. The slow growth was sufficient to explain recalcitrance to antibiotics treatment. Therefore, slow-growing antibiotic-tolerant bacteria lodged within dendritic cells can explain poor in vivo antibiotic activity and relapse. Administration of LPS or CpG, known elicitors of innate immune defense, reduced the loads of tolerant bacteria. Thus, manipulating innate immunity may augment the in vivo activity of antibiotics. link: http://identifiers.org/pubmed/24558351

Parameters:

NameDescription
mu1 = 297.78957 per_day; r3 = 4.5867007 per_day; t1 = 1.0 day; c10 = 2.43E-7 per_day; r1 = 2.8195198 per_day; t5 = 5.0 day; t3 = 3.0 day; r10 = 0.3757764 per_day; mu5 = 0.0 per_day; r5 = 1.8812956 per_day; c5 = 2.497735 per_day; t10 = 10.0 day; c1 = 0.0 per_day; c3 = 5.042901 per_day; mu10 = 0.0 per_day; mu3 = 0.0 per_dayReaction: L = piecewise(mu1+(r1-c1)*L, (time >= 0) && (time <= t1), mu3+(r3-c3)*L, (time > t1) && (time <= t3), mu5+(r5-c5)*L, (time > t3) && (time <= t5), mu10+(r10-c10)*L, (time > t5) && (time <= t10)), Rate Law: piecewise(mu1+(r1-c1)*L, (time >= 0) && (time <= t1), mu3+(r3-c3)*L, (time > t1) && (time <= t3), mu5+(r5-c5)*L, (time > t3) && (time <= t5), mu10+(r10-c10)*L, (time > t5) && (time <= t10))

States:

NameDescription
L[Salmonella enterica subsp. enterica serovar Typhimurium str. DT104]

Kaizu2010_BuddingYeastCellCycle_Map: MODEL1011020000v0.0.1

This is the map described in and accompanying the article: **A comprehensive molecular interaction map of the budding…

Details

With the accumulation of data on complex molecular machineries coordinating cell-cycle dynamics, coupled with its central function in disease patho-physiologies, it is becoming increasingly important to collate the disparate knowledge sources into a comprehensive molecular network amenable to systems-level analyses. In this work, we present a comprehensive map of the budding yeast cell-cycle, curating reactions from ∼600 original papers. Toward leveraging the map as a framework to explore the underlying network architecture, we abstract the molecular components into three planes–signaling, cell-cycle core and structural planes. The planar view together with topological analyses facilitates network-centric identification of functions and control mechanisms. Further, we perform a comparative motif analysis to identify around 194 motifs including feed-forward, mutual inhibitory and feedback mechanisms contributing to cell-cycle robustness. We envisage the open access, comprehensive cell-cycle map to open roads toward community-based deeper understanding of cell-cycle dynamics. link: http://identifiers.org/pubmed/20865008

Kallenberger2014 - CD95L induced apoptosis initiated by caspase-8, CD95 HeLa cells (cis/trans variant): BIOMD0000000523v0.0.1

Kallenberger2014 - CD95L induced apoptosis initiated by caspase-8, CD95 HeLa cells (cis/trans variant)The paper describe…

Details

Apoptosis in response to the ligand CD95L (also known as Fas ligand) is initiated by caspase-8, which is activated by dimerization and self-cleavage at death-inducing signaling complexes (DISCs). Previous work indicated that the degree of substrate cleavage by caspase-8 determines whether a cell dies or survives in response to a death stimulus. To determine how a death ligand stimulus is effectively translated into caspase-8 activity, we assessed this activity over time in single cells with compartmentalized probes that are cleaved by caspase-8 and used multiscale modeling to simultaneously describe single-cell and population data with an ensemble of single-cell models. We derived and experimentally validated a minimal model in which cleavage of caspase-8 in the enzymatic domain occurs in an interdimeric manner through interaction between DISCs, whereas prodomain cleavage sites are cleaved in an intradimeric manner within DISCs. Modeling indicated that sustained membrane-bound caspase-8 activity is followed by transient cytosolic activity, which can be interpreted as a molecular timer mechanism reflected by a limited lifetime of active caspase-8. The activation of caspase-8 by combined intra- and interdimeric cleavage ensures weak signaling at low concentrations of CD95L and strongly accelerated activation at higher ligand concentrations, thereby contributing to precise control of apoptosis. link: http://identifiers.org/pubmed/24619646

Parameters:

NameDescription
kD374probe = 0.00152252549827479Reaction: PrNES_mCherry => PrNES + mCherry; p43, p18, PrNES_mCherry, p43, p18, Rate Law: kD374probe*PrNES_mCherry*(p43+p18)*cell
kdiss_p18 = 0.0949914492651531Reaction: p18 => p18inactive; p18, Rate Law: kdiss_p18*p18*cell
kD216 = 0.0114186392006403Reaction: p43 => p18 + DISC; p43, Rate Law: kD216*p43*cell
kon_FADD = 8.11711012144556E-4; CD95act = 0.0Reaction: FADD => DISC; FADD, Rate Law: kon_FADD*CD95act*FADD*cell
kDISC = 4.91828591049766E-4Reaction: p55free + DISC => DISCp55; p55free, DISC, Rate Law: kDISC*p55free*DISC*cell
kD374trans_p55 = 4.46994772958953E-4Reaction: p30 => p18 + DISC; DISCp55, p30, p30, DISCp55, Rate Law: kD374trans_p55*p30*(DISCp55+p30)*cell
kD374trans_p43 = 0.00343995957326369Reaction: p30 => p18 + DISC; p43, p30, p43, Rate Law: kD374trans_p43*p30*p43*cell
koff_FADD = 0.00566528253772301Reaction: DISC => FADD; DISC, Rate Law: koff_FADD*DISC*cell
kBid = 5.2867403363568E-4Reaction: Bid => tBid; p43, p18, Bid, p43, p18, Rate Law: kBid*Bid*(p43+p18)*cell

States:

NameDescription
Bid[BH3-interacting domain death agonist]
PrNES mCherry[SBO:0000178; probe; Red fluorescent protein drFP583; nuclear_export_signal]
p30[CASP8]
FADD[FAS-associated death domain protein]
p18[Caspase-8]
p43[CASP8 and FADD-like apoptosis regulator]
DISC[death-inducing signaling complex]
p55free[CASP8]
PrER[probe; Calnexin]
mCherry[Red fluorescent protein drFP583]
DISCp55[CASP8; death-inducing signaling complex]
PrER mGFP[SBO:0000178; probe; Calnexin; Green fluorescent protein]
mGFP[Green fluorescent protein]
PrNES[probe; nuclear_export_signal]
p18inactive[inactive; Caspase-8]
tBid[BH3-interacting domain death agonist; mitochondrial outer membrane permeabilization]

Kallenberger2014 - CD95L induced apoptosis initiated by caspase-8, CD95 HeLa cells (cis/trans-cis/trans variant): BIOMD0000000525v0.0.1

Kallenberger2014 - CD95L induced apoptosis initiated by caspase-8, CD95 HeLa cells (cis/trans-cis/trans variant)The pape…

Details

Apoptosis in response to the ligand CD95L (also known as Fas ligand) is initiated by caspase-8, which is activated by dimerization and self-cleavage at death-inducing signaling complexes (DISCs). Previous work indicated that the degree of substrate cleavage by caspase-8 determines whether a cell dies or survives in response to a death stimulus. To determine how a death ligand stimulus is effectively translated into caspase-8 activity, we assessed this activity over time in single cells with compartmentalized probes that are cleaved by caspase-8 and used multiscale modeling to simultaneously describe single-cell and population data with an ensemble of single-cell models. We derived and experimentally validated a minimal model in which cleavage of caspase-8 in the enzymatic domain occurs in an interdimeric manner through interaction between DISCs, whereas prodomain cleavage sites are cleaved in an intradimeric manner within DISCs. Modeling indicated that sustained membrane-bound caspase-8 activity is followed by transient cytosolic activity, which can be interpreted as a molecular timer mechanism reflected by a limited lifetime of active caspase-8. The activation of caspase-8 by combined intra- and interdimeric cleavage ensures weak signaling at low concentrations of CD95L and strongly accelerated activation at higher ligand concentrations, thereby contributing to precise control of apoptosis. link: http://identifiers.org/pubmed/24619646

Parameters:

NameDescription
kD374trans_p55 = 5.43518631342483E-4Reaction: DISCp55 => p43; DISCp55, p30, DISCp55, p30, Rate Law: kD374trans_p55*DISCp55*(DISCp55+p30)*cell
kD374 = 6.44612643975149E-4Reaction: DISCp55 => p43; DISCp55, Rate Law: kD374*DISCp55*cell
kDISC = 3.64965874405544E-4Reaction: p55free + DISC => DISCp55; p55free, DISC, Rate Law: kDISC*p55free*DISC*cell
kdiss_p18 = 0.064713651554491Reaction: p18 => p18inactive; p18, Rate Law: kdiss_p18*p18*cell
kD374trans_p43 = 0.00413530054938906Reaction: p30 => p18 + DISC; p43, p30, p43, Rate Law: kD374trans_p43*p30*p43*cell
kD374probe = 0.00153710001025539Reaction: PrNES_mCherry => PrNES + mCherry; p43, p18, PrNES_mCherry, p43, p18, Rate Law: kD374probe*PrNES_mCherry*(p43+p18)*cell
kD216trans_p43 = 5.29906975294056E-5Reaction: p43 => p18 + DISC; p43, p43, Rate Law: kD216trans_p43*p43*p43*cell
kBid = 5.2134055139547E-4Reaction: Bid => tBid; p43, p18, Bid, p43, p18, Rate Law: kBid*Bid*(p43+p18)*cell
kD216trans_p55 = 2.23246421372882E-4Reaction: p43 => p18 + DISC; DISCp55, p30, p43, DISCp55, p30, Rate Law: kD216trans_p55*p43*(DISCp55+p30)*cell
koff_FADD = 0.00130854998177646Reaction: DISC => FADD; DISC, Rate Law: koff_FADD*DISC*cell
kD216 = 0.00639775937416746Reaction: DISCp55 => p30; DISCp55, Rate Law: kD216*DISCp55*cell
CD95act = 0.0; kon_FADD = 0.00108871858684363Reaction: FADD => DISC; FADD, Rate Law: kon_FADD*CD95act*FADD*cell

States:

NameDescription
Bid[BH3-interacting domain death agonist]
PrNES mCherry[SBO:0000178; probe; Red fluorescent protein drFP583; nuclear_export_signal]
p30[CASP8]
FADD[FAS-associated death domain protein]
p18[Caspase-8]
p43[CASP8 and FADD-like apoptosis regulator]
DISC[death-inducing signaling complex]
p55free[CASP8]
PrER[probe; Calnexin]
mCherry[Red fluorescent protein drFP583]
PrER mGFP[SBO:0000178; probe; Calnexin; Green fluorescent protein]
DISCp55[CASP8; death-inducing signaling complex]
mGFP[Green fluorescent protein]
PrNES[probe; nuclear_export_signal]
p18inactive[inactive; Caspase-8]
tBid[BH3-interacting domain death agonist; mitochondrial outer membrane permeabilization]

Kallenberger2014 - CD95L induced apoptosis initiated by caspase-8, wild-type HeLa cells (cis/trans variant): BIOMD0000000524v0.0.1

Kallenberger2014 - CD95L induced apoptosis initiated by caspase-8, wild-type HeLa cells (cis/trans variant)The paper des…

Details

Apoptosis in response to the ligand CD95L (also known as Fas ligand) is initiated by caspase-8, which is activated by dimerization and self-cleavage at death-inducing signaling complexes (DISCs). Previous work indicated that the degree of substrate cleavage by caspase-8 determines whether a cell dies or survives in response to a death stimulus. To determine how a death ligand stimulus is effectively translated into caspase-8 activity, we assessed this activity over time in single cells with compartmentalized probes that are cleaved by caspase-8 and used multiscale modeling to simultaneously describe single-cell and population data with an ensemble of single-cell models. We derived and experimentally validated a minimal model in which cleavage of caspase-8 in the enzymatic domain occurs in an interdimeric manner through interaction between DISCs, whereas prodomain cleavage sites are cleaved in an intradimeric manner within DISCs. Modeling indicated that sustained membrane-bound caspase-8 activity is followed by transient cytosolic activity, which can be interpreted as a molecular timer mechanism reflected by a limited lifetime of active caspase-8. The activation of caspase-8 by combined intra- and interdimeric cleavage ensures weak signaling at low concentrations of CD95L and strongly accelerated activation at higher ligand concentrations, thereby contributing to precise control of apoptosis. link: http://identifiers.org/pubmed/24619646

Parameters:

NameDescription
kD374probe = 0.00152252549827479Reaction: PrNES_mCherry => PrNES + mCherry; p43, p18, PrNES_mCherry, p43, p18, Rate Law: kD374probe*PrNES_mCherry*(p43+p18)*cell
kdiss_p18 = 0.0949914492651531Reaction: p18 => p18inactive; p18, Rate Law: kdiss_p18*p18*cell
kon_FADD = 8.11711012144556E-4; CD95act = 0.0Reaction: FADD => DISC; FADD, Rate Law: kon_FADD*CD95act*FADD*cell
kD216 = 0.0114186392006403Reaction: p43 => p18 + DISC; p43, Rate Law: kD216*p43*cell
kDISC = 4.91828591049766E-4Reaction: p55free + DISC => DISCp55; p55free, DISC, Rate Law: kDISC*p55free*DISC*cell
kBid = 5.2867403363568E-4Reaction: Bid => tBid; p43, p18, Bid, p43, p18, Rate Law: kBid*Bid*(p43+p18)*cell
kD374trans_p55 = 4.46994772958953E-4Reaction: p30 => p18 + DISC; DISCp55, p30, p30, DISCp55, Rate Law: kD374trans_p55*p30*(DISCp55+p30)*cell
koff_FADD = 0.00566528253772301Reaction: DISC => FADD; DISC, Rate Law: koff_FADD*DISC*cell
kD374trans_p43 = 0.00343995957326369Reaction: p30 => p18 + DISC; p43, p30, p43, Rate Law: kD374trans_p43*p30*p43*cell

States:

NameDescription
Bid[BH3-interacting domain death agonist]
PrNES mCherry[SBO:0000178; probe; Red fluorescent protein drFP583; nuclear_export_signal]
p30[CASP8]
FADD[FAS-associated death domain protein]
p18[Caspase-8]
p43[CASP8 and FADD-like apoptosis regulator]
DISC[death-inducing signaling complex]
p55free[CASP8]
PrER[probe; Calnexin]
mCherry[Red fluorescent protein drFP583]
PrER mGFP[SBO:0000178; probe; Calnexin; Green fluorescent protein]
DISCp55[CASP8; death-inducing signaling complex]
mGFP[Green fluorescent protein]
PrNES[probe; nuclear_export_signal]
p18inactive[inactive; Caspase-8]
tBid[BH3-interacting domain death agonist; mitochondrial outer membrane permeabilization]

Kallenberger2014 - CD95L induced apoptosis initiated by caspase-8, wild-type HeLa cells (cis/trans-cis/trans variant): BIOMD0000000526v0.0.1

Kallenberger2014 - CD95L induced apoptosis initiated by caspase-8, wild-type HeLa cells (cis/trans-cis/trans variant)The…

Details

Apoptosis in response to the ligand CD95L (also known as Fas ligand) is initiated by caspase-8, which is activated by dimerization and self-cleavage at death-inducing signaling complexes (DISCs). Previous work indicated that the degree of substrate cleavage by caspase-8 determines whether a cell dies or survives in response to a death stimulus. To determine how a death ligand stimulus is effectively translated into caspase-8 activity, we assessed this activity over time in single cells with compartmentalized probes that are cleaved by caspase-8 and used multiscale modeling to simultaneously describe single-cell and population data with an ensemble of single-cell models. We derived and experimentally validated a minimal model in which cleavage of caspase-8 in the enzymatic domain occurs in an interdimeric manner through interaction between DISCs, whereas prodomain cleavage sites are cleaved in an intradimeric manner within DISCs. Modeling indicated that sustained membrane-bound caspase-8 activity is followed by transient cytosolic activity, which can be interpreted as a molecular timer mechanism reflected by a limited lifetime of active caspase-8. The activation of caspase-8 by combined intra- and interdimeric cleavage ensures weak signaling at low concentrations of CD95L and strongly accelerated activation at higher ligand concentrations, thereby contributing to precise control of apoptosis. link: http://identifiers.org/pubmed/24619646

Parameters:

NameDescription
kD374trans_p55 = 5.43518631342483E-4Reaction: DISCp55 => p43; DISCp55, p30, DISCp55, p30, Rate Law: kD374trans_p55*DISCp55*(DISCp55+p30)*cell
kD374 = 6.44612643975149E-4Reaction: DISCp55 => p43; DISCp55, Rate Law: kD374*DISCp55*cell
kdiss_p18 = 0.064713651554491Reaction: p18 => p18inactive; p18, Rate Law: kdiss_p18*p18*cell
kDISC = 3.64965874405544E-4Reaction: p55free + DISC => DISCp55; p55free, DISC, Rate Law: kDISC*p55free*DISC*cell
kD374trans_p43 = 0.00413530054938906Reaction: DISCp55 => p43; p43, DISCp55, p43, Rate Law: kD374trans_p43*DISCp55*p43*cell
kD374probe = 0.00153710001025539Reaction: PrNES_mCherry => PrNES + mCherry; p43, p18, PrNES_mCherry, p43, p18, Rate Law: kD374probe*PrNES_mCherry*(p43+p18)*cell
kD216trans_p43 = 5.29906975294056E-5Reaction: DISCp55 => p30; p43, DISCp55, p43, Rate Law: kD216trans_p43*DISCp55*p43*cell
kBid = 5.2134055139547E-4Reaction: Bid => tBid; p43, p18, Bid, p43, p18, Rate Law: kBid*Bid*(p43+p18)*cell
kD216trans_p55 = 2.23246421372882E-4Reaction: DISCp55 => p30; DISCp55, p30, DISCp55, p30, Rate Law: kD216trans_p55*DISCp55*(DISCp55+p30)*cell
koff_FADD = 0.00130854998177646Reaction: DISC => FADD; DISC, Rate Law: koff_FADD*DISC*cell
kD216 = 0.00639775937416746Reaction: DISCp55 => p30; DISCp55, Rate Law: kD216*DISCp55*cell
CD95act = 0.0; kon_FADD = 0.00108871858684363Reaction: FADD => DISC; FADD, Rate Law: kon_FADD*CD95act*FADD*cell

States:

NameDescription
Bid[BH3-interacting domain death agonist]
PrNES mCherry[SBO:0000178; probe; Red fluorescent protein drFP583; nuclear_export_signal]
p30[CASP8]
FADD[FAS-associated death domain protein]
p18[Caspase-8]
p43[CASP8 and FADD-like apoptosis regulator]
DISC[death-inducing signaling complex]
p55free[CASP8]
PrER[probe; Calnexin]
mCherry[Red fluorescent protein drFP583]
DISCp55[CASP8; death-inducing signaling complex]
mGFP[Green fluorescent protein]
PrER mGFP[SBO:0000178; probe; Calnexin; Green fluorescent protein]
PrNES[probe; nuclear_export_signal]
p18inactive[inactive; Caspase-8]
tBid[BH3-interacting domain death agonist; mitochondrial outer membrane permeabilization]

Kamihira2000 - calcitonin fibrillation kinetics: BIOMD0000000614v0.0.1

Kamihira2000 - calcitonin fibrillation kineticsThis model studies the kinetics of human calcitonin fibrillation describe…

Details

Conformational transitions of human calcitonin (hCT) during fibril formation in the acidic and neutral conditions were investigated by high-resolution solid-state 13C NMR spectroscopy. In aqueous acetic acid solution (pH 3.3), a local alpha-helical form is present around Gly10 whereas a random coil form is dominant as viewed from Phe22, Ala26, and Ala31 in the monomer form on the basis of the 13C chemical shifts. On the other hand, a local beta-sheet form as viewed from Gly10 and Phe22, and both beta-sheet and random coil as viewed from Ala26 and Ala31 were detected in the fibril at pH 3.3. The results indicate that conformational transitions from alpha-helix to beta-sheet, and from random coil to beta-sheet forms occurred in the central and C-terminus regions, respectively, during the fibril formation. The increased 13C resonance intensities of fibrils after a certain delay time suggests that the fibrillation can be explained by a two-step reaction mechanism in which the first step is a homogeneous association to form a nucleus, and the second step is an autocatalytic heterogeneous fibrillation. In contrast to the fibril at pH 3.3, the fibril at pH 7.5 formed a local beta-sheet conformation at the central region and exhibited a random coil at the C-terminus region. Not only a hydrophobic interaction among the amphiphilic alpha-helices, but also an electrostatic interaction between charged side chains can play an important role for the fibril formation at pH 7.5 and 3.3 acting as electrostatically favorable and unfavorable interactions, respectively. These results suggest that hCT fibrils are formed by stacking antiparallel beta-sheets at pH 7.5 and a mixture of antiparallel and parallel beta-sheets at pH 3.3. link: http://identifiers.org/pubmed/10850796

Parameters:

NameDescription
k2 = 2.29; a = 5.85E-5Reaction: => f, Rate Law: compartment_*k2*a*f
k1 = 2.79E-6Reaction: => f, Rate Law: compartment_*k1

States:

NameDescription
f[Calcitonin]

Kamminga2017 - Metabolic model of Mycoplasma hyopneumoniae growth: MODEL1704250001v0.0.1

Kamminga2017 - Metabolic model of Mycoplasma hyopneumoniae growthThis model is described in the article: [Metabolic mod…

Details

Mycoplasma hyopneumoniae is cultured on large-scale to produce antigen for inactivated whole-cell vaccines against respiratory disease in pigs. However, the fastidious nutrient requirements of this minimal bacterium and the low growth rate make it challenging to reach sufficient biomass yield for antigen production. In this study, we sequenced the genome of M. hyopneumoniae strain 11 and constructed a high quality constraint-based genome-scale metabolic model of 284 chemical reactions and 298 metabolites. We validated the model with time-series data of duplicate fermentation cultures to aim for an integrated model describing the dynamic profiles measured in fermentations. The model predicted that 84% of cellular energy in a standard M. hyopneumoniae cultivation was used for non-growth associated maintenance and only 16% of cellular energy was used for growth and growth associated maintenance. Following a cycle of model-driven experimentation in dedicated fermentation experiments, we were able to increase the fraction of cellular energy used for growth through pyruvate addition to the medium. This increase in turn led to an increase in growth rate and a 2.3 times increase in the total biomass concentration reached after 3-4 days of fermentation, enhancing the productivity of the overall process. The model presented provides a solid basis to understand and further improve M. hyopneumoniae fermentation processes. Biotechnol. Bioeng. 2017;9999: 1-9. © 2017 Wiley Periodicals, Inc. link: http://identifiers.org/pubmed/28600895

Karagiannis2004_CollagenIproteolysis: MODEL0911270007v0.0.1

This a model from the article: A theoretical model of type I collagen proteolysis by matrix metalloproteinase (MMP) 2…

Details

One well documented family of enzymes responsible for the proteolytic processes that occur in the extracellular matrix is the soluble and membrane-associated matrix metalloproteinases. Here we present the first theoretical model of the biochemical network describing the proteolysis of collagen I by matrix metalloproteinases 2 (MMP2) and membrane type 1 matrix metalloproteinases (MT1-MMP) in the presence of the tissue inhibitor of metalloproteinases 2 (TIMP2) in a bulk, cell-free, well stirred environment. The model can serve as a tool for describing quantitatively the activation of the MMP2 proenzyme (pro-MMP2), the ectodomain shedding of MT1-MMP, and the collagenolysis arising from both of the enzymes. We show that pro-MMP2 activation, a process that involves a trimer formation of the proenzyme with TIMP2 and MT1-MMP, is suppressed at high inhibitor levels and paradoxically attains maximum only at intermediate TIMP2 concentrations. We also calculate the conditions for which pro-MMP2 activation is maximal. Furthermore we demonstrate that the ectodomain shedding of MT1-MMP can serve as a mechanism controlling the MT1-MMP availability and therefore the pro-MMP2 activation. Finally the proteolytic synergism of MMP2 and MT1-MMP is introduced and described quantitatively. The model provides us a tool to determine the conditions under which the synergism is optimized. Our approach is the first step toward a more complete description of the proteolytic processes that occur in the extracellular matrix and include a wider spectrum of enzymes and substrates as well as naturally occurring or artificial inhibitors. link: http://identifiers.org/pubmed/15252025

Karapetyan2016 - Genetic oscillatory network - Activator Titration Circuit (ATC): BIOMD0000000586v0.0.1

Karapetyan2016 - Genetic oscillatory network - Activator Titration Circuit (ATC)This model is described in the article:…

Details

Genetic oscillators, such as circadian clocks, are constantly perturbed by molecular noise arising from the small number of molecules involved in gene regulation. One of the strongest sources of stochasticity is the binary noise that arises from the binding of a regulatory protein to a promoter in the chromosomal DNA. In this study, we focus on two minimal oscillators based on activator titration and repressor titration to understand the key parameters that are important for oscillations and for overcoming binary noise. We show that the rate of unbinding from the DNA, despite traditionally being considered a fast parameter, needs to be slow to broaden the space of oscillatory solutions. The addition of multiple, independent DNA binding sites further expands the oscillatory parameter space for the repressor-titration oscillator and lengthens the period of both oscillators. This effect is a combination of increased effective delay of the unbinding kinetics due to multiple binding sites and increased promoter ultrasensitivity that is specific for repression. We then use stochastic simulation to show that multiple binding sites increase the coherence of oscillations by mitigating the binary noise. Slow values of DNA unbinding rate are also effective in alleviating molecular noise due to the increased distance from the bifurcation point. Our work demonstrates how the number of DNA binding sites and slow unbinding kinetics, which are often omitted in biophysical models of gene circuits, can have a significant impact on the temporal and stochastic dynamics of genetic oscillators. link: http://identifiers.org/pubmed/26764732

Parameters:

NameDescription
t_32 = 0.0Reaction: G3 => G2 + A2; G3, Rate Law: yeast*t_32*G3
a_01 = 2.49202551834131E-4Reaction: G0 + A2 => G1; G0, A2, Rate Law: yeast*a_01*G0*A2
delta_m = 0.0186Reaction: rA => ; rA, Rate Law: yeast*delta_m*rA
a_23 = 0.0Reaction: G2 + A2 => G3; G2, A2, Rate Law: yeast*a_23*G2*A2
delta_p = 0.0077Reaction: I => ; I, Rate Law: yeast*delta_p*I
t_21 = 0.0Reaction: G2 => G1 + A2; G2, Rate Law: yeast*t_21*G2
epsilon_1 = 6.0Reaction: A2 => A; A2, Rate Law: yeast*epsilon_1*A2
a_12 = 0.0Reaction: G1 + A2 => G2; G1, A2, Rate Law: yeast*a_12*G1*A2
rho_f = 0.1781Reaction: G0 => G0 + rI; G0, Rate Law: yeast*rho_f*G0
epsilon = 0.024Reaction: AI => A + I; AI, Rate Law: yeast*epsilon*AI
beta = 14.109Reaction: rA => rA + A; rA, Rate Law: yeast*beta*rA
rho_b = 5.343Reaction: G1 => G1 + rI; G1, Rate Law: yeast*rho_b*G1
t_10 = 0.02Reaction: G1 => G0 + A2; G1, Rate Law: yeast*t_10*G1
gamma = 0.025Reaction: A + I => AI; A, I, Rate Law: yeast*gamma*A*I
rho_0 = 0.975493874916701Reaction: => rA, Rate Law: yeast*rho_0

States:

NameDescription
G3G3
II
rIrI
AA
G1G1
A2[IPR004827]
rArA
G2G2
G0G0
AIAI

Karapetyan2016 - Genetic oscillatory network - Repressor Titration Circuit (RTC): BIOMD0000000587v0.0.1

Karapetyan2016 - Genetic oscillatory network - Repressor Titration Circuit (RTC)This model is described in the article:…

Details

Genetic oscillators, such as circadian clocks, are constantly perturbed by molecular noise arising from the small number of molecules involved in gene regulation. One of the strongest sources of stochasticity is the binary noise that arises from the binding of a regulatory protein to a promoter in the chromosomal DNA. In this study, we focus on two minimal oscillators based on activator titration and repressor titration to understand the key parameters that are important for oscillations and for overcoming binary noise. We show that the rate of unbinding from the DNA, despite traditionally being considered a fast parameter, needs to be slow to broaden the space of oscillatory solutions. The addition of multiple, independent DNA binding sites further expands the oscillatory parameter space for the repressor-titration oscillator and lengthens the period of both oscillators. This effect is a combination of increased effective delay of the unbinding kinetics due to multiple binding sites and increased promoter ultrasensitivity that is specific for repression. We then use stochastic simulation to show that multiple binding sites increase the coherence of oscillations by mitigating the binary noise. Slow values of DNA unbinding rate are also effective in alleviating molecular noise due to the increased distance from the bifurcation point. Our work demonstrates how the number of DNA binding sites and slow unbinding kinetics, which are often omitted in biophysical models of gene circuits, can have a significant impact on the temporal and stochastic dynamics of genetic oscillators. link: http://identifiers.org/pubmed/26764732

Parameters:

NameDescription
t_32 = 0.0Reaction: G3 => G2 + R2; G3, Rate Law: yeast*t_32*G3
a_01 = 2.49202551834131E-4Reaction: G0 + R2 => G1; G0, R2, Rate Law: yeast*a_01*G0*R2
a_23 = 0.0Reaction: G2 + R2 => G3; G2, R2, Rate Law: yeast*a_23*G2*R2
rho_0 = 0.468598473029544Reaction: => rI, Rate Law: yeast*rho_0
delta_p = 0.0077Reaction: I => ; I, Rate Law: yeast*delta_p*I
t_21 = 0.0Reaction: G2 => G1 + R2; G2, Rate Law: yeast*t_21*G2
rho_b = 0.245950413223141Reaction: G3 => G3 + rR; G3, Rate Law: yeast*rho_b*G3
rho_f = 0.8928Reaction: G0 => G0 + rR; G0, Rate Law: yeast*rho_f*G0
epsilon_1 = 6.0Reaction: R2 => R; R2, Rate Law: yeast*epsilon_1*R2
a_12 = 0.0Reaction: G1 + R2 => G2; G1, R2, Rate Law: yeast*a_12*G1*R2
epsilon = 0.024Reaction: RI => R + I; RI, Rate Law: yeast*epsilon*RI
beta = 14.109Reaction: rI => rI + I; rI, Rate Law: yeast*beta*rI
delta_m = 0.0159Reaction: rR => ; rR, Rate Law: yeast*delta_m*rR
t_10 = 0.02Reaction: G1 => G0 + R2; G1, Rate Law: yeast*t_10*G1
gamma = 0.025Reaction: R + I => RI; R, I, Rate Law: yeast*gamma*R*I

States:

NameDescription
G3G3
II
rIrI
G1G1
RIRI
G2G2
G0G0
R2R2
rRrR
RR

Karlstaedt2012 - CardioNet, A Human Metabolic Network: MODEL1212040000v0.0.1

Karlstaedt2012 - CardioNet, A Human Metabolic NetworkCardioNet is a functionally validated metabolic network of the huma…

Details

BACKGROUND: Availability of oxygen and nutrients in the coronary circulation is a crucial determinant of cardiac performance. Nutrient composition of coronary blood may significantly vary in specific physiological and pathological conditions, for example, administration of special diets, long-term starvation, physical exercise or diabetes. Quantitative analysis of cardiac metabolism from a systems biology perspective may help to a better understanding of the relationship between nutrient supply and efficiency of metabolic processes required for an adequate cardiac output. RESULTS: Here we present CardioNet, the first large-scale reconstruction of the metabolic network of the human cardiomyocyte comprising 1793 metabolic reactions, including 560 transport processes in six compartments. We use flux-balance analysis to demonstrate the capability of the network to accomplish a set of 368 metabolic functions required for maintaining the structural and functional integrity of the cell. Taking the maintenance of ATP, biosynthesis of ceramide, cardiolipin and further important phospholipids as examples, we analyse how a changed supply of glucose, lactate, fatty acids and ketone bodies may influence the efficiency of these essential processes. CONCLUSIONS: CardioNet is a functionally validated metabolic network of the human cardiomyocyte that enables theorectical studies of cellular metabolic processes crucial for the accomplishment of an adequate cardiac output. link: http://identifiers.org/pubmed/22929619

Karlstaedt2019 - G6P accumulation via Phosphoglucose isomerase inhibition in heart muscles: MODEL1910170001v0.0.1

This model is described in the article: **Glucose 6-phosphate accumulates via phosphoglucose isomerase inhibition in he…

Details

Rationale: Metabolic and structural remodeling is a hallmark of heart failure. This remodeling involves activation of the mammalian target of rapamycin (mTOR) signaling pathway, but little is known on how intermediary metabolites are integrated as metabolic signals. Objective: We investigated the metabolic control of cardiac glycolysis and explored the potential of glucose 6-phosphate to regulate glycolytic flux and mTOR activation. Methods and Results: We developed a kinetic model of cardiomyocyte carbohydrate metabolism, CardioGlyco, to study the metabolic control of myocardial glycolysis and glucose 6-phosphate levels. Metabolic control analysis revealed that glucose 6-phosphate concentration is dependent on phosphoglucose isomerase activity. Next, we integrated ex vivo tracer studies with mathematical simulations to test how changes in glucose supply and glycolytic flux affect mTOR activation. Nutrient deprivation promoted a tight coupling between glucose uptake and oxidation, glucose 6-phosphate reduction, and increased protein-protein interaction between hexokinase II and mTOR. We validated the in silico modeling in cultured adult mouse ventricular cardiomyocytes by modulating phosphoglucose isomerase activity using erythrose 4-phosphate. Inhibition of glycolytic flux at the level of phosphoglucose isomerase caused glucose 6-phosphate accumulation, which correlated with increased mTOR activation. Using click chemistry, we labeled newly synthesized proteins and confirmed that inhibition of phosphoglucose isomerase increases protein synthesis. Conclusions: The reduction of phosphoglucose isomerase activity directly affects myocyte growth by regulating mTOR activation. link: http://identifiers.org/pubmed/31698999

Kavšček2015 - Genome-scale metabolic model of Yarrowia lipolytica (iMK735): MODEL1510060001v0.0.1

Kavšček2015 - Genome-scale metabolic model of Yarrowia lipolytica (iMK735)This model is described in the article: [Opti…

Details

Yarrowia lipolytica is a non-conventional yeast that is extensively investigated for its ability to excrete citrate or to accumulate large amounts of storage lipids, which is of great significance for single cell oil production. Both traits are thus of interest for basic research as well as for biotechnological applications but they typically occur simultaneously thus lowering the respective yields. Therefore, engineering of strains with high lipid content relies on novel concepts such as computational simulation to better understand the two competing processes and to eliminate citrate excretion.Using a genome-scale model (GSM) of baker's yeast as a scaffold, we reconstructed the metabolic network of Y. lipolytica and optimized it for use in flux balance analysis (FBA), with the aim to simulate growth and lipid production phases of this yeast. We validated our model and found the predictions of the growth behavior of Y. lipolytica in excellent agreement with experimental data. Based on these data, we successfully designed a fed-batch strategy to avoid citrate excretion during the lipid production phase. Further analysis of the network suggested that the oxygen demand of Y. lipolytica is reduced upon induction of lipid synthesis. According to this finding we hypothesized that a reduced aeration rate might induce lipid accumulation. This prediction was indeed confirmed experimentally. In a fermentation combining these two strategies lipid content of the biomass was increased by 80%, and lipid yield was improved more than four-fold, compared to standard conditions.Genome scale network reconstructions provide a powerful tool to predict the effects of genetic modifications and the metabolic response to environmental conditions. The high accuracy and the predictive value of a newly reconstructed GSM of Y. lipolytica to optimize growth conditions for lipid accumulation are demonstrated. Based on these findings, further strategies for engineering Y. lipolytica towards higher efficiency in single cell oil production are discussed. link: http://identifiers.org/pubmed/26503450

Kawka2014 - Revealing the role of SGK1 in the dynamics of medulloblastoma using a mathematical model: MODEL1912090002v0.0.1

This is a mathematical model of the Wnt signaling pathway in medulloblastoma comprised of two compartments. Composed of…

Details

Deregulation of signaling pathways and subsequent abnormal interactions of downstream genes very often results in carcinogenesis. In this paper, we propose a two-compartment model describing intricate dynamics of the target genes of the Wnt signaling pathway in medulloblastoma. The system of nine nonlinear ordinary differential equations accounts for the formation and dissociation of complexes as well as for the transcription, translation and transport between the cytoplasm and the nucleus. We focus on the interplay between MYC and SGK1 (serum and glucocorticoid-inducible kinase 1), which are the products of Wnt/β-catenin signaling pathway, and GSK3β (glycogen synthase kinase). Numerical simulations of the model solutions yield a better understanding of the process and indicate the importance of the SGK1 gene in the development of medulloblastoma, which has been confirmed in our recent experiments. The model is calibrated based on the gene expression microarray data for two types of medulloblastoma, characterized by monosomy and trisomy of chromosome 6q to highlight the difference between diagnoses. link: http://identifiers.org/pubmed/24685888

Keener2001_OscillatoryInsulinSecretionModel: MODEL1201140001v0.0.1

This a model from the article: Diffusion induced oscillatory insulin secretion. Keener JP. Bull Math Biol. 2001 Jul…

Details

Oscillatory secretion of insulin has been observed in many different experimental preparations. Here we examine a mathematical model for in vitro insulin secretion from pancreatic beta cells in a flow-through reactor. The analysis shows that oscillations result because of an important interplay between flow rate of the reactor and insulin diffusion. In particular, if the ratio of flow rate to volume of the reaction bed is too large, oscillations are eliminated, in contradiction to the conclusions of Maki and Keizer (L. W. Maki and Keizer J. Mathematical analysis of a proposed mechanism for oscillatory insulin secretion in perifused HIT-15 cells. Bull. Math. Biol., 57(1995), 569-591). Furthermore, with reasonable numbers for the experimental parameters and the diffusion of insulin, the model equations do not exhibit oscillations. link: http://identifiers.org/pubmed/11497161

Kees2018 - Genome-scale constraint-based model of the mucin-degrader <I>Akkermansia muciniphila</I>: MODEL1710040000v0.0.1

Kees2018 - Genome-scale constraint-based model of the mucin-degrader <I>Akkermansia muciniphila</I>This model is descr…

Details

The abundance of the human intestinal symbiont Akkermansia muciniphila has found to be inversely correlated with several diseases, including metabolic syndrome and obesity. A. muciniphila is known to use mucin as sole carbon and nitrogen source. To study the physiology and the potential for therapeutic applications of this bacterium, we designed a defined minimal medium. The composition of the medium was based on the genome-scale metabolic model of A. muciniphila and the composition of mucin. Our results indicate that A. muciniphila does not code for GlmS, the enzyme that mediates the conversion of fructose-6-phosphate (Fru6P) to glucosamine-6-phosphate (GlcN6P), which is essential in peptidoglycan formation. The only annotated enzyme that could mediate this conversion is Amuc-NagB on locus Amuc_1822. We found that Amuc-NagB was unable to form GlcN6P from Fru6P at physiological conditions, while it efficiently catalyzed the reverse reaction. To overcome this inability, N-acetylglucosamine needs to be present in the medium for A. muciniphila growth. With these findings, the genome-scale metabolic model was updated and used to accurately predict growth of A. muciniphila on synthetic media. The finding that A. muciniphila has a necessity for GlcNAc, which is present in mucin further prompts the adaptation to its mucosal niche. link: http://identifiers.org/pubmed/29377524

Keizer1996_Ryanodine_receptor_adaptation: BIOMD0000000060v0.0.1

The model reproduces the time profile of Open probability of the ryanodine receptor as shown in Fig 2A and 2B of the pap…

Details

A simplified mechanism that mimics "adaptation" of the ryanodine receptor (RyR) has been developed and its significance for Ca2+(-)induced Ca2+ release and Ca2+ oscillations investigated. For parameters that reproduce experimental data for the RyR from cardiac cells, adaptation of the RyR in combination with sarco/endoplasmic reticulum Ca2+ ATPase Ca2+ pumps in the internal stores can give rise to either low [Cai2+] steady states or Ca2+ oscillations coexisting with unphysiologically high [Cai2+] steady states. In this closed-cell-type model rapid, adaptation-dependent Ca2+ oscillations occur only in limited ranges of parameters. In the presence of Ca2+ influx and efflux from outside the cell (open-cell model) Ca2+ oscillations occur for a wide range of physiological parameter values and have a period that is determined by the rate of Ca2+ refilling of the stores. Although the rate of adaptation of the RyR has a role in determining the shape and the period of the Ca2+ spike, it is not essential for their existence. This is in marked contrast with what is observed for the inositol 1,4,5-trisphosphate receptor for which the biphasic activation and inhibition of its activity by Ca2+ are sufficient to produce oscillations. Results for this model are compared with those based on Ca2+(-)induced Ca2+ release alone in the bullfrog sympathetic neuron. This kinetic model should be suitable for analyzing phenomena associated with "Ca2+ sparks," including their merger into Ca2+ waves in cardiac myocytes. link: http://identifiers.org/pubmed/8968617

Parameters:

NameDescription
kc_minus=0.1 per_second; kc_plus=1.75 per_secondReaction: Po1 => Pc2; Po1, Rate Law: kc_plus*Po1-kc_minus*Pc2
ka_plus=1500.0 microM-4sec-1; ka_minus=28.8 per_second; Ca=0.9 microM; n=4.0 dimensionlessReaction: Po1 => Pc1; Po1, Rate Law: ka_minus*Po1-ka_plus*Ca^n*Pc1
m=3.0 dimensionless; kb_plus=1500.0 microM-3sec-1; kb_minus=385.9 per_second; Ca=0.9 microMReaction: Po1 => Po2; Po1, Rate Law: kb_plus*Ca^m*Po1-kb_minus*Po2

States:

NameDescription
Po2[Ryanodine receptor 1]
Pc2[Ryanodine receptor 1]
Pc1[Ryanodine receptor 1]
Po1[Ryanodine receptor 1]

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei - MODEL B: BIOMD0000000514v0.0.1

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei - MODEL BThere are six models (Model A, B, C, C-fru…

Details

Dynamic models of metabolism can be useful in identifying potential drug targets, especially in unicellular organisms. A model of glycolysis in the causative agent of human African trypanosomiasis, Trypanosoma brucei, has already shown the utility of this approach. Here we add the pentose phosphate pathway (PPP) of T. brucei to the glycolytic model. The PPP is localized to both the cytosol and the glycosome and adding it to the glycolytic model without further adjustments leads to a draining of the essential bound-phosphate moiety within the glycosome. This phosphate "leak" must be resolved for the model to be a reasonable representation of parasite physiology. Two main types of theoretical solution to the problem could be identified: (i) including additional enzymatic reactions in the glycosome, or (ii) adding a mechanism to transfer bound phosphates between cytosol and glycosome. One example of the first type of solution would be the presence of a glycosomal ribokinase to regenerate ATP from ribose 5-phosphate and ADP. Experimental characterization of ribokinase in T. brucei showed that very low enzyme levels are sufficient for parasite survival, indicating that other mechanisms are required in controlling the phosphate leak. Examples of the second type would involve the presence of an ATP:ADP exchanger or recently described permeability pores in the glycosomal membrane, although the current absence of identified genes encoding such molecules impedes experimental testing by genetic manipulation. Confronted with this uncertainty, we present a modeling strategy that identifies robust predictions in the context of incomplete system characterization. We illustrate this strategy by exploring the mechanism underlying the essential function of one of the PPP enzymes, and validate it by confirming the model predictions experimentally. link: http://identifiers.org/pubmed/24339766

Parameters:

NameDescription
PPI_c_Keq=5.6; PPI_c_Vmax=72.0; PPI_c_KmRul5P=1.4; PPI_c_KmRib5P=4.0Reaction: Rul5P_c => Rib5P_c; Rul5P_c, Rib5P_c, Rate Law: PPI_c_Vmax*Rul5P_c*(1-Rib5P_c/(PPI_c_Keq*Rul5P_c))/(PPI_c_KmRul5P*(1+Rul5P_c/PPI_c_KmRul5P+Rib5P_c/PPI_c_KmRib5P))
GDA_g_k=600.0Reaction: Gly3P_g + DHAP_c => Gly3P_c + DHAP_g; Gly3P_g, DHAP_c, Gly3P_c, DHAP_g, Rate Law: Gly3P_g*GDA_g_k*DHAP_c-Gly3P_c*GDA_g_k*DHAP_g
_3PGAT_g_k=250.0Reaction: _3PGA_g => _3PGA_c; _3PGA_g, _3PGA_c, Rate Law: _3PGAT_g_k*_3PGA_g-_3PGAT_g_k*_3PGA_c
GPO_c_Vmax=368.0; GPO_c_KmGly3P=1.7Reaction: Gly3P_c => DHAP_c; Gly3P_c, Rate Law: GPO_c_Vmax*Gly3P_c/(GPO_c_KmGly3P*(1+Gly3P_c/GPO_c_KmGly3P))
HXK_g_Vmax=1774.68; HXK_g_KmADP=0.126; HXK_g_Keq=759.0; HXK_g_KmATP=0.116; HXK_g_KmGlc6P=2.7; HXK_g_KmGlc=0.1Reaction: ATP_g + Glc_g => Glc6P_g + ADP_g; Glc_g, ATP_g, Glc6P_g, ADP_g, Rate Law: HXK_g_Vmax*Glc_g*ATP_g*(1-Glc6P_g*ADP_g/(HXK_g_Keq*Glc_g*ATP_g))/(HXK_g_KmGlc*HXK_g_KmATP*(1+Glc_g/HXK_g_KmGlc+Glc6P_g/HXK_g_KmGlc6P)*(1+ATP_g/HXK_g_KmATP+ADP_g/HXK_g_KmADP))
PGI_g_Vmax=1305.0; PGI_g_Ki6PG=0.14; PGI_g_KmGlc6P=0.4; PGI_g_Keq=0.457; PGI_g_KmFru6P=0.12Reaction: Glc6P_g => Fru6P_g; _6PG_g, Glc6P_g, Fru6P_g, _6PG_g, Rate Law: PGI_g_Vmax*Glc6P_g*(1-Fru6P_g/(PGI_g_Keq*Glc6P_g))/(PGI_g_KmGlc6P*(1+Glc6P_g/PGI_g_KmGlc6P+Fru6P_g/PGI_g_KmFru6P+_6PG_g/PGI_g_Ki6PG))
NADPHu_c_k=2.0Reaction: NADPH_c => NADP_c; NADPH_c, Rate Law: NADPHu_c_k*NADPH_c
ALD_g_KmDHAP=0.015; ALD_g_KiGA3P=0.098; ALD_g_KmGA3P=0.067; ALD_g_Vmax=560.0; ALD_g_KmFru16BP=0.009; ALD_g_KiADP=1.51; ALD_g_KiAMP=3.65; ALD_g_Keq=0.084; ALD_g_KiATP=0.68Reaction: Fru16BP_g => GA3P_g + DHAP_g; ATP_g, ADP_g, AMP_g, Fru16BP_g, GA3P_g, DHAP_g, ATP_g, ADP_g, AMP_g, Rate Law: ALD_g_Vmax*Fru16BP_g*(1-GA3P_g*DHAP_g/(Fru16BP_g*ALD_g_Keq))/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP)*(1+GA3P_g/ALD_g_KmGA3P+DHAP_g/ALD_g_KmDHAP+Fru16BP_g/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))+GA3P_g*DHAP_g/(ALD_g_KmGA3P*ALD_g_KmDHAP)+Fru16BP_g*GA3P_g/(ALD_g_KmFru16BP*ALD_g_KiGA3P*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))))
G6PP_c_KmGlc6P=5.6; G6PP_c_Vmax=28.0; G6PP_c_Keq=263.0; G6PP_c_KmGlc=5.6Reaction: Glc6P_c => Glc_c; Glc6P_c, Glc_c, Rate Law: G6PP_c_Vmax*Glc6P_c*(1-Glc_c/(G6PP_c_Keq*Glc6P_c))/(G6PP_c_KmGlc6P*(1+Glc6P_c/G6PP_c_KmGlc6P+Glc_c/G6PP_c_KmGlc))
PYK_c_KmPyr=50.0; PYK_c_KiADP=0.64; PYK_c_Vmax=1020.0; PYK_c_KmADP=0.114; PYK_c_Keq=10800.0; PYK_c_KiATP=0.57; PYK_c_KmATP=15.0; PYK_c_KmPEP=0.34; PYK_c_n=2.5Reaction: PEP_c + ADP_c => Pyr_c + ATP_c; ADP_c, Pyr_c, ATP_c, PEP_c, Rate Law: PYK_c_Vmax*ADP_c*(1-Pyr_c*ATP_c/(PYK_c_Keq*PEP_c*ADP_c))*(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n/(PYK_c_KmADP*(1+(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n+Pyr_c/PYK_c_KmPyr)*(1+ADP_c/PYK_c_KmADP+ATP_c/PYK_c_KmATP))
ATPu_c_k=50.0Reaction: ATP_c => ADP_c; ATP_c, ADP_c, Rate Law: ATPu_c_k*ATP_c/ADP_c
G6PDH_g_KmNADPH=1.0E-4; G6PDH_g_Keq=5.02; G6PDH_g_KmNADP=0.0094; G6PDH_g_KmGlc6P=0.058; G6PDH_g_Vmax=8.4; G6PDH_g_Km6PGL=0.04Reaction: Glc6P_g + NADP_g => _6PGL_g + NADPH_g; Glc6P_g, NADP_g, _6PGL_g, NADPH_g, Rate Law: G6PDH_g_Vmax*Glc6P_g*NADP_g*(1-_6PGL_g*NADPH_g/(G6PDH_g_Keq*Glc6P_g*NADP_g))/(G6PDH_g_KmGlc6P*G6PDH_g_KmNADP*(1+Glc6P_g/G6PDH_g_KmGlc6P+_6PGL_g/G6PDH_g_Km6PGL)*(1+NADP_g/G6PDH_g_KmNADP+NADPH_g/G6PDH_g_KmNADPH))
TR_c_KmTS2=0.0069; TR_c_KmTSH2=0.0018; TR_c_KmNADPH=7.7E-4; TR_c_Vmax=252.0; TR_c_Keq=434.0; TR_c_KmNADP=0.081Reaction: TS2_c + NADPH_c => NADP_c + TSH2_c; TS2_c, NADPH_c, TSH2_c, NADP_c, Rate Law: TR_c_Vmax*TS2_c*NADPH_c*(1-TSH2_c*NADP_c/(TR_c_Keq*TS2_c*NADPH_c))/(TR_c_KmTS2*TR_c_KmNADPH*(1+TS2_c/TR_c_KmTS2+TSH2_c/TR_c_KmTSH2)*(1+NADPH_c/TR_c_KmNADPH+NADP_c/TR_c_KmNADP))
AK_g_k2=1000.0; AK_g_k1=480.0Reaction: ADP_g => AMP_g + ATP_g; ADP_g, AMP_g, ATP_g, Rate Law: AK_g_k1*ADP_g^2-AMP_g*ATP_g*AK_g_k2
PyrT_c_Vmax=230.0; PyrT_c_KmPyr=1.96Reaction: Pyr_c => Pyr_e; Pyr_c, Rate Law: PyrT_c_Vmax*Pyr_c/(PyrT_c_KmPyr*(1+Pyr_c/PyrT_c_KmPyr))
G6PDH_c_Vmax=8.4; G6PDH_c_Keq=5.02; G6PDH_c_KmNADP=0.0094; G6PDH_c_KmNADPH=1.0E-4; G6PDH_c_Km6PGL=0.04; G6PDH_c_KmGlc6P=0.058Reaction: Glc6P_c + NADP_c => NADPH_c + _6PGL_c; Glc6P_c, NADP_c, _6PGL_c, NADPH_c, Rate Law: G6PDH_c_Vmax*Glc6P_c*NADP_c*(1-_6PGL_c*NADPH_c/(G6PDH_c_Keq*Glc6P_c*NADP_c))/(G6PDH_c_KmGlc6P*G6PDH_c_KmNADP*(1+Glc6P_c/G6PDH_c_KmGlc6P+_6PGL_c/G6PDH_c_Km6PGL)*(1+NADP_c/G6PDH_c_KmNADP+NADPH_c/G6PDH_c_KmNADPH))
HXK_c_KmATP=0.116; HXK_c_KmGlc=0.1; HXK_c_Vmax=154.32; HXK_c_KmADP=0.126; HXK_c_Keq=759.0; HXK_c_KmGlc6P=2.7Reaction: Glc_c + ATP_c => Glc6P_c + ADP_c; Glc_c, ATP_c, Glc6P_c, ADP_c, Rate Law: HXK_c_Vmax*Glc_c*ATP_c*(1-Glc6P_c*ADP_c/(HXK_c_Keq*Glc_c*ATP_c))/(HXK_c_KmGlc*HXK_c_KmATP*(1+Glc_c/HXK_c_KmGlc+Glc6P_c/HXK_c_KmGlc6P)*(1+ATP_c/HXK_c_KmATP+ADP_c/HXK_c_KmADP))
GAPDH_g_Vmax=720.9; GAPDH_g_Km13BPGA=0.1; GAPDH_g_KmNAD=0.45; GAPDH_g_KmNADH=0.02; GAPDH_g_KmGA3P=0.15; GAPDH_g_Keq=0.066Reaction: GA3P_g + NAD_g + Pi_g => NADH_g + _13BPGA_g; GA3P_g, NAD_g, _13BPGA_g, NADH_g, Rate Law: GAPDH_g_Vmax*GA3P_g*NAD_g*(1-_13BPGA_g*NADH_g/(GAPDH_g_Keq*GA3P_g*NAD_g))/(GAPDH_g_KmGA3P*GAPDH_g_KmNAD*(1+GA3P_g/GAPDH_g_KmGA3P+_13BPGA_g/GAPDH_g_Km13BPGA)*(1+NAD_g/GAPDH_g_KmNAD+NADH_g/GAPDH_g_KmNADH))
PGL_g_Km6PGL=0.05; PGL_g_Km6PG=0.05; PGL_g_Vmax=5.0; PGL_g_Keq=20000.0; PGL_g_k=0.055Reaction: _6PGL_g => _6PG_g; _6PGL_g, _6PG_g, Rate Law: glycosome*PGL_g_k*(_6PGL_g-_6PG_g/PGL_g_Keq)+PGL_g_Vmax*_6PGL_g*(1-_6PG_g/(PGL_g_Keq*_6PGL_g))/(PGL_g_Km6PGL*(1+_6PGL_g/PGL_g_Km6PGL+_6PG_g/PGL_g_Km6PG))
TOX_c_k=2.0Reaction: TSH2_c => TS2_c; TSH2_c, Rate Law: TOX_c_k*TSH2_c
ENO_c_Vmax=598.0; ENO_c_Km2PGA=0.054; ENO_c_Keq=4.17; ENO_c_KmPEP=0.24Reaction: _2PGA_c => PEP_c; _2PGA_c, PEP_c, Rate Law: ENO_c_Vmax*_2PGA_c*(1-PEP_c/(ENO_c_Keq*_2PGA_c))/(ENO_c_Km2PGA*(1+_2PGA_c/ENO_c_Km2PGA+PEP_c/ENO_c_KmPEP))
PFK_g_Vmax=1708.0; PFK_g_KsATP=0.0393; PFK_g_KmFru6P=0.999; PFK_g_KmADP=1.0; PFK_g_KmATP=0.0648; PFK_g_Ki2=10.7; PFK_g_Ki1=15.8; PFK_g_Keq=1035.0Reaction: ATP_g + Fru6P_g => Fru16BP_g + ADP_g; Fru6P_g, ATP_g, Fru16BP_g, ADP_g, Rate Law: PFK_g_Vmax*PFK_g_Ki1*Fru6P_g*ATP_g*(1-Fru16BP_g*ADP_g/(PFK_g_Keq*Fru6P_g*ATP_g))/(PFK_g_KmFru6P*PFK_g_KmATP*(Fru16BP_g+PFK_g_Ki1)*(PFK_g_KsATP/PFK_g_KmATP+Fru6P_g/PFK_g_KmFru6P+ATP_g/PFK_g_KmATP+ADP_g/PFK_g_KmADP+Fru16BP_g*ADP_g/(PFK_g_KmADP*PFK_g_Ki2)+Fru6P_g*ATP_g/(PFK_g_KmFru6P*PFK_g_KmATP)))
_6PGDH_g_KmNADP=0.001; _6PGDH_g_KmNADPH=6.0E-4; _6PGDH_g_Keq=47.0; _6PGDH_g_Km6PG=0.0035; _6PGDH_g_KmRul5P=0.03; _6PGDH_g_Vmax=10.6Reaction: _6PG_g + NADP_g => Rul5P_g + CO2_g + NADPH_g; _6PG_g, NADP_g, Rul5P_g, NADPH_g, Rate Law: _6PGDH_g_Vmax*_6PG_g*NADP_g*(1-Rul5P_g*NADPH_g/(_6PGDH_g_Keq*_6PG_g*NADP_g))/(_6PGDH_g_Km6PG*_6PGDH_g_KmNADP*(1+_6PG_g/_6PGDH_g_Km6PG+Rul5P_g/_6PGDH_g_KmRul5P)*(1+NADP_g/_6PGDH_g_KmNADP+NADPH_g/_6PGDH_g_KmNADPH))
GlcT_c_Vmax=111.7; GlcT_c_KmGlc=1.0; GlcT_c_alpha=0.75Reaction: Glc_e => Glc_c; Glc_e, Glc_c, Rate Law: GlcT_c_Vmax*(Glc_e-Glc_c)/(GlcT_c_KmGlc+Glc_e+Glc_c+GlcT_c_alpha*Glc_e*Glc_c/GlcT_c_KmGlc)
TPI_g_Keq=0.046; TPI_g_KmDHAP=1.2; TPI_g_KmGA3P=0.25; TPI_g_Vmax=999.3Reaction: DHAP_g => GA3P_g; DHAP_g, GA3P_g, Rate Law: TPI_g_Vmax*DHAP_g*(1-GA3P_g/(TPI_g_Keq*DHAP_g))/(TPI_g_KmDHAP*(1+DHAP_g/TPI_g_KmDHAP+GA3P_g/TPI_g_KmGA3P))
NADPHu_g_k=2.0Reaction: NADPH_g => NADP_g; NADPH_g, Rate Law: NADPHu_g_k*NADPH_g
PGAM_c_Km3PGA=0.15; PGAM_c_Vmax=225.0; PGAM_c_Km2PGA=0.16; PGAM_c_Keq=0.17Reaction: _3PGA_c => _2PGA_c; _3PGA_c, _2PGA_c, Rate Law: PGAM_c_Vmax*_3PGA_c*(1-_2PGA_c/(PGAM_c_Keq*_3PGA_c))/(PGAM_c_Km3PGA*(1+_3PGA_c/PGAM_c_Km3PGA+_2PGA_c/PGAM_c_Km2PGA))
G3PDH_g_KmDHAP=0.1; G3PDH_g_KmNAD=0.4; G3PDH_g_Vmax=465.0; G3PDH_g_Keq=17085.0; G3PDH_g_KmNADH=0.01; G3PDH_g_KmGly3P=2.0Reaction: NADH_g + DHAP_g => Gly3P_g + NAD_g; DHAP_g, NADH_g, Gly3P_g, NAD_g, Rate Law: G3PDH_g_Vmax*DHAP_g*NADH_g*(1-Gly3P_g*NAD_g/(G3PDH_g_Keq*DHAP_g*NADH_g))/(G3PDH_g_KmDHAP*G3PDH_g_KmNADH*(1+DHAP_g/G3PDH_g_KmDHAP+Gly3P_g/G3PDH_g_KmGly3P)*(1+NADH_g/G3PDH_g_KmNADH+NAD_g/G3PDH_g_KmNAD))
GK_g_Keq=8.37E-4; GK_g_KmATP=0.24; GK_g_KmGly=0.44; GK_g_KmADP=0.56; GK_g_KmGly3P=3.83; GK_g_Vmax=200.0Reaction: Gly3P_g + ADP_g => Gly_e + ATP_g; Gly3P_g, ADP_g, Gly_e, ATP_g, Rate Law: GK_g_Vmax*Gly3P_g*ADP_g*(1-Gly_e*ATP_g/(GK_g_Keq*Gly3P_g*ADP_g))/(GK_g_KmGly3P*GK_g_KmADP*(1+Gly3P_g/GK_g_KmGly3P+Gly_e/GK_g_KmGly)*(1+ADP_g/GK_g_KmADP+ATP_g/GK_g_KmATP))
GlcT_g_k2=250000.0; GlcT_g_k1=250000.0Reaction: Glc_c => Glc_g; Glc_c, Glc_g, Rate Law: GlcT_g_k1*Glc_c-GlcT_g_k2*Glc_g
PGL_c_Km6PG=0.05; PGL_c_k=0.055; PGL_c_Vmax=28.0; PGL_c_Km6PGL=0.05; PGL_c_Keq=20000.0Reaction: _6PGL_c => _6PG_c; _6PGL_c, _6PG_c, Rate Law: PGL_c_k*cytosol*(_6PGL_c-_6PG_c/PGL_c_Keq)+PGL_c_Vmax*_6PGL_c*(1-_6PG_c/(PGL_c_Keq*_6PGL_c))/(PGL_c_Km6PGL*(1+_6PGL_c/PGL_c_Km6PGL+_6PG_c/PGL_c_Km6PG))
_6PGDH_c_KmNADP=0.001; _6PGDH_c_Keq=47.0; _6PGDH_c_Vmax=10.6; _6PGDH_c_KmNADPH=6.0E-4; _6PGDH_c_Km6PG=0.0035; _6PGDH_c_KmRul5P=0.03Reaction: NADP_c + _6PG_c => CO2_c + NADPH_c + Rul5P_c; _6PG_c, NADP_c, Rul5P_c, NADPH_c, Rate Law: _6PGDH_c_Vmax*_6PG_c*NADP_c*(1-Rul5P_c*NADPH_c/(_6PGDH_c_Keq*_6PG_c*NADP_c))/(_6PGDH_c_Km6PG*_6PGDH_c_KmNADP*(1+_6PG_c/_6PGDH_c_Km6PG+Rul5P_c/_6PGDH_c_KmRul5P)*(1+NADP_c/_6PGDH_c_KmNADP+NADPH_c/_6PGDH_c_KmNADPH))
PPI_g_KmRul5P=1.4; PPI_g_Vmax=72.0; PPI_g_Keq=5.6; PPI_g_KmRib5P=4.0Reaction: Rul5P_g => Rib5P_g; Rul5P_g, Rib5P_g, Rate Law: PPI_g_Vmax*Rul5P_g*(1-Rib5P_g/(PPI_g_Keq*Rul5P_g))/(PPI_g_KmRul5P*(1+Rul5P_g/PPI_g_KmRul5P+Rib5P_g/PPI_g_KmRib5P))
AK_c_k1=480.0; AK_c_k2=1000.0Reaction: ADP_c => AMP_c + ATP_c; ADP_c, AMP_c, ATP_c, Rate Law: AK_c_k1*ADP_c^2-AMP_c*ATP_c*AK_c_k2
PGK_g_Km13BPGA=0.003; PGK_g_Vmax=2862.0; PGK_g_KmADP=0.1; PGK_g_Km3PGA=1.62; PGK_g_KmATP=0.29; PGK_g_Keq=3377.0Reaction: _13BPGA_g + ADP_g => _3PGA_g + ATP_g; _13BPGA_g, ADP_g, _3PGA_g, ATP_g, Rate Law: PGK_g_Vmax*_13BPGA_g*ADP_g*(1-_3PGA_g*ATP_g/(PGK_g_Keq*_13BPGA_g*ADP_g))/(PGK_g_Km13BPGA*PGK_g_KmADP*(1+_13BPGA_g/PGK_g_Km13BPGA+_3PGA_g/PGK_g_Km3PGA)*(1+ADP_g/PGK_g_KmADP+ATP_g/PGK_g_KmATP))

States:

NameDescription
6PG g[6-phospho-D-gluconic acid]
6PG c[6-phospho-D-gluconic acid]
TS2 c[trypanothione]
Rul5P g[D-ribulose 5-phosphate(2-)]
Glc6P c[D-glucopyranose 6-phosphate]
PEP c[phosphoenolpyruvate]
ATP g[ATP]
CO2 g[carbon dioxide]
Glc c[glucose]
GA3P g[glyceraldehyde 3-phosphate]
Fru16BP g[alpha-D-fructofuranose 1,6-bisphosphate]
Glc g[glucose]
Glc e[glucose]
TSH2 c[trypanothione disulfide]
Pyr e[pyruvate]
ADP g[ADP]
13BPGA g[683]
NADP c[NADP(+)]
DHAP g[glycerone phosphate(2-)]
NAD g[NAD]
NADH g[NADH]
Pyr c[pyruvate]
CO2 c[carbon dioxide]
6PGL g[6-phosphogluconolactonase3.1.1.17]
Fru6P g[444848]
2PGA c[59]
Gly3P c[glycerol 1-phosphate]
Rib5P c[aldehydo-D-ribose 5-phosphate(2-)]
3PGA c[3-phospho-D-glyceric acid]
NADP g[NADP(+)]
Rib5P g[aldehydo-D-ribose 5-phosphate(2-)]
Pi g[phosphatidylinositol]
Glc6P g[D-glucopyranose 6-phosphate]
ATP c[ATP]
DHAP c[glycerone phosphate(2-)]
NADPH c[NADPH]
Gly e[glycerol]
6PGL c[6-phosphogluconolactonase3.1.1.17]
Gly3P g[glycerol 1-phosphate]
ADP c[ADP]
AMP g[AMP]
Rul5P c[D-ribulose 5-phosphate(2-)]
3PGA g[3-phospho-D-glyceric acid]
AMP c[AMP]
NADPH g[NADPH]

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei - MODEL C (with glucosomal ribokinase): BIOMD0000000510v0.0.1

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei -MODEL C (with glucosomal ribokinase)There are six…

Details

Dynamic models of metabolism can be useful in identifying potential drug targets, especially in unicellular organisms. A model of glycolysis in the causative agent of human African trypanosomiasis, Trypanosoma brucei, has already shown the utility of this approach. Here we add the pentose phosphate pathway (PPP) of T. brucei to the glycolytic model. The PPP is localized to both the cytosol and the glycosome and adding it to the glycolytic model without further adjustments leads to a draining of the essential bound-phosphate moiety within the glycosome. This phosphate "leak" must be resolved for the model to be a reasonable representation of parasite physiology. Two main types of theoretical solution to the problem could be identified: (i) including additional enzymatic reactions in the glycosome, or (ii) adding a mechanism to transfer bound phosphates between cytosol and glycosome. One example of the first type of solution would be the presence of a glycosomal ribokinase to regenerate ATP from ribose 5-phosphate and ADP. Experimental characterization of ribokinase in T. brucei showed that very low enzyme levels are sufficient for parasite survival, indicating that other mechanisms are required in controlling the phosphate leak. Examples of the second type would involve the presence of an ATP:ADP exchanger or recently described permeability pores in the glycosomal membrane, although the current absence of identified genes encoding such molecules impedes experimental testing by genetic manipulation. Confronted with this uncertainty, we present a modeling strategy that identifies robust predictions in the context of incomplete system characterization. We illustrate this strategy by exploring the mechanism underlying the essential function of one of the PPP enzymes, and validate it by confirming the model predictions experimentally. link: http://identifiers.org/pubmed/24339766

Parameters:

NameDescription
RK_g_Keq=0.0036; RK_g_KmRib=0.51; RK_g_Vmax=10.0; RK_g_KmRib5P=0.39; RK_g_KmADP=0.25; RK_g_KmATP=0.24Reaction: ADP_g + Rib5P_g => ATP_g + Rib_g; Rib5P_g, ADP_g, Rib_g, ATP_g, Rate Law: RK_g_Vmax*Rib5P_g*ADP_g*(1-Rib_g*ATP_g/(RK_g_Keq*Rib5P_g*ADP_g))/(RK_g_KmRib5P*RK_g_KmADP*(1+Rib5P_g/RK_g_KmRib5P+Rib_g/RK_g_KmRib)*(1+ADP_g/RK_g_KmADP+ATP_g/RK_g_KmATP))
PPI_c_Keq=5.6; PPI_c_Vmax=72.0; PPI_c_KmRul5P=1.4; PPI_c_KmRib5P=4.0Reaction: Rul5P_c => Rib5P_c; Rul5P_c, Rib5P_c, Rate Law: PPI_c_Vmax*Rul5P_c*(1-Rib5P_c/(PPI_c_Keq*Rul5P_c))/(PPI_c_KmRul5P*(1+Rul5P_c/PPI_c_KmRul5P+Rib5P_c/PPI_c_KmRib5P))
GDA_g_k=600.0Reaction: Gly3P_g + DHAP_c => Gly3P_c + DHAP_g; Gly3P_g, DHAP_c, Gly3P_c, DHAP_g, Rate Law: Gly3P_g*GDA_g_k*DHAP_c-Gly3P_c*GDA_g_k*DHAP_g
GPO_c_Vmax=368.0; GPO_c_KmGly3P=1.7Reaction: Gly3P_c => DHAP_c; Gly3P_c, Rate Law: GPO_c_Vmax*Gly3P_c/(GPO_c_KmGly3P*(1+Gly3P_c/GPO_c_KmGly3P))
_3PGAT_g_k=250.0Reaction: _3PGA_g => _3PGA_c; _3PGA_g, _3PGA_c, Rate Law: _3PGAT_g_k*_3PGA_g-_3PGAT_g_k*_3PGA_c
PGI_g_Vmax=1305.0; PGI_g_Ki6PG=0.14; PGI_g_KmGlc6P=0.4; PGI_g_Keq=0.457; PGI_g_KmFru6P=0.12Reaction: Glc6P_g => Fru6P_g; _6PG_g, Glc6P_g, Fru6P_g, _6PG_g, Rate Law: PGI_g_Vmax*Glc6P_g*(1-Fru6P_g/(PGI_g_Keq*Glc6P_g))/(PGI_g_KmGlc6P*(1+Glc6P_g/PGI_g_KmGlc6P+Fru6P_g/PGI_g_KmFru6P+_6PG_g/PGI_g_Ki6PG))
HXK_g_Vmax=1774.68; HXK_g_KmADP=0.126; HXK_g_Keq=759.0; HXK_g_KmATP=0.116; HXK_g_KmGlc6P=2.7; HXK_g_KmGlc=0.1Reaction: ATP_g + Glc_g => Glc6P_g + ADP_g; Glc_g, ATP_g, Glc6P_g, ADP_g, Rate Law: HXK_g_Vmax*Glc_g*ATP_g*(1-Glc6P_g*ADP_g/(HXK_g_Keq*Glc_g*ATP_g))/(HXK_g_KmGlc*HXK_g_KmATP*(1+Glc_g/HXK_g_KmGlc+Glc6P_g/HXK_g_KmGlc6P)*(1+ATP_g/HXK_g_KmATP+ADP_g/HXK_g_KmADP))
ALD_g_KmDHAP=0.015; ALD_g_KiGA3P=0.098; ALD_g_KmGA3P=0.067; ALD_g_Vmax=560.0; ALD_g_KmFru16BP=0.009; ALD_g_KiADP=1.51; ALD_g_KiAMP=3.65; ALD_g_Keq=0.084; ALD_g_KiATP=0.68Reaction: Fru16BP_g => GA3P_g + DHAP_g; ATP_g, ADP_g, AMP_g, Fru16BP_g, GA3P_g, DHAP_g, ATP_g, ADP_g, AMP_g, Rate Law: ALD_g_Vmax*Fru16BP_g*(1-GA3P_g*DHAP_g/(Fru16BP_g*ALD_g_Keq))/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP)*(1+GA3P_g/ALD_g_KmGA3P+DHAP_g/ALD_g_KmDHAP+Fru16BP_g/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))+GA3P_g*DHAP_g/(ALD_g_KmGA3P*ALD_g_KmDHAP)+Fru16BP_g*GA3P_g/(ALD_g_KmFru16BP*ALD_g_KiGA3P*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))))
NADPHu_c_k=2.0Reaction: NADPH_c => NADP_c; NADPH_c, Rate Law: NADPHu_c_k*NADPH_c
ATPu_c_k=50.0Reaction: ATP_c => ADP_c; ATP_c, ADP_c, Rate Law: ATPu_c_k*ATP_c/ADP_c
PYK_c_KmPyr=50.0; PYK_c_KiADP=0.64; PYK_c_Vmax=1020.0; PYK_c_KmADP=0.114; PYK_c_Keq=10800.0; PYK_c_KiATP=0.57; PYK_c_KmATP=15.0; PYK_c_KmPEP=0.34; PYK_c_n=2.5Reaction: PEP_c + ADP_c => Pyr_c + ATP_c; ADP_c, Pyr_c, ATP_c, PEP_c, Rate Law: PYK_c_Vmax*ADP_c*(1-Pyr_c*ATP_c/(PYK_c_Keq*PEP_c*ADP_c))*(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n/(PYK_c_KmADP*(1+(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n+Pyr_c/PYK_c_KmPyr)*(1+ADP_c/PYK_c_KmADP+ATP_c/PYK_c_KmATP))
G6PP_c_KmGlc6P=5.6; G6PP_c_Vmax=28.0; G6PP_c_Keq=263.0; G6PP_c_KmGlc=5.6Reaction: Glc6P_c => Glc_c; Glc6P_c, Glc_c, Rate Law: G6PP_c_Vmax*Glc6P_c*(1-Glc_c/(G6PP_c_Keq*Glc6P_c))/(G6PP_c_KmGlc6P*(1+Glc6P_c/G6PP_c_KmGlc6P+Glc_c/G6PP_c_KmGlc))
G6PDH_g_KmNADPH=1.0E-4; G6PDH_g_Keq=5.02; G6PDH_g_KmNADP=0.0094; G6PDH_g_KmGlc6P=0.058; G6PDH_g_Vmax=8.4; G6PDH_g_Km6PGL=0.04Reaction: Glc6P_g + NADP_g => _6PGL_g + NADPH_g; Glc6P_g, NADP_g, _6PGL_g, NADPH_g, Rate Law: G6PDH_g_Vmax*Glc6P_g*NADP_g*(1-_6PGL_g*NADPH_g/(G6PDH_g_Keq*Glc6P_g*NADP_g))/(G6PDH_g_KmGlc6P*G6PDH_g_KmNADP*(1+Glc6P_g/G6PDH_g_KmGlc6P+_6PGL_g/G6PDH_g_Km6PGL)*(1+NADP_g/G6PDH_g_KmNADP+NADPH_g/G6PDH_g_KmNADPH))
TR_c_KmTS2=0.0069; TR_c_KmTSH2=0.0018; TR_c_KmNADPH=7.7E-4; TR_c_Vmax=252.0; TR_c_Keq=434.0; TR_c_KmNADP=0.081Reaction: TS2_c + NADPH_c => NADP_c + TSH2_c; TS2_c, NADPH_c, TSH2_c, NADP_c, Rate Law: TR_c_Vmax*TS2_c*NADPH_c*(1-TSH2_c*NADP_c/(TR_c_Keq*TS2_c*NADPH_c))/(TR_c_KmTS2*TR_c_KmNADPH*(1+TS2_c/TR_c_KmTS2+TSH2_c/TR_c_KmTSH2)*(1+NADPH_c/TR_c_KmNADPH+NADP_c/TR_c_KmNADP))
AK_g_k2=1000.0; AK_g_k1=480.0Reaction: ADP_g => AMP_g + ATP_g; ADP_g, AMP_g, ATP_g, Rate Law: AK_g_k1*ADP_g^2-AMP_g*ATP_g*AK_g_k2
PyrT_c_Vmax=230.0; PyrT_c_KmPyr=1.96Reaction: Pyr_c => Pyr_e; Pyr_c, Rate Law: PyrT_c_Vmax*Pyr_c/(PyrT_c_KmPyr*(1+Pyr_c/PyrT_c_KmPyr))
G6PDH_c_Vmax=8.4; G6PDH_c_Keq=5.02; G6PDH_c_KmNADP=0.0094; G6PDH_c_KmNADPH=1.0E-4; G6PDH_c_Km6PGL=0.04; G6PDH_c_KmGlc6P=0.058Reaction: Glc6P_c + NADP_c => NADPH_c + _6PGL_c; Glc6P_c, NADP_c, _6PGL_c, NADPH_c, Rate Law: G6PDH_c_Vmax*Glc6P_c*NADP_c*(1-_6PGL_c*NADPH_c/(G6PDH_c_Keq*Glc6P_c*NADP_c))/(G6PDH_c_KmGlc6P*G6PDH_c_KmNADP*(1+Glc6P_c/G6PDH_c_KmGlc6P+_6PGL_c/G6PDH_c_Km6PGL)*(1+NADP_c/G6PDH_c_KmNADP+NADPH_c/G6PDH_c_KmNADPH))
HXK_c_KmATP=0.116; HXK_c_KmGlc=0.1; HXK_c_Vmax=154.32; HXK_c_KmADP=0.126; HXK_c_Keq=759.0; HXK_c_KmGlc6P=2.7Reaction: Glc_c + ATP_c => Glc6P_c + ADP_c; Glc_c, ATP_c, Glc6P_c, ADP_c, Rate Law: HXK_c_Vmax*Glc_c*ATP_c*(1-Glc6P_c*ADP_c/(HXK_c_Keq*Glc_c*ATP_c))/(HXK_c_KmGlc*HXK_c_KmATP*(1+Glc_c/HXK_c_KmGlc+Glc6P_c/HXK_c_KmGlc6P)*(1+ATP_c/HXK_c_KmATP+ADP_c/HXK_c_KmADP))
GAPDH_g_Vmax=720.9; GAPDH_g_Km13BPGA=0.1; GAPDH_g_KmNAD=0.45; GAPDH_g_KmNADH=0.02; GAPDH_g_KmGA3P=0.15; GAPDH_g_Keq=0.066Reaction: GA3P_g + NAD_g + Pi_g => NADH_g + _13BPGA_g; GA3P_g, NAD_g, _13BPGA_g, NADH_g, Rate Law: GAPDH_g_Vmax*GA3P_g*NAD_g*(1-_13BPGA_g*NADH_g/(GAPDH_g_Keq*GA3P_g*NAD_g))/(GAPDH_g_KmGA3P*GAPDH_g_KmNAD*(1+GA3P_g/GAPDH_g_KmGA3P+_13BPGA_g/GAPDH_g_Km13BPGA)*(1+NAD_g/GAPDH_g_KmNAD+NADH_g/GAPDH_g_KmNADH))
TOX_c_k=2.0Reaction: TSH2_c => TS2_c; TSH2_c, Rate Law: TOX_c_k*TSH2_c
PGL_g_Km6PGL=0.05; PGL_g_Km6PG=0.05; PGL_g_Vmax=5.0; PGL_g_Keq=20000.0; PGL_g_k=0.055Reaction: _6PGL_g => _6PG_g; _6PGL_g, _6PG_g, Rate Law: glycosome*PGL_g_k*(_6PGL_g-_6PG_g/PGL_g_Keq)+PGL_g_Vmax*_6PGL_g*(1-_6PG_g/(PGL_g_Keq*_6PGL_g))/(PGL_g_Km6PGL*(1+_6PGL_g/PGL_g_Km6PGL+_6PG_g/PGL_g_Km6PG))
ENO_c_Vmax=598.0; ENO_c_Km2PGA=0.054; ENO_c_Keq=4.17; ENO_c_KmPEP=0.24Reaction: _2PGA_c => PEP_c; _2PGA_c, PEP_c, Rate Law: ENO_c_Vmax*_2PGA_c*(1-PEP_c/(ENO_c_Keq*_2PGA_c))/(ENO_c_Km2PGA*(1+_2PGA_c/ENO_c_Km2PGA+PEP_c/ENO_c_KmPEP))
PFK_g_Vmax=1708.0; PFK_g_KsATP=0.0393; PFK_g_KmFru6P=0.999; PFK_g_KmADP=1.0; PFK_g_KmATP=0.0648; PFK_g_Ki2=10.7; PFK_g_Ki1=15.8; PFK_g_Keq=1035.0Reaction: ATP_g + Fru6P_g => Fru16BP_g + ADP_g; Fru6P_g, ATP_g, Fru16BP_g, ADP_g, Rate Law: PFK_g_Vmax*PFK_g_Ki1*Fru6P_g*ATP_g*(1-Fru16BP_g*ADP_g/(PFK_g_Keq*Fru6P_g*ATP_g))/(PFK_g_KmFru6P*PFK_g_KmATP*(Fru16BP_g+PFK_g_Ki1)*(PFK_g_KsATP/PFK_g_KmATP+Fru6P_g/PFK_g_KmFru6P+ATP_g/PFK_g_KmATP+ADP_g/PFK_g_KmADP+Fru16BP_g*ADP_g/(PFK_g_KmADP*PFK_g_Ki2)+Fru6P_g*ATP_g/(PFK_g_KmFru6P*PFK_g_KmATP)))
_6PGDH_g_KmNADP=0.001; _6PGDH_g_KmNADPH=6.0E-4; _6PGDH_g_Keq=47.0; _6PGDH_g_Km6PG=0.0035; _6PGDH_g_KmRul5P=0.03; _6PGDH_g_Vmax=10.6Reaction: _6PG_g + NADP_g => Rul5P_g + CO2_g + NADPH_g; _6PG_g, NADP_g, Rul5P_g, NADPH_g, Rate Law: _6PGDH_g_Vmax*_6PG_g*NADP_g*(1-Rul5P_g*NADPH_g/(_6PGDH_g_Keq*_6PG_g*NADP_g))/(_6PGDH_g_Km6PG*_6PGDH_g_KmNADP*(1+_6PG_g/_6PGDH_g_Km6PG+Rul5P_g/_6PGDH_g_KmRul5P)*(1+NADP_g/_6PGDH_g_KmNADP+NADPH_g/_6PGDH_g_KmNADPH))
GlcT_c_Vmax=111.7; GlcT_c_KmGlc=1.0; GlcT_c_alpha=0.75Reaction: Glc_e => Glc_c; Glc_e, Glc_c, Rate Law: GlcT_c_Vmax*(Glc_e-Glc_c)/(GlcT_c_KmGlc+Glc_e+Glc_c+GlcT_c_alpha*Glc_e*Glc_c/GlcT_c_KmGlc)
TPI_g_Keq=0.046; TPI_g_KmDHAP=1.2; TPI_g_KmGA3P=0.25; TPI_g_Vmax=999.3Reaction: DHAP_g => GA3P_g; DHAP_g, GA3P_g, Rate Law: TPI_g_Vmax*DHAP_g*(1-GA3P_g/(TPI_g_Keq*DHAP_g))/(TPI_g_KmDHAP*(1+DHAP_g/TPI_g_KmDHAP+GA3P_g/TPI_g_KmGA3P))
NADPHu_g_k=2.0Reaction: NADPH_g => NADP_g; NADPH_g, Rate Law: NADPHu_g_k*NADPH_g
PGAM_c_Km3PGA=0.15; PGAM_c_Vmax=225.0; PGAM_c_Km2PGA=0.16; PGAM_c_Keq=0.17Reaction: _3PGA_c => _2PGA_c; _3PGA_c, _2PGA_c, Rate Law: PGAM_c_Vmax*_3PGA_c*(1-_2PGA_c/(PGAM_c_Keq*_3PGA_c))/(PGAM_c_Km3PGA*(1+_3PGA_c/PGAM_c_Km3PGA+_2PGA_c/PGAM_c_Km2PGA))
G3PDH_g_KmDHAP=0.1; G3PDH_g_KmNAD=0.4; G3PDH_g_Vmax=465.0; G3PDH_g_Keq=17085.0; G3PDH_g_KmNADH=0.01; G3PDH_g_KmGly3P=2.0Reaction: NADH_g + DHAP_g => Gly3P_g + NAD_g; DHAP_g, NADH_g, Gly3P_g, NAD_g, Rate Law: G3PDH_g_Vmax*DHAP_g*NADH_g*(1-Gly3P_g*NAD_g/(G3PDH_g_Keq*DHAP_g*NADH_g))/(G3PDH_g_KmDHAP*G3PDH_g_KmNADH*(1+DHAP_g/G3PDH_g_KmDHAP+Gly3P_g/G3PDH_g_KmGly3P)*(1+NADH_g/G3PDH_g_KmNADH+NAD_g/G3PDH_g_KmNAD))
GK_g_Keq=8.37E-4; GK_g_KmATP=0.24; GK_g_KmGly=0.44; GK_g_KmADP=0.56; GK_g_KmGly3P=3.83; GK_g_Vmax=200.0Reaction: Gly3P_g + ADP_g => Gly_e + ATP_g; Gly3P_g, ADP_g, Gly_e, ATP_g, Rate Law: GK_g_Vmax*Gly3P_g*ADP_g*(1-Gly_e*ATP_g/(GK_g_Keq*Gly3P_g*ADP_g))/(GK_g_KmGly3P*GK_g_KmADP*(1+Gly3P_g/GK_g_KmGly3P+Gly_e/GK_g_KmGly)*(1+ADP_g/GK_g_KmADP+ATP_g/GK_g_KmATP))
GlcT_g_k2=250000.0; GlcT_g_k1=250000.0Reaction: Glc_c => Glc_g; Glc_c, Glc_g, Rate Law: GlcT_g_k1*Glc_c-GlcT_g_k2*Glc_g
PGL_c_Km6PG=0.05; PGL_c_k=0.055; PGL_c_Vmax=28.0; PGL_c_Km6PGL=0.05; PGL_c_Keq=20000.0Reaction: _6PGL_c => _6PG_c; _6PGL_c, _6PG_c, Rate Law: PGL_c_k*cytosol*(_6PGL_c-_6PG_c/PGL_c_Keq)+PGL_c_Vmax*_6PGL_c*(1-_6PG_c/(PGL_c_Keq*_6PGL_c))/(PGL_c_Km6PGL*(1+_6PGL_c/PGL_c_Km6PGL+_6PG_c/PGL_c_Km6PG))
AK_c_k1=480.0; AK_c_k2=1000.0Reaction: ADP_c => AMP_c + ATP_c; ADP_c, AMP_c, ATP_c, Rate Law: AK_c_k1*ADP_c^2-AMP_c*ATP_c*AK_c_k2
_6PGDH_c_KmNADP=0.001; _6PGDH_c_Keq=47.0; _6PGDH_c_Vmax=10.6; _6PGDH_c_KmNADPH=6.0E-4; _6PGDH_c_Km6PG=0.0035; _6PGDH_c_KmRul5P=0.03Reaction: NADP_c + _6PG_c => CO2_c + NADPH_c + Rul5P_c; _6PG_c, NADP_c, Rul5P_c, NADPH_c, Rate Law: _6PGDH_c_Vmax*_6PG_c*NADP_c*(1-Rul5P_c*NADPH_c/(_6PGDH_c_Keq*_6PG_c*NADP_c))/(_6PGDH_c_Km6PG*_6PGDH_c_KmNADP*(1+_6PG_c/_6PGDH_c_Km6PG+Rul5P_c/_6PGDH_c_KmRul5P)*(1+NADP_c/_6PGDH_c_KmNADP+NADPH_c/_6PGDH_c_KmNADPH))
PPI_g_KmRul5P=1.4; PPI_g_Vmax=72.0; PPI_g_Keq=5.6; PPI_g_KmRib5P=4.0Reaction: Rul5P_g => Rib5P_g; Rul5P_g, Rib5P_g, Rate Law: PPI_g_Vmax*Rul5P_g*(1-Rib5P_g/(PPI_g_Keq*Rul5P_g))/(PPI_g_KmRul5P*(1+Rul5P_g/PPI_g_KmRul5P+Rib5P_g/PPI_g_KmRib5P))
PGK_g_Km13BPGA=0.003; PGK_g_Vmax=2862.0; PGK_g_KmADP=0.1; PGK_g_Km3PGA=1.62; PGK_g_KmATP=0.29; PGK_g_Keq=3377.0Reaction: _13BPGA_g + ADP_g => _3PGA_g + ATP_g; _13BPGA_g, ADP_g, _3PGA_g, ATP_g, Rate Law: PGK_g_Vmax*_13BPGA_g*ADP_g*(1-_3PGA_g*ATP_g/(PGK_g_Keq*_13BPGA_g*ADP_g))/(PGK_g_Km13BPGA*PGK_g_KmADP*(1+_13BPGA_g/PGK_g_Km13BPGA+_3PGA_g/PGK_g_Km3PGA)*(1+ADP_g/PGK_g_KmADP+ATP_g/PGK_g_KmATP))

States:

NameDescription
6PG g[6-phospho-D-gluconic acid]
6PG c[6-phospho-D-gluconic acid]
Rul5P g[D-ribulose 5-phosphate(2-)]
TS2 c[trypanothione]
Glc6P c[D-glucopyranose 6-phosphate]
PEP c[phosphoenolpyruvate]
CO2 g[carbon dioxide]
ATP g[ATP]
GA3P g[glyceraldehyde 3-phosphate]
Glc c[glucose]
Fru16BP g[alpha-D-fructofuranose 1,6-bisphosphate]
Glc g[glucose]
Glc e[glucose]
TSH2 c[trypanothione disulfide]
Pyr e[pyruvate]
ADP g[ADP]
13BPGA g[683]
NADP c[NADP(+)]
DHAP g[glycerone phosphate(2-)]
NAD g[NAD]
NADH g[NADH]
Pyr c[pyruvate]
CO2 c[carbon dioxide]
6PGL g[6-phosphogluconolactonase3.1.1.17]
Fru6P g[444848]
2PGA c[59]
Gly3P c[glycerol 1-phosphate]
Rib5P c[aldehydo-D-ribose 5-phosphate(2-)]
3PGA c[3-phospho-D-glyceric acid]
NADP g[NADP(+)]
Rib g[ribose]
Rib5P g[aldehydo-D-ribose 5-phosphate(2-)]
Pi g[phosphatidylinositol]
Glc6P g[D-glucopyranose 6-phosphate]
ATP c[ATP]
DHAP c[glycerone phosphate(2-)]
NADPH c[NADPH]
Gly e[glycerol]
6PGL c[6-phosphogluconolactonase3.1.1.17]
Gly3P g[glycerol 1-phosphate]
ADP c[ADP]
AMP g[AMP]
Rul5P c[D-ribulose 5-phosphate(2-)]
3PGA g[3-phospho-D-glyceric acid]
AMP c[AMP]
NADPH g[NADPH]

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei - MODEL C in fructose medium (with glucosomal ribokinase): BIOMD0000000515v0.0.1

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei - MODEL C in fructose medium (with glucosomal ribok…

Details

Dynamic models of metabolism can be useful in identifying potential drug targets, especially in unicellular organisms. A model of glycolysis in the causative agent of human African trypanosomiasis, Trypanosoma brucei, has already shown the utility of this approach. Here we add the pentose phosphate pathway (PPP) of T. brucei to the glycolytic model. The PPP is localized to both the cytosol and the glycosome and adding it to the glycolytic model without further adjustments leads to a draining of the essential bound-phosphate moiety within the glycosome. This phosphate "leak" must be resolved for the model to be a reasonable representation of parasite physiology. Two main types of theoretical solution to the problem could be identified: (i) including additional enzymatic reactions in the glycosome, or (ii) adding a mechanism to transfer bound phosphates between cytosol and glycosome. One example of the first type of solution would be the presence of a glycosomal ribokinase to regenerate ATP from ribose 5-phosphate and ADP. Experimental characterization of ribokinase in T. brucei showed that very low enzyme levels are sufficient for parasite survival, indicating that other mechanisms are required in controlling the phosphate leak. Examples of the second type would involve the presence of an ATP:ADP exchanger or recently described permeability pores in the glycosomal membrane, although the current absence of identified genes encoding such molecules impedes experimental testing by genetic manipulation. Confronted with this uncertainty, we present a modeling strategy that identifies robust predictions in the context of incomplete system characterization. We illustrate this strategy by exploring the mechanism underlying the essential function of one of the PPP enzymes, and validate it by confirming the model predictions experimentally. link: http://identifiers.org/pubmed/24339766

Parameters:

NameDescription
RK_g_Keq=0.0036; RK_g_KmRib=0.51; RK_g_Vmax=10.0; RK_g_KmRib5P=0.39; RK_g_KmADP=0.25; RK_g_KmATP=0.24Reaction: ADP_g + Rib5P_g => ATP_g + Rib_g; Rib5P_g, ADP_g, Rib_g, ATP_g, Rate Law: RK_g_Vmax*Rib5P_g*ADP_g*(1-Rib_g*ATP_g/(RK_g_Keq*Rib5P_g*ADP_g))/(RK_g_KmRib5P*RK_g_KmADP*(1+Rib5P_g/RK_g_KmRib5P+Rib_g/RK_g_KmRib)*(1+ADP_g/RK_g_KmADP+ATP_g/RK_g_KmATP))
_3PGAT_g_k=250.0Reaction: _3PGA_g => _3PGA_c; _3PGA_g, _3PGA_c, Rate Law: _3PGAT_g_k*_3PGA_g-_3PGAT_g_k*_3PGA_c
GDA_g_k=600.0Reaction: Gly3P_g + DHAP_c => Gly3P_c + DHAP_g; Gly3P_g, DHAP_c, Gly3P_c, DHAP_g, Rate Law: Gly3P_g*GDA_g_k*DHAP_c-Gly3P_c*GDA_g_k*DHAP_g
GPO_c_Vmax=368.0; GPO_c_KmGly3P=1.7Reaction: Gly3P_c => DHAP_c; Gly3P_c, Rate Law: GPO_c_Vmax*Gly3P_c/(GPO_c_KmGly3P*(1+Gly3P_c/GPO_c_KmGly3P))
PPI_c_Keq=5.6; PPI_c_Vmax=72.0; PPI_c_KmRul5P=1.4; PPI_c_KmRib5P=4.0Reaction: Rul5P_c => Rib5P_c; Rul5P_c, Rib5P_c, Rate Law: PPI_c_Vmax*Rul5P_c*(1-Rib5P_c/(PPI_c_Keq*Rul5P_c))/(PPI_c_KmRul5P*(1+Rul5P_c/PPI_c_KmRul5P+Rib5P_c/PPI_c_KmRib5P))
PGI_g_Vmax=1305.0; PGI_g_Ki6PG=0.14; PGI_g_KmGlc6P=0.4; PGI_g_Keq=0.457; PGI_g_KmFru6P=0.12Reaction: Glc6P_g => Fru6P_g; _6PG_g, Glc6P_g, Fru6P_g, _6PG_g, Rate Law: PGI_g_Vmax*Glc6P_g*(1-Fru6P_g/(PGI_g_Keq*Glc6P_g))/(PGI_g_KmGlc6P*(1+Glc6P_g/PGI_g_KmGlc6P+Fru6P_g/PGI_g_KmFru6P+_6PG_g/PGI_g_Ki6PG))
ALD_g_KmDHAP=0.015; ALD_g_KiGA3P=0.098; ALD_g_KmGA3P=0.067; ALD_g_Vmax=560.0; ALD_g_KmFru16BP=0.009; ALD_g_KiADP=1.51; ALD_g_KiAMP=3.65; ALD_g_Keq=0.084; ALD_g_KiATP=0.68Reaction: Fru16BP_g => GA3P_g + DHAP_g; ATP_g, ADP_g, AMP_g, Fru16BP_g, GA3P_g, DHAP_g, ATP_g, ADP_g, AMP_g, Rate Law: ALD_g_Vmax*Fru16BP_g*(1-GA3P_g*DHAP_g/(Fru16BP_g*ALD_g_Keq))/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP)*(1+GA3P_g/ALD_g_KmGA3P+DHAP_g/ALD_g_KmDHAP+Fru16BP_g/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))+GA3P_g*DHAP_g/(ALD_g_KmGA3P*ALD_g_KmDHAP)+Fru16BP_g*GA3P_g/(ALD_g_KmFru16BP*ALD_g_KiGA3P*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))))
ATPu_c_k=50.0Reaction: ATP_c => ADP_c; ATP_c, ADP_c, Rate Law: ATPu_c_k*ATP_c/ADP_c
NADPHu_c_k=2.0Reaction: NADPH_c => NADP_c; NADPH_c, Rate Law: NADPHu_c_k*NADPH_c
PYK_c_KmPyr=50.0; PYK_c_KiADP=0.64; PYK_c_Vmax=1020.0; PYK_c_KmADP=0.114; PYK_c_Keq=10800.0; PYK_c_KiATP=0.57; PYK_c_KmATP=15.0; PYK_c_KmPEP=0.34; PYK_c_n=2.5Reaction: PEP_c + ADP_c => Pyr_c + ATP_c; ADP_c, Pyr_c, ATP_c, PEP_c, Rate Law: PYK_c_Vmax*ADP_c*(1-Pyr_c*ATP_c/(PYK_c_Keq*PEP_c*ADP_c))*(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n/(PYK_c_KmADP*(1+(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n+Pyr_c/PYK_c_KmPyr)*(1+ADP_c/PYK_c_KmADP+ATP_c/PYK_c_KmATP))
G6PP_c_KmGlc6P=5.6; G6PP_c_Vmax=28.0; G6PP_c_Keq=263.0; G6PP_c_KmGlc=5.6Reaction: Glc6P_c => Glc_c; Glc6P_c, Glc_c, Rate Law: G6PP_c_Vmax*Glc6P_c*(1-Glc_c/(G6PP_c_Keq*Glc6P_c))/(G6PP_c_KmGlc6P*(1+Glc6P_c/G6PP_c_KmGlc6P+Glc_c/G6PP_c_KmGlc))
G6PDH_g_KmNADPH=1.0E-4; G6PDH_g_Keq=5.02; G6PDH_g_KmNADP=0.0094; G6PDH_g_KmGlc6P=0.058; G6PDH_g_Vmax=8.4; G6PDH_g_Km6PGL=0.04Reaction: Glc6P_g + NADP_g => _6PGL_g + NADPH_g; Glc6P_g, NADP_g, _6PGL_g, NADPH_g, Rate Law: G6PDH_g_Vmax*Glc6P_g*NADP_g*(1-_6PGL_g*NADPH_g/(G6PDH_g_Keq*Glc6P_g*NADP_g))/(G6PDH_g_KmGlc6P*G6PDH_g_KmNADP*(1+Glc6P_g/G6PDH_g_KmGlc6P+_6PGL_g/G6PDH_g_Km6PGL)*(1+NADP_g/G6PDH_g_KmNADP+NADPH_g/G6PDH_g_KmNADPH))
TR_c_KmTS2=0.0069; TR_c_KmTSH2=0.0018; TR_c_KmNADPH=7.7E-4; TR_c_Vmax=252.0; TR_c_Keq=434.0; TR_c_KmNADP=0.081Reaction: TS2_c + NADPH_c => NADP_c + TSH2_c; TS2_c, NADPH_c, TSH2_c, NADP_c, Rate Law: TR_c_Vmax*TS2_c*NADPH_c*(1-TSH2_c*NADP_c/(TR_c_Keq*TS2_c*NADPH_c))/(TR_c_KmTS2*TR_c_KmNADPH*(1+TS2_c/TR_c_KmTS2+TSH2_c/TR_c_KmTSH2)*(1+NADPH_c/TR_c_KmNADPH+NADP_c/TR_c_KmNADP))
AK_g_k2=1000.0; AK_g_k1=480.0Reaction: ADP_g => AMP_g + ATP_g; ADP_g, AMP_g, ATP_g, Rate Law: AK_g_k1*ADP_g^2-AMP_g*ATP_g*AK_g_k2
HXK_g_Vmax=1774.68; HXK_g_KiFru6P=2.7; HXK_g_KiFru=0.35; HXK_g_KmADP=0.126; HXK_g_Keq=759.0; HXK_g_KmATP=0.116; HXK_g_KmGlc6P=2.7; HXK_g_KmGlc=0.1Reaction: ATP_g + Glc_g => Glc6P_g + ADP_g; Fru_g, Fru6P_g, Glc_g, ATP_g, Glc6P_g, ADP_g, Fru_g, Fru6P_g, Rate Law: HXK_g_Vmax*Glc_g*ATP_g*(1-Glc6P_g*ADP_g/(HXK_g_Keq*Glc_g*ATP_g))/(HXK_g_KmGlc*HXK_g_KmATP*(1+Glc_g/HXK_g_KmGlc+Glc6P_g/HXK_g_KmGlc6P)*(1+ATP_g/HXK_g_KmATP+ADP_g/HXK_g_KmADP+Fru_g/HXK_g_KiFru+Fru6P_g/HXK_g_KiFru6P))
G6PDH_c_Vmax=8.4; G6PDH_c_Keq=5.02; G6PDH_c_KmNADP=0.0094; G6PDH_c_KmNADPH=1.0E-4; G6PDH_c_Km6PGL=0.04; G6PDH_c_KmGlc6P=0.058Reaction: Glc6P_c + NADP_c => NADPH_c + _6PGL_c; Glc6P_c, NADP_c, _6PGL_c, NADPH_c, Rate Law: G6PDH_c_Vmax*Glc6P_c*NADP_c*(1-_6PGL_c*NADPH_c/(G6PDH_c_Keq*Glc6P_c*NADP_c))/(G6PDH_c_KmGlc6P*G6PDH_c_KmNADP*(1+Glc6P_c/G6PDH_c_KmGlc6P+_6PGL_c/G6PDH_c_Km6PGL)*(1+NADP_c/G6PDH_c_KmNADP+NADPH_c/G6PDH_c_KmNADPH))
FruT_c_Vmax=69.1; FruT_c_alpha=0.75; FruT_c_KmFru=3.91Reaction: Fru_e => Fru_c; Fru_e, Fru_c, Rate Law: FruT_c_Vmax*(Fru_e-Fru_c)/(FruT_c_KmFru+Fru_e+Fru_c+FruT_c_alpha*Fru_e*Fru_c/FruT_c_KmFru)
GAPDH_g_Vmax=720.9; GAPDH_g_Km13BPGA=0.1; GAPDH_g_KmNAD=0.45; GAPDH_g_KmNADH=0.02; GAPDH_g_KmGA3P=0.15; GAPDH_g_Keq=0.066Reaction: GA3P_g + NAD_g + Pi_g => NADH_g + _13BPGA_g; GA3P_g, NAD_g, _13BPGA_g, NADH_g, Rate Law: GAPDH_g_Vmax*GA3P_g*NAD_g*(1-_13BPGA_g*NADH_g/(GAPDH_g_Keq*GA3P_g*NAD_g))/(GAPDH_g_KmGA3P*GAPDH_g_KmNAD*(1+GA3P_g/GAPDH_g_KmGA3P+_13BPGA_g/GAPDH_g_Km13BPGA)*(1+NAD_g/GAPDH_g_KmNAD+NADH_g/GAPDH_g_KmNADH))
TOX_c_k=2.0Reaction: TSH2_c => TS2_c; TSH2_c, Rate Law: TOX_c_k*TSH2_c
PGL_g_Km6PGL=0.05; PGL_g_Km6PG=0.05; PGL_g_Vmax=5.0; PGL_g_Keq=20000.0; PGL_g_k=0.055Reaction: _6PGL_g => _6PG_g; _6PGL_g, _6PG_g, Rate Law: glycosome*PGL_g_k*(_6PGL_g-_6PG_g/PGL_g_Keq)+PGL_g_Vmax*_6PGL_g*(1-_6PG_g/(PGL_g_Keq*_6PGL_g))/(PGL_g_Km6PGL*(1+_6PGL_g/PGL_g_Km6PGL+_6PG_g/PGL_g_Km6PG))
ENO_c_Vmax=598.0; ENO_c_Km2PGA=0.054; ENO_c_Keq=4.17; ENO_c_KmPEP=0.24Reaction: _2PGA_c => PEP_c; _2PGA_c, PEP_c, Rate Law: ENO_c_Vmax*_2PGA_c*(1-PEP_c/(ENO_c_Keq*_2PGA_c))/(ENO_c_Km2PGA*(1+_2PGA_c/ENO_c_Km2PGA+PEP_c/ENO_c_KmPEP))
PFK_g_Vmax=1708.0; PFK_g_KsATP=0.0393; PFK_g_KmFru6P=0.999; PFK_g_KmADP=1.0; PFK_g_KmATP=0.0648; PFK_g_Ki2=10.7; PFK_g_Ki1=15.8; PFK_g_Keq=1035.0Reaction: ATP_g + Fru6P_g => Fru16BP_g + ADP_g; Fru6P_g, ATP_g, Fru16BP_g, ADP_g, Rate Law: PFK_g_Vmax*PFK_g_Ki1*Fru6P_g*ATP_g*(1-Fru16BP_g*ADP_g/(PFK_g_Keq*Fru6P_g*ATP_g))/(PFK_g_KmFru6P*PFK_g_KmATP*(Fru16BP_g+PFK_g_Ki1)*(PFK_g_KsATP/PFK_g_KmATP+Fru6P_g/PFK_g_KmFru6P+ATP_g/PFK_g_KmATP+ADP_g/PFK_g_KmADP+Fru16BP_g*ADP_g/(PFK_g_KmADP*PFK_g_Ki2)+Fru6P_g*ATP_g/(PFK_g_KmFru6P*PFK_g_KmATP)))
_6PGDH_g_KmNADP=0.001; _6PGDH_g_KmNADPH=6.0E-4; _6PGDH_g_Keq=47.0; _6PGDH_g_Km6PG=0.0035; _6PGDH_g_KmRul5P=0.03; _6PGDH_g_Vmax=10.6Reaction: _6PG_g + NADP_g => Rul5P_g + CO2_g + NADPH_g; _6PG_g, NADP_g, Rul5P_g, NADPH_g, Rate Law: _6PGDH_g_Vmax*_6PG_g*NADP_g*(1-Rul5P_g*NADPH_g/(_6PGDH_g_Keq*_6PG_g*NADP_g))/(_6PGDH_g_Km6PG*_6PGDH_g_KmNADP*(1+_6PG_g/_6PGDH_g_Km6PG+Rul5P_g/_6PGDH_g_KmRul5P)*(1+NADP_g/_6PGDH_g_KmNADP+NADPH_g/_6PGDH_g_KmNADPH))
TPI_g_Keq=0.046; TPI_g_KmDHAP=1.2; TPI_g_KmGA3P=0.25; TPI_g_Vmax=999.3Reaction: DHAP_g => GA3P_g; DHAP_g, GA3P_g, Rate Law: TPI_g_Vmax*DHAP_g*(1-GA3P_g/(TPI_g_Keq*DHAP_g))/(TPI_g_KmDHAP*(1+DHAP_g/TPI_g_KmDHAP+GA3P_g/TPI_g_KmGA3P))
FruT_g_k1=250000.0; FruT_g_k2=250000.0Reaction: Fru_c => Fru_g; Fru_c, Fru_g, Rate Law: FruT_g_k1*Fru_c-FruT_g_k2*Fru_g
GlcT_c_Vmax=111.7; GlcT_c_KmGlc=1.0; GlcT_c_alpha=0.75Reaction: Glc_e => Glc_c; Glc_e, Glc_c, Rate Law: GlcT_c_Vmax*(Glc_e-Glc_c)/(GlcT_c_KmGlc+Glc_e+Glc_c+GlcT_c_alpha*Glc_e*Glc_c/GlcT_c_KmGlc)
NADPHu_g_k=2.0Reaction: NADPH_g => NADP_g; NADPH_g, Rate Law: NADPHu_g_k*NADPH_g
PGAM_c_Km3PGA=0.15; PGAM_c_Vmax=225.0; PGAM_c_Km2PGA=0.16; PGAM_c_Keq=0.17Reaction: _3PGA_c => _2PGA_c; _3PGA_c, _2PGA_c, Rate Law: PGAM_c_Vmax*_3PGA_c*(1-_2PGA_c/(PGAM_c_Keq*_3PGA_c))/(PGAM_c_Km3PGA*(1+_3PGA_c/PGAM_c_Km3PGA+_2PGA_c/PGAM_c_Km2PGA))
G3PDH_g_KmDHAP=0.1; G3PDH_g_KmNAD=0.4; G3PDH_g_Vmax=465.0; G3PDH_g_Keq=17085.0; G3PDH_g_KmNADH=0.01; G3PDH_g_KmGly3P=2.0Reaction: NADH_g + DHAP_g => Gly3P_g + NAD_g; DHAP_g, NADH_g, Gly3P_g, NAD_g, Rate Law: G3PDH_g_Vmax*DHAP_g*NADH_g*(1-Gly3P_g*NAD_g/(G3PDH_g_Keq*DHAP_g*NADH_g))/(G3PDH_g_KmDHAP*G3PDH_g_KmNADH*(1+DHAP_g/G3PDH_g_KmDHAP+Gly3P_g/G3PDH_g_KmGly3P)*(1+NADH_g/G3PDH_g_KmNADH+NAD_g/G3PDH_g_KmNAD))
GK_g_Keq=8.37E-4; GK_g_KmATP=0.24; GK_g_KmGly=0.44; GK_g_KmADP=0.56; GK_g_KmGly3P=3.83; GK_g_Vmax=200.0Reaction: Gly3P_g + ADP_g => Gly_e + ATP_g; Gly3P_g, ADP_g, Gly_e, ATP_g, Rate Law: GK_g_Vmax*Gly3P_g*ADP_g*(1-Gly_e*ATP_g/(GK_g_Keq*Gly3P_g*ADP_g))/(GK_g_KmGly3P*GK_g_KmADP*(1+Gly3P_g/GK_g_KmGly3P+Gly_e/GK_g_KmGly)*(1+ADP_g/GK_g_KmADP+ATP_g/GK_g_KmATP))
GlcT_g_k2=250000.0; GlcT_g_k1=250000.0Reaction: Glc_c => Glc_g; Glc_c, Glc_g, Rate Law: GlcT_g_k1*Glc_c-GlcT_g_k2*Glc_g
PGL_c_Km6PG=0.05; PGL_c_k=0.055; PGL_c_Vmax=28.0; PGL_c_Km6PGL=0.05; PGL_c_Keq=20000.0Reaction: _6PGL_c => _6PG_c; _6PGL_c, _6PG_c, Rate Law: PGL_c_k*cytosol*(_6PGL_c-_6PG_c/PGL_c_Keq)+PGL_c_Vmax*_6PGL_c*(1-_6PG_c/(PGL_c_Keq*_6PGL_c))/(PGL_c_Km6PGL*(1+_6PGL_c/PGL_c_Km6PGL+_6PG_c/PGL_c_Km6PG))
HXK_c_KmATP=0.116; HXK_c_KmGlc=0.1; HXK_c_KiFru6P=2.7; HXK_c_Vmax=154.32; HXK_c_KmADP=0.126; HXK_c_KiFru=0.35; HXK_c_Keq=759.0; HXK_c_KmGlc6P=2.7Reaction: Glc_c + ATP_c => Glc6P_c + ADP_c; Fru_c, Fru6P_c, Glc_c, ATP_c, Glc6P_c, ADP_c, Fru_c, Fru6P_c, Rate Law: HXK_c_Vmax*Glc_c*ATP_c*(1-Glc6P_c*ADP_c/(HXK_c_Keq*Glc_c*ATP_c))/(HXK_c_KmGlc*HXK_c_KmATP*(1+Glc_c/HXK_c_KmGlc+Glc6P_c/HXK_c_KmGlc6P)*(1+ATP_c/HXK_c_KmATP+ADP_c/HXK_c_KmADP+Fru_c/HXK_c_KiFru+Fru6P_c/HXK_c_KiFru6P))
_6PGDH_c_KmNADP=0.001; _6PGDH_c_Keq=47.0; _6PGDH_c_Vmax=10.6; _6PGDH_c_KmNADPH=6.0E-4; _6PGDH_c_Km6PG=0.0035; _6PGDH_c_KmRul5P=0.03Reaction: NADP_c + _6PG_c => CO2_c + NADPH_c + Rul5P_c; _6PG_c, NADP_c, Rul5P_c, NADPH_c, Rate Law: _6PGDH_c_Vmax*_6PG_c*NADP_c*(1-Rul5P_c*NADPH_c/(_6PGDH_c_Keq*_6PG_c*NADP_c))/(_6PGDH_c_Km6PG*_6PGDH_c_KmNADP*(1+_6PG_c/_6PGDH_c_Km6PG+Rul5P_c/_6PGDH_c_KmRul5P)*(1+NADP_c/_6PGDH_c_KmNADP+NADPH_c/_6PGDH_c_KmNADPH))
AK_c_k1=480.0; AK_c_k2=1000.0Reaction: ADP_c => AMP_c + ATP_c; ADP_c, AMP_c, ATP_c, Rate Law: AK_c_k1*ADP_c^2-AMP_c*ATP_c*AK_c_k2
PPI_g_KmRul5P=1.4; PPI_g_Vmax=72.0; PPI_g_Keq=5.6; PPI_g_KmRib5P=4.0Reaction: Rul5P_g => Rib5P_g; Rul5P_g, Rib5P_g, Rate Law: PPI_g_Vmax*Rul5P_g*(1-Rib5P_g/(PPI_g_Keq*Rul5P_g))/(PPI_g_KmRul5P*(1+Rul5P_g/PPI_g_KmRul5P+Rib5P_g/PPI_g_KmRib5P))
HXKfru_c_KmFru=0.35; HXKfru_c_KiGlc6P=2.7; HXKfru_c_KmADP=0.126; HXKfru_c_Keq=631.0; HXKfru_c_KmATP=0.116; HXKfru_c_KmFru6P=2.7; HXKfru_c_Vmax=154.32; HXKfru_c_KiGlc=0.1Reaction: Fru_c + ATP_c => ADP_c + Fru6P_c; Glc_c, Glc6P_c, Fru_c, ATP_c, Fru6P_c, ADP_c, Glc_c, Glc6P_c, Rate Law: HXKfru_c_Vmax*Fru_c*ATP_c*(1-Fru6P_c*ADP_c/(HXKfru_c_Keq*Fru_c*ATP_c))/(HXKfru_c_KmFru*HXKfru_c_KmATP*(1+Fru_c/HXKfru_c_KmFru+Fru6P_c/HXKfru_c_KmFru6P)*(1+ATP_c/HXKfru_c_KmATP+ADP_c/HXKfru_c_KmADP+Glc_c/HXKfru_c_KiGlc+Glc6P_c/HXKfru_c_KiGlc6P))
PGK_g_Km13BPGA=0.003; PGK_g_Vmax=2862.0; PGK_g_KmADP=0.1; PGK_g_Km3PGA=1.62; PGK_g_KmATP=0.29; PGK_g_Keq=3377.0Reaction: _13BPGA_g + ADP_g => _3PGA_g + ATP_g; _13BPGA_g, ADP_g, _3PGA_g, ATP_g, Rate Law: PGK_g_Vmax*_13BPGA_g*ADP_g*(1-_3PGA_g*ATP_g/(PGK_g_Keq*_13BPGA_g*ADP_g))/(PGK_g_Km13BPGA*PGK_g_KmADP*(1+_13BPGA_g/PGK_g_Km13BPGA+_3PGA_g/PGK_g_Km3PGA)*(1+ADP_g/PGK_g_KmADP+ATP_g/PGK_g_KmATP))
HXKfru_g_Keq=631.0; HXKfru_g_KmATP=0.116; HXKfru_g_KmFru6P=2.7; HXKfru_g_KiGlc6P=2.7; HXKfru_g_KmADP=0.126; HXKfru_g_KiGlc=0.1; HXKfru_g_Vmax=1774.68; HXKfru_g_KmFru=0.35Reaction: Fru_g + ATP_g => ADP_g + Fru6P_g; Glc_g, Glc6P_g, Fru_g, ATP_g, Fru6P_g, ADP_g, Glc_g, Glc6P_g, Rate Law: HXKfru_g_Vmax*Fru_g*ATP_g*(1-Fru6P_g*ADP_g/(HXKfru_g_Keq*Fru_g*ATP_g))/(HXKfru_g_KmFru*HXKfru_g_KmATP*(1+Fru_g/HXKfru_g_KmFru+Fru6P_g/HXKfru_g_KmFru6P)*(1+ATP_g/HXKfru_g_KmATP+ADP_g/HXKfru_g_KmADP+Glc_g/HXKfru_g_KiGlc+Glc6P_g/HXKfru_g_KiGlc6P))

States:

NameDescription
6PG g[6-phospho-D-gluconic acid]
6PG c[6-phospho-D-gluconic acid]
TS2 c[trypanothione]
PEP c[phosphoenolpyruvate]
Glc6P c[D-glucopyranose 6-phosphate]
Rul5P g[D-ribulose 5-phosphate(2-)]
ATP g[ATP]
CO2 g[carbon dioxide]
Glc c[glucose]
Fru6P c[444848]
Fru g[fructose]
Glc g[glucose]
Glc e[glucose]
TSH2 c[trypanothione disulfide]
ADP g[ADP]
13BPGA g[683]
NADP c[NADP(+)]
Fru c[fructose]
DHAP g[glycerone phosphate(2-)]
NADH g[NADH]
Fru e[fructose]
CO2 c[carbon dioxide]
6PGL g[6-phosphogluconolactonase3.1.1.17]
Fru6P g[444848]
2PGA c[59]
Gly3P c[glycerol 1-phosphate]
Rib5P c[aldehydo-D-ribose 5-phosphate(2-)]
3PGA c[3-phospho-D-glyceric acid]
NADP g[NADP(+)]
Rib g[aromatic annulene]
Rib5P g[ribose]
Pi g[phosphatidylinositol]
Glc6P g[D-glucopyranose 6-phosphate]
ATP c[ATP]
DHAP c[glycerone phosphate(2-)]
NADPH c[NADPH]
Gly3P g[glycerol 1-phosphate]
ADP c[ADP]
AMP g[AMP]
Rul5P c[D-ribulose 5-phosphate(2-)]
3PGA g[3-phospho-D-glyceric acid]
AMP c[AMP]
NADPH g[NADPH]

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei - MODEL D (with ATP:ADP antiporter): BIOMD0000000511v0.0.1

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei - MODEL D (with ATP:ADP antiporter)There are six mo…

Details

Dynamic models of metabolism can be useful in identifying potential drug targets, especially in unicellular organisms. A model of glycolysis in the causative agent of human African trypanosomiasis, Trypanosoma brucei, has already shown the utility of this approach. Here we add the pentose phosphate pathway (PPP) of T. brucei to the glycolytic model. The PPP is localized to both the cytosol and the glycosome and adding it to the glycolytic model without further adjustments leads to a draining of the essential bound-phosphate moiety within the glycosome. This phosphate "leak" must be resolved for the model to be a reasonable representation of parasite physiology. Two main types of theoretical solution to the problem could be identified: (i) including additional enzymatic reactions in the glycosome, or (ii) adding a mechanism to transfer bound phosphates between cytosol and glycosome. One example of the first type of solution would be the presence of a glycosomal ribokinase to regenerate ATP from ribose 5-phosphate and ADP. Experimental characterization of ribokinase in T. brucei showed that very low enzyme levels are sufficient for parasite survival, indicating that other mechanisms are required in controlling the phosphate leak. Examples of the second type would involve the presence of an ATP:ADP exchanger or recently described permeability pores in the glycosomal membrane, although the current absence of identified genes encoding such molecules impedes experimental testing by genetic manipulation. Confronted with this uncertainty, we present a modeling strategy that identifies robust predictions in the context of incomplete system characterization. We illustrate this strategy by exploring the mechanism underlying the essential function of one of the PPP enzymes, and validate it by confirming the model predictions experimentally. link: http://identifiers.org/pubmed/24339766

Parameters:

NameDescription
ATPT_g_KmATP=0.02; ATPT_g_Keq=1.0; ATPT_g_Vmax=1.5; ATPT_g_KmADP=0.02Reaction: ADP_g + ATP_c => ATP_g + ADP_c; ADP_g, ATP_c, ADP_c, ATP_g, Rate Law: ATPT_g_Vmax*ADP_g*ATP_c*(1-ADP_c*ATP_g/(ATPT_g_Keq*ADP_g*ATP_c))/(ATPT_g_KmADP*ATPT_g_KmATP*(1+ADP_g/ATPT_g_KmADP+ADP_c/ATPT_g_KmADP)*(1+ATP_c/ATPT_g_KmATP+ATP_g/ATPT_g_KmATP))
PPI_c_Keq=5.6; PPI_c_Vmax=72.0; PPI_c_KmRul5P=1.4; PPI_c_KmRib5P=4.0Reaction: Rul5P_c => Rib5P_c; Rul5P_c, Rib5P_c, Rate Law: PPI_c_Vmax*Rul5P_c*(1-Rib5P_c/(PPI_c_Keq*Rul5P_c))/(PPI_c_KmRul5P*(1+Rul5P_c/PPI_c_KmRul5P+Rib5P_c/PPI_c_KmRib5P))
GDA_g_k=600.0Reaction: Gly3P_g + DHAP_c => Gly3P_c + DHAP_g; Gly3P_g, DHAP_c, Gly3P_c, DHAP_g, Rate Law: Gly3P_g*GDA_g_k*DHAP_c-Gly3P_c*GDA_g_k*DHAP_g
GPO_c_Vmax=368.0; GPO_c_KmGly3P=1.7Reaction: Gly3P_c => DHAP_c; Gly3P_c, Rate Law: GPO_c_Vmax*Gly3P_c/(GPO_c_KmGly3P*(1+Gly3P_c/GPO_c_KmGly3P))
_3PGAT_g_k=250.0Reaction: _3PGA_g => _3PGA_c; _3PGA_g, _3PGA_c, Rate Law: _3PGAT_g_k*_3PGA_g-_3PGAT_g_k*_3PGA_c
PGI_g_Vmax=1305.0; PGI_g_Ki6PG=0.14; PGI_g_KmGlc6P=0.4; PGI_g_Keq=0.457; PGI_g_KmFru6P=0.12Reaction: Glc6P_g => Fru6P_g; _6PG_g, Glc6P_g, Fru6P_g, _6PG_g, Rate Law: PGI_g_Vmax*Glc6P_g*(1-Fru6P_g/(PGI_g_Keq*Glc6P_g))/(PGI_g_KmGlc6P*(1+Glc6P_g/PGI_g_KmGlc6P+Fru6P_g/PGI_g_KmFru6P+_6PG_g/PGI_g_Ki6PG))
HXK_g_Vmax=1774.68; HXK_g_KmADP=0.126; HXK_g_Keq=759.0; HXK_g_KmATP=0.116; HXK_g_KmGlc6P=2.7; HXK_g_KmGlc=0.1Reaction: ATP_g + Glc_g => Glc6P_g + ADP_g; Glc_g, ATP_g, Glc6P_g, ADP_g, Rate Law: HXK_g_Vmax*Glc_g*ATP_g*(1-Glc6P_g*ADP_g/(HXK_g_Keq*Glc_g*ATP_g))/(HXK_g_KmGlc*HXK_g_KmATP*(1+Glc_g/HXK_g_KmGlc+Glc6P_g/HXK_g_KmGlc6P)*(1+ATP_g/HXK_g_KmATP+ADP_g/HXK_g_KmADP))
NADPHu_c_k=2.0Reaction: NADPH_c => NADP_c; NADPH_c, Rate Law: NADPHu_c_k*NADPH_c
ATPu_c_k=50.0Reaction: ATP_c => ADP_c; ATP_c, ADP_c, Rate Law: ATPu_c_k*ATP_c/ADP_c
G6PP_c_KmGlc6P=5.6; G6PP_c_Vmax=28.0; G6PP_c_Keq=263.0; G6PP_c_KmGlc=5.6Reaction: Glc6P_c => Glc_c; Glc6P_c, Glc_c, Rate Law: G6PP_c_Vmax*Glc6P_c*(1-Glc_c/(G6PP_c_Keq*Glc6P_c))/(G6PP_c_KmGlc6P*(1+Glc6P_c/G6PP_c_KmGlc6P+Glc_c/G6PP_c_KmGlc))
PYK_c_KmPyr=50.0; PYK_c_KiADP=0.64; PYK_c_Vmax=1020.0; PYK_c_KmADP=0.114; PYK_c_Keq=10800.0; PYK_c_KiATP=0.57; PYK_c_KmATP=15.0; PYK_c_KmPEP=0.34; PYK_c_n=2.5Reaction: PEP_c + ADP_c => Pyr_c + ATP_c; ADP_c, Pyr_c, ATP_c, PEP_c, Rate Law: PYK_c_Vmax*ADP_c*(1-Pyr_c*ATP_c/(PYK_c_Keq*PEP_c*ADP_c))*(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n/(PYK_c_KmADP*(1+(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n+Pyr_c/PYK_c_KmPyr)*(1+ADP_c/PYK_c_KmADP+ATP_c/PYK_c_KmATP))
ALD_g_KmDHAP=0.015; ALD_g_KiGA3P=0.098; ALD_g_KmGA3P=0.067; ALD_g_Vmax=560.0; ALD_g_KmFru16BP=0.009; ALD_g_KiADP=1.51; ALD_g_KiAMP=3.65; ALD_g_Keq=0.084; ALD_g_KiATP=0.68Reaction: Fru16BP_g => GA3P_g + DHAP_g; ATP_g, ADP_g, AMP_g, Fru16BP_g, GA3P_g, DHAP_g, ATP_g, ADP_g, AMP_g, Rate Law: ALD_g_Vmax*Fru16BP_g*(1-GA3P_g*DHAP_g/(Fru16BP_g*ALD_g_Keq))/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP)*(1+GA3P_g/ALD_g_KmGA3P+DHAP_g/ALD_g_KmDHAP+Fru16BP_g/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))+GA3P_g*DHAP_g/(ALD_g_KmGA3P*ALD_g_KmDHAP)+Fru16BP_g*GA3P_g/(ALD_g_KmFru16BP*ALD_g_KiGA3P*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))))
G6PDH_g_KmNADPH=1.0E-4; G6PDH_g_Keq=5.02; G6PDH_g_KmNADP=0.0094; G6PDH_g_KmGlc6P=0.058; G6PDH_g_Vmax=8.4; G6PDH_g_Km6PGL=0.04Reaction: Glc6P_g + NADP_g => _6PGL_g + NADPH_g; Glc6P_g, NADP_g, _6PGL_g, NADPH_g, Rate Law: G6PDH_g_Vmax*Glc6P_g*NADP_g*(1-_6PGL_g*NADPH_g/(G6PDH_g_Keq*Glc6P_g*NADP_g))/(G6PDH_g_KmGlc6P*G6PDH_g_KmNADP*(1+Glc6P_g/G6PDH_g_KmGlc6P+_6PGL_g/G6PDH_g_Km6PGL)*(1+NADP_g/G6PDH_g_KmNADP+NADPH_g/G6PDH_g_KmNADPH))
TR_c_KmTS2=0.0069; TR_c_KmTSH2=0.0018; TR_c_KmNADPH=7.7E-4; TR_c_Vmax=252.0; TR_c_Keq=434.0; TR_c_KmNADP=0.081Reaction: TS2_c + NADPH_c => NADP_c + TSH2_c; TS2_c, NADPH_c, TSH2_c, NADP_c, Rate Law: TR_c_Vmax*TS2_c*NADPH_c*(1-TSH2_c*NADP_c/(TR_c_Keq*TS2_c*NADPH_c))/(TR_c_KmTS2*TR_c_KmNADPH*(1+TS2_c/TR_c_KmTS2+TSH2_c/TR_c_KmTSH2)*(1+NADPH_c/TR_c_KmNADPH+NADP_c/TR_c_KmNADP))
AK_g_k2=1000.0; AK_g_k1=480.0Reaction: ADP_g => AMP_g + ATP_g; ADP_g, AMP_g, ATP_g, Rate Law: AK_g_k1*ADP_g^2-AMP_g*ATP_g*AK_g_k2
PyrT_c_Vmax=230.0; PyrT_c_KmPyr=1.96Reaction: Pyr_c => Pyr_e; Pyr_c, Rate Law: PyrT_c_Vmax*Pyr_c/(PyrT_c_KmPyr*(1+Pyr_c/PyrT_c_KmPyr))
G6PDH_c_Vmax=8.4; G6PDH_c_Keq=5.02; G6PDH_c_KmNADP=0.0094; G6PDH_c_KmNADPH=1.0E-4; G6PDH_c_Km6PGL=0.04; G6PDH_c_KmGlc6P=0.058Reaction: Glc6P_c + NADP_c => NADPH_c + _6PGL_c; Glc6P_c, NADP_c, _6PGL_c, NADPH_c, Rate Law: G6PDH_c_Vmax*Glc6P_c*NADP_c*(1-_6PGL_c*NADPH_c/(G6PDH_c_Keq*Glc6P_c*NADP_c))/(G6PDH_c_KmGlc6P*G6PDH_c_KmNADP*(1+Glc6P_c/G6PDH_c_KmGlc6P+_6PGL_c/G6PDH_c_Km6PGL)*(1+NADP_c/G6PDH_c_KmNADP+NADPH_c/G6PDH_c_KmNADPH))
HXK_c_KmATP=0.116; HXK_c_KmGlc=0.1; HXK_c_Vmax=154.32; HXK_c_KmADP=0.126; HXK_c_Keq=759.0; HXK_c_KmGlc6P=2.7Reaction: Glc_c + ATP_c => Glc6P_c + ADP_c; Glc_c, ATP_c, Glc6P_c, ADP_c, Rate Law: HXK_c_Vmax*Glc_c*ATP_c*(1-Glc6P_c*ADP_c/(HXK_c_Keq*Glc_c*ATP_c))/(HXK_c_KmGlc*HXK_c_KmATP*(1+Glc_c/HXK_c_KmGlc+Glc6P_c/HXK_c_KmGlc6P)*(1+ATP_c/HXK_c_KmATP+ADP_c/HXK_c_KmADP))
GAPDH_g_Vmax=720.9; GAPDH_g_Km13BPGA=0.1; GAPDH_g_KmNAD=0.45; GAPDH_g_KmNADH=0.02; GAPDH_g_KmGA3P=0.15; GAPDH_g_Keq=0.066Reaction: GA3P_g + NAD_g + Pi_g => NADH_g + _13BPGA_g; GA3P_g, NAD_g, _13BPGA_g, NADH_g, Rate Law: GAPDH_g_Vmax*GA3P_g*NAD_g*(1-_13BPGA_g*NADH_g/(GAPDH_g_Keq*GA3P_g*NAD_g))/(GAPDH_g_KmGA3P*GAPDH_g_KmNAD*(1+GA3P_g/GAPDH_g_KmGA3P+_13BPGA_g/GAPDH_g_Km13BPGA)*(1+NAD_g/GAPDH_g_KmNAD+NADH_g/GAPDH_g_KmNADH))
PGL_g_Km6PGL=0.05; PGL_g_Km6PG=0.05; PGL_g_Vmax=5.0; PGL_g_Keq=20000.0; PGL_g_k=0.055Reaction: _6PGL_g => _6PG_g; _6PGL_g, _6PG_g, Rate Law: glycosome*PGL_g_k*(_6PGL_g-_6PG_g/PGL_g_Keq)+PGL_g_Vmax*_6PGL_g*(1-_6PG_g/(PGL_g_Keq*_6PGL_g))/(PGL_g_Km6PGL*(1+_6PGL_g/PGL_g_Km6PGL+_6PG_g/PGL_g_Km6PG))
TOX_c_k=2.0Reaction: TSH2_c => TS2_c; TSH2_c, Rate Law: TOX_c_k*TSH2_c
ENO_c_Vmax=598.0; ENO_c_Km2PGA=0.054; ENO_c_Keq=4.17; ENO_c_KmPEP=0.24Reaction: _2PGA_c => PEP_c; _2PGA_c, PEP_c, Rate Law: ENO_c_Vmax*_2PGA_c*(1-PEP_c/(ENO_c_Keq*_2PGA_c))/(ENO_c_Km2PGA*(1+_2PGA_c/ENO_c_Km2PGA+PEP_c/ENO_c_KmPEP))
PFK_g_Vmax=1708.0; PFK_g_KsATP=0.0393; PFK_g_KmFru6P=0.999; PFK_g_KmADP=1.0; PFK_g_KmATP=0.0648; PFK_g_Ki2=10.7; PFK_g_Ki1=15.8; PFK_g_Keq=1035.0Reaction: ATP_g + Fru6P_g => Fru16BP_g + ADP_g; Fru6P_g, ATP_g, Fru16BP_g, ADP_g, Rate Law: PFK_g_Vmax*PFK_g_Ki1*Fru6P_g*ATP_g*(1-Fru16BP_g*ADP_g/(PFK_g_Keq*Fru6P_g*ATP_g))/(PFK_g_KmFru6P*PFK_g_KmATP*(Fru16BP_g+PFK_g_Ki1)*(PFK_g_KsATP/PFK_g_KmATP+Fru6P_g/PFK_g_KmFru6P+ATP_g/PFK_g_KmATP+ADP_g/PFK_g_KmADP+Fru16BP_g*ADP_g/(PFK_g_KmADP*PFK_g_Ki2)+Fru6P_g*ATP_g/(PFK_g_KmFru6P*PFK_g_KmATP)))
GlcT_c_Vmax=111.7; GlcT_c_KmGlc=1.0; GlcT_c_alpha=0.75Reaction: Glc_e => Glc_c; Glc_e, Glc_c, Rate Law: GlcT_c_Vmax*(Glc_e-Glc_c)/(GlcT_c_KmGlc+Glc_e+Glc_c+GlcT_c_alpha*Glc_e*Glc_c/GlcT_c_KmGlc)
TPI_g_Keq=0.046; TPI_g_KmDHAP=1.2; TPI_g_KmGA3P=0.25; TPI_g_Vmax=999.3Reaction: DHAP_g => GA3P_g; DHAP_g, GA3P_g, Rate Law: TPI_g_Vmax*DHAP_g*(1-GA3P_g/(TPI_g_Keq*DHAP_g))/(TPI_g_KmDHAP*(1+DHAP_g/TPI_g_KmDHAP+GA3P_g/TPI_g_KmGA3P))
_6PGDH_g_KmNADP=0.001; _6PGDH_g_KmNADPH=6.0E-4; _6PGDH_g_Keq=47.0; _6PGDH_g_Km6PG=0.0035; _6PGDH_g_KmRul5P=0.03; _6PGDH_g_Vmax=10.6Reaction: _6PG_g + NADP_g => Rul5P_g + CO2_g + NADPH_g; _6PG_g, NADP_g, Rul5P_g, NADPH_g, Rate Law: _6PGDH_g_Vmax*_6PG_g*NADP_g*(1-Rul5P_g*NADPH_g/(_6PGDH_g_Keq*_6PG_g*NADP_g))/(_6PGDH_g_Km6PG*_6PGDH_g_KmNADP*(1+_6PG_g/_6PGDH_g_Km6PG+Rul5P_g/_6PGDH_g_KmRul5P)*(1+NADP_g/_6PGDH_g_KmNADP+NADPH_g/_6PGDH_g_KmNADPH))
NADPHu_g_k=2.0Reaction: NADPH_g => NADP_g; NADPH_g, Rate Law: NADPHu_g_k*NADPH_g
PGAM_c_Km3PGA=0.15; PGAM_c_Vmax=225.0; PGAM_c_Km2PGA=0.16; PGAM_c_Keq=0.17Reaction: _3PGA_c => _2PGA_c; _3PGA_c, _2PGA_c, Rate Law: PGAM_c_Vmax*_3PGA_c*(1-_2PGA_c/(PGAM_c_Keq*_3PGA_c))/(PGAM_c_Km3PGA*(1+_3PGA_c/PGAM_c_Km3PGA+_2PGA_c/PGAM_c_Km2PGA))
G3PDH_g_KmDHAP=0.1; G3PDH_g_KmNAD=0.4; G3PDH_g_Vmax=465.0; G3PDH_g_Keq=17085.0; G3PDH_g_KmNADH=0.01; G3PDH_g_KmGly3P=2.0Reaction: NADH_g + DHAP_g => Gly3P_g + NAD_g; DHAP_g, NADH_g, Gly3P_g, NAD_g, Rate Law: G3PDH_g_Vmax*DHAP_g*NADH_g*(1-Gly3P_g*NAD_g/(G3PDH_g_Keq*DHAP_g*NADH_g))/(G3PDH_g_KmDHAP*G3PDH_g_KmNADH*(1+DHAP_g/G3PDH_g_KmDHAP+Gly3P_g/G3PDH_g_KmGly3P)*(1+NADH_g/G3PDH_g_KmNADH+NAD_g/G3PDH_g_KmNAD))
GK_g_Keq=8.37E-4; GK_g_KmATP=0.24; GK_g_KmGly=0.44; GK_g_KmADP=0.56; GK_g_KmGly3P=3.83; GK_g_Vmax=200.0Reaction: Gly3P_g + ADP_g => Gly_e + ATP_g; Gly3P_g, ADP_g, Gly_e, ATP_g, Rate Law: GK_g_Vmax*Gly3P_g*ADP_g*(1-Gly_e*ATP_g/(GK_g_Keq*Gly3P_g*ADP_g))/(GK_g_KmGly3P*GK_g_KmADP*(1+Gly3P_g/GK_g_KmGly3P+Gly_e/GK_g_KmGly)*(1+ADP_g/GK_g_KmADP+ATP_g/GK_g_KmATP))
GlcT_g_k2=250000.0; GlcT_g_k1=250000.0Reaction: Glc_c => Glc_g; Glc_c, Glc_g, Rate Law: GlcT_g_k1*Glc_c-GlcT_g_k2*Glc_g
PGL_c_Km6PG=0.05; PGL_c_k=0.055; PGL_c_Vmax=28.0; PGL_c_Km6PGL=0.05; PGL_c_Keq=20000.0Reaction: _6PGL_c => _6PG_c; _6PGL_c, _6PG_c, Rate Law: PGL_c_k*cytosol*(_6PGL_c-_6PG_c/PGL_c_Keq)+PGL_c_Vmax*_6PGL_c*(1-_6PG_c/(PGL_c_Keq*_6PGL_c))/(PGL_c_Km6PGL*(1+_6PGL_c/PGL_c_Km6PGL+_6PG_c/PGL_c_Km6PG))
AK_c_k1=480.0; AK_c_k2=1000.0Reaction: ADP_c => AMP_c + ATP_c; ADP_c, AMP_c, ATP_c, Rate Law: AK_c_k1*ADP_c^2-AMP_c*ATP_c*AK_c_k2
_6PGDH_c_KmNADP=0.001; _6PGDH_c_Keq=47.0; _6PGDH_c_Vmax=10.6; _6PGDH_c_KmNADPH=6.0E-4; _6PGDH_c_Km6PG=0.0035; _6PGDH_c_KmRul5P=0.03Reaction: NADP_c + _6PG_c => CO2_c + NADPH_c + Rul5P_c; _6PG_c, NADP_c, Rul5P_c, NADPH_c, Rate Law: _6PGDH_c_Vmax*_6PG_c*NADP_c*(1-Rul5P_c*NADPH_c/(_6PGDH_c_Keq*_6PG_c*NADP_c))/(_6PGDH_c_Km6PG*_6PGDH_c_KmNADP*(1+_6PG_c/_6PGDH_c_Km6PG+Rul5P_c/_6PGDH_c_KmRul5P)*(1+NADP_c/_6PGDH_c_KmNADP+NADPH_c/_6PGDH_c_KmNADPH))
PPI_g_KmRul5P=1.4; PPI_g_Vmax=72.0; PPI_g_Keq=5.6; PPI_g_KmRib5P=4.0Reaction: Rul5P_g => Rib5P_g; Rul5P_g, Rib5P_g, Rate Law: PPI_g_Vmax*Rul5P_g*(1-Rib5P_g/(PPI_g_Keq*Rul5P_g))/(PPI_g_KmRul5P*(1+Rul5P_g/PPI_g_KmRul5P+Rib5P_g/PPI_g_KmRib5P))
PGK_g_Km13BPGA=0.003; PGK_g_Vmax=2862.0; PGK_g_KmADP=0.1; PGK_g_Km3PGA=1.62; PGK_g_KmATP=0.29; PGK_g_Keq=3377.0Reaction: _13BPGA_g + ADP_g => _3PGA_g + ATP_g; _13BPGA_g, ADP_g, _3PGA_g, ATP_g, Rate Law: PGK_g_Vmax*_13BPGA_g*ADP_g*(1-_3PGA_g*ATP_g/(PGK_g_Keq*_13BPGA_g*ADP_g))/(PGK_g_Km13BPGA*PGK_g_KmADP*(1+_13BPGA_g/PGK_g_Km13BPGA+_3PGA_g/PGK_g_Km3PGA)*(1+ADP_g/PGK_g_KmADP+ATP_g/PGK_g_KmATP))

States:

NameDescription
6PG g[6-phospho-D-gluconic acid]
6PG c[6-phospho-D-gluconic acid]
Rul5P g[D-ribulose 5-phosphate(2-)]
TS2 c[trypanothione]
Glc6P c[D-glucopyranose 6-phosphate]
PEP c[phosphoenolpyruvate]
CO2 g[carbon dioxide]
ATP g[ATP]
Glc c[glucose]
GA3P g[glyceraldehyde 3-phosphate]
Fru16BP g[alpha-D-fructofuranose 1,6-bisphosphate]
Glc g[glucose]
Glc e[glucose]
TSH2 c[trypanothione disulfide]
Pyr e[pyruvate]
ADP g[ADP]
13BPGA g[683]
NADP c[NADP(+)]
DHAP g[CHEBI_57622]
NAD g[NAD]
NADH g[NADH]
Pyr c[pyruvate]
CO2 c[carbon dioxide]
6PGL g[6-phosphogluconolactonase3.1.1.17]
Fru6P g[444848]
2PGA c[59]
Gly3P c[glycerol 1-phosphate]
Rib5P c[aldehydo-D-ribose 5-phosphate(2-)]
NADP g[NADP(+)]
Rib5P g[aldehydo-D-ribose 5-phosphate(2-)]
Pi g[phosphatidylinositol]
Glc6P g[D-glucopyranose 6-phosphate]
ATP c[ATP]
DHAP c[glycerone phosphate(2-)]
NADPH c[NADPH]
Gly e[glycerol]
6PGL c[6-phosphogluconolactonase3.1.1.17]
Gly3P g[glycerol 1-phosphate]
ADP c[ADP]
AMP g[AMP]
Rul5P c[D-ribulose 5-phosphate(2-)]
3PGA g[3-phospho-D-glyceric acid]
AMP c[AMP]
NADPH g[NADPH]

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei - MODEL D in fructose medium (with ATP:ADP antiporter): BIOMD0000000516v0.0.1

Kerkhoven2013 - Glycolysis and Pentose Phosphate Pathway in T.brucei - MODEL D in fructose medium (with ATP:ADP antiport…

Details

Dynamic models of metabolism can be useful in identifying potential drug targets, especially in unicellular organisms. A model of glycolysis in the causative agent of human African trypanosomiasis, Trypanosoma brucei, has already shown the utility of this approach. Here we add the pentose phosphate pathway (PPP) of T. brucei to the glycolytic model. The PPP is localized to both the cytosol and the glycosome and adding it to the glycolytic model without further adjustments leads to a draining of the essential bound-phosphate moiety within the glycosome. This phosphate "leak" must be resolved for the model to be a reasonable representation of parasite physiology. Two main types of theoretical solution to the problem could be identified: (i) including additional enzymatic reactions in the glycosome, or (ii) adding a mechanism to transfer bound phosphates between cytosol and glycosome. One example of the first type of solution would be the presence of a glycosomal ribokinase to regenerate ATP from ribose 5-phosphate and ADP. Experimental characterization of ribokinase in T. brucei showed that very low enzyme levels are sufficient for parasite survival, indicating that other mechanisms are required in controlling the phosphate leak. Examples of the second type would involve the presence of an ATP:ADP exchanger or recently described permeability pores in the glycosomal membrane, although the current absence of identified genes encoding such molecules impedes experimental testing by genetic manipulation. Confronted with this uncertainty, we present a modeling strategy that identifies robust predictions in the context of incomplete system characterization. We illustrate this strategy by exploring the mechanism underlying the essential function of one of the PPP enzymes, and validate it by confirming the model predictions experimentally. link: http://identifiers.org/pubmed/24339766

Parameters:

NameDescription
ATPT_g_KmATP=0.02; ATPT_g_Keq=1.0; ATPT_g_Vmax=1.5; ATPT_g_KmADP=0.02Reaction: ADP_g + ATP_c => ATP_g + ADP_c; ADP_g, ATP_c, ADP_c, ATP_g, Rate Law: ATPT_g_Vmax*ADP_g*ATP_c*(1-ADP_c*ATP_g/(ATPT_g_Keq*ADP_g*ATP_c))/(ATPT_g_KmADP*ATPT_g_KmATP*(1+ADP_g/ATPT_g_KmADP+ADP_c/ATPT_g_KmADP)*(1+ATP_c/ATPT_g_KmATP+ATP_g/ATPT_g_KmATP))
_3PGAT_g_k=250.0Reaction: _3PGA_g => _3PGA_c; _3PGA_g, _3PGA_c, Rate Law: _3PGAT_g_k*_3PGA_g-_3PGAT_g_k*_3PGA_c
PPI_c_Keq=5.6; PPI_c_Vmax=72.0; PPI_c_KmRul5P=1.4; PPI_c_KmRib5P=4.0Reaction: Rul5P_c => Rib5P_c; Rul5P_c, Rib5P_c, Rate Law: PPI_c_Vmax*Rul5P_c*(1-Rib5P_c/(PPI_c_Keq*Rul5P_c))/(PPI_c_KmRul5P*(1+Rul5P_c/PPI_c_KmRul5P+Rib5P_c/PPI_c_KmRib5P))
GDA_g_k=600.0Reaction: Gly3P_g + DHAP_c => Gly3P_c + DHAP_g; Gly3P_g, DHAP_c, Gly3P_c, DHAP_g, Rate Law: Gly3P_g*GDA_g_k*DHAP_c-Gly3P_c*GDA_g_k*DHAP_g
GPO_c_Vmax=368.0; GPO_c_KmGly3P=1.7Reaction: Gly3P_c => DHAP_c; Gly3P_c, Rate Law: GPO_c_Vmax*Gly3P_c/(GPO_c_KmGly3P*(1+Gly3P_c/GPO_c_KmGly3P))
NADPHu_c_k=2.0Reaction: NADPH_c => NADP_c; NADPH_c, Rate Law: NADPHu_c_k*NADPH_c
ATPu_c_k=50.0Reaction: ATP_c => ADP_c; ATP_c, ADP_c, Rate Law: ATPu_c_k*ATP_c/ADP_c
ALD_g_KmDHAP=0.015; ALD_g_KiGA3P=0.098; ALD_g_KmGA3P=0.067; ALD_g_Vmax=560.0; ALD_g_KmFru16BP=0.009; ALD_g_KiADP=1.51; ALD_g_KiAMP=3.65; ALD_g_Keq=0.084; ALD_g_KiATP=0.68Reaction: Fru16BP_g => GA3P_g + DHAP_g; ATP_g, ADP_g, AMP_g, Fru16BP_g, GA3P_g, DHAP_g, ATP_g, ADP_g, AMP_g, Rate Law: ALD_g_Vmax*Fru16BP_g*(1-GA3P_g*DHAP_g/(Fru16BP_g*ALD_g_Keq))/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP)*(1+GA3P_g/ALD_g_KmGA3P+DHAP_g/ALD_g_KmDHAP+Fru16BP_g/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))+GA3P_g*DHAP_g/(ALD_g_KmGA3P*ALD_g_KmDHAP)+Fru16BP_g*GA3P_g/(ALD_g_KmFru16BP*ALD_g_KiGA3P*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))))
PYK_c_KmPyr=50.0; PYK_c_KiADP=0.64; PYK_c_Vmax=1020.0; PYK_c_KmADP=0.114; PYK_c_Keq=10800.0; PYK_c_KiATP=0.57; PYK_c_KmATP=15.0; PYK_c_KmPEP=0.34; PYK_c_n=2.5Reaction: PEP_c + ADP_c => Pyr_c + ATP_c; ADP_c, Pyr_c, ATP_c, PEP_c, Rate Law: PYK_c_Vmax*ADP_c*(1-Pyr_c*ATP_c/(PYK_c_Keq*PEP_c*ADP_c))*(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n/(PYK_c_KmADP*(1+(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n+Pyr_c/PYK_c_KmPyr)*(1+ADP_c/PYK_c_KmADP+ATP_c/PYK_c_KmATP))
G6PP_c_KmGlc6P=5.6; G6PP_c_Vmax=28.0; G6PP_c_Keq=263.0; G6PP_c_KmGlc=5.6Reaction: Glc6P_c => Glc_c; Glc6P_c, Glc_c, Rate Law: G6PP_c_Vmax*Glc6P_c*(1-Glc_c/(G6PP_c_Keq*Glc6P_c))/(G6PP_c_KmGlc6P*(1+Glc6P_c/G6PP_c_KmGlc6P+Glc_c/G6PP_c_KmGlc))
G6PDH_g_KmNADPH=1.0E-4; G6PDH_g_Keq=5.02; G6PDH_g_KmNADP=0.0094; G6PDH_g_KmGlc6P=0.058; G6PDH_g_Vmax=8.4; G6PDH_g_Km6PGL=0.04Reaction: Glc6P_g + NADP_g => _6PGL_g + NADPH_g; Glc6P_g, NADP_g, _6PGL_g, NADPH_g, Rate Law: G6PDH_g_Vmax*Glc6P_g*NADP_g*(1-_6PGL_g*NADPH_g/(G6PDH_g_Keq*Glc6P_g*NADP_g))/(G6PDH_g_KmGlc6P*G6PDH_g_KmNADP*(1+Glc6P_g/G6PDH_g_KmGlc6P+_6PGL_g/G6PDH_g_Km6PGL)*(1+NADP_g/G6PDH_g_KmNADP+NADPH_g/G6PDH_g_KmNADPH))
TR_c_KmTS2=0.0069; TR_c_KmTSH2=0.0018; TR_c_KmNADPH=7.7E-4; TR_c_Vmax=252.0; TR_c_Keq=434.0; TR_c_KmNADP=0.081Reaction: TS2_c + NADPH_c => NADP_c + TSH2_c; TS2_c, NADPH_c, TSH2_c, NADP_c, Rate Law: TR_c_Vmax*TS2_c*NADPH_c*(1-TSH2_c*NADP_c/(TR_c_Keq*TS2_c*NADPH_c))/(TR_c_KmTS2*TR_c_KmNADPH*(1+TS2_c/TR_c_KmTS2+TSH2_c/TR_c_KmTSH2)*(1+NADPH_c/TR_c_KmNADPH+NADP_c/TR_c_KmNADP))
AK_g_k2=1000.0; AK_g_k1=480.0Reaction: ADP_g => AMP_g + ATP_g; ADP_g, AMP_g, ATP_g, Rate Law: AK_g_k1*ADP_g^2-AMP_g*ATP_g*AK_g_k2
PyrT_c_Vmax=230.0; PyrT_c_KmPyr=1.96Reaction: Pyr_c => Pyr_e; Pyr_c, Rate Law: PyrT_c_Vmax*Pyr_c/(PyrT_c_KmPyr*(1+Pyr_c/PyrT_c_KmPyr))
HXK_g_Vmax=1774.68; HXK_g_KiFru6P=2.7; HXK_g_KiFru=0.35; HXK_g_KmADP=0.126; HXK_g_Keq=759.0; HXK_g_KmATP=0.116; HXK_g_KmGlc6P=2.7; HXK_g_KmGlc=0.1Reaction: ATP_g + Glc_g => Glc6P_g + ADP_g; Fru_g, Fru6P_g, Glc_g, ATP_g, Glc6P_g, ADP_g, Fru_g, Fru6P_g, Rate Law: HXK_g_Vmax*Glc_g*ATP_g*(1-Glc6P_g*ADP_g/(HXK_g_Keq*Glc_g*ATP_g))/(HXK_g_KmGlc*HXK_g_KmATP*(1+Glc_g/HXK_g_KmGlc+Glc6P_g/HXK_g_KmGlc6P)*(1+ATP_g/HXK_g_KmATP+ADP_g/HXK_g_KmADP+Fru_g/HXK_g_KiFru+Fru6P_g/HXK_g_KiFru6P))
G6PDH_c_Vmax=8.4; G6PDH_c_Keq=5.02; G6PDH_c_KmNADP=0.0094; G6PDH_c_KmNADPH=1.0E-4; G6PDH_c_Km6PGL=0.04; G6PDH_c_KmGlc6P=0.058Reaction: Glc6P_c + NADP_c => NADPH_c + _6PGL_c; Glc6P_c, NADP_c, _6PGL_c, NADPH_c, Rate Law: G6PDH_c_Vmax*Glc6P_c*NADP_c*(1-_6PGL_c*NADPH_c/(G6PDH_c_Keq*Glc6P_c*NADP_c))/(G6PDH_c_KmGlc6P*G6PDH_c_KmNADP*(1+Glc6P_c/G6PDH_c_KmGlc6P+_6PGL_c/G6PDH_c_Km6PGL)*(1+NADP_c/G6PDH_c_KmNADP+NADPH_c/G6PDH_c_KmNADPH))
GAPDH_g_Vmax=720.9; GAPDH_g_Km13BPGA=0.1; GAPDH_g_KmNAD=0.45; GAPDH_g_KmNADH=0.02; GAPDH_g_KmGA3P=0.15; GAPDH_g_Keq=0.066Reaction: GA3P_g + NAD_g + Pi_g => NADH_g + _13BPGA_g; GA3P_g, NAD_g, _13BPGA_g, NADH_g, Rate Law: GAPDH_g_Vmax*GA3P_g*NAD_g*(1-_13BPGA_g*NADH_g/(GAPDH_g_Keq*GA3P_g*NAD_g))/(GAPDH_g_KmGA3P*GAPDH_g_KmNAD*(1+GA3P_g/GAPDH_g_KmGA3P+_13BPGA_g/GAPDH_g_Km13BPGA)*(1+NAD_g/GAPDH_g_KmNAD+NADH_g/GAPDH_g_KmNADH))
FruT_c_Vmax=69.1; FruT_c_alpha=0.75; FruT_c_KmFru=3.91Reaction: Fru_e => Fru_c; Fru_e, Fru_c, Rate Law: FruT_c_Vmax*(Fru_e-Fru_c)/(FruT_c_KmFru+Fru_e+Fru_c+FruT_c_alpha*Fru_e*Fru_c/FruT_c_KmFru)
PGL_g_Km6PGL=0.05; PGL_g_Km6PG=0.05; PGL_g_Vmax=5.0; PGL_g_Keq=20000.0; PGL_g_k=0.055Reaction: _6PGL_g => _6PG_g; _6PGL_g, _6PG_g, Rate Law: glycosome*PGL_g_k*(_6PGL_g-_6PG_g/PGL_g_Keq)+PGL_g_Vmax*_6PGL_g*(1-_6PG_g/(PGL_g_Keq*_6PGL_g))/(PGL_g_Km6PGL*(1+_6PGL_g/PGL_g_Km6PGL+_6PG_g/PGL_g_Km6PG))
TOX_c_k=2.0Reaction: TSH2_c => TS2_c; TSH2_c, Rate Law: TOX_c_k*TSH2_c
ENO_c_Vmax=598.0; ENO_c_Km2PGA=0.054; ENO_c_Keq=4.17; ENO_c_KmPEP=0.24Reaction: _2PGA_c => PEP_c; _2PGA_c, PEP_c, Rate Law: ENO_c_Vmax*_2PGA_c*(1-PEP_c/(ENO_c_Keq*_2PGA_c))/(ENO_c_Km2PGA*(1+_2PGA_c/ENO_c_Km2PGA+PEP_c/ENO_c_KmPEP))
PFK_g_Vmax=1708.0; PFK_g_KsATP=0.0393; PFK_g_KmFru6P=0.999; PFK_g_KmADP=1.0; PFK_g_KmATP=0.0648; PFK_g_Ki2=10.7; PFK_g_Ki1=15.8; PFK_g_Keq=1035.0Reaction: ATP_g + Fru6P_g => Fru16BP_g + ADP_g; Fru6P_g, ATP_g, Fru16BP_g, ADP_g, Rate Law: PFK_g_Vmax*PFK_g_Ki1*Fru6P_g*ATP_g*(1-Fru16BP_g*ADP_g/(PFK_g_Keq*Fru6P_g*ATP_g))/(PFK_g_KmFru6P*PFK_g_KmATP*(Fru16BP_g+PFK_g_Ki1)*(PFK_g_KsATP/PFK_g_KmATP+Fru6P_g/PFK_g_KmFru6P+ATP_g/PFK_g_KmATP+ADP_g/PFK_g_KmADP+Fru16BP_g*ADP_g/(PFK_g_KmADP*PFK_g_Ki2)+Fru6P_g*ATP_g/(PFK_g_KmFru6P*PFK_g_KmATP)))
_6PGDH_g_KmNADP=0.001; _6PGDH_g_KmNADPH=6.0E-4; _6PGDH_g_Keq=47.0; _6PGDH_g_Km6PG=0.0035; _6PGDH_g_KmRul5P=0.03; _6PGDH_g_Vmax=10.6Reaction: _6PG_g + NADP_g => Rul5P_g + CO2_g + NADPH_g; _6PG_g, NADP_g, Rul5P_g, NADPH_g, Rate Law: _6PGDH_g_Vmax*_6PG_g*NADP_g*(1-Rul5P_g*NADPH_g/(_6PGDH_g_Keq*_6PG_g*NADP_g))/(_6PGDH_g_Km6PG*_6PGDH_g_KmNADP*(1+_6PG_g/_6PGDH_g_Km6PG+Rul5P_g/_6PGDH_g_KmRul5P)*(1+NADP_g/_6PGDH_g_KmNADP+NADPH_g/_6PGDH_g_KmNADPH))
TPI_g_Keq=0.046; TPI_g_KmDHAP=1.2; TPI_g_KmGA3P=0.25; TPI_g_Vmax=999.3Reaction: DHAP_g => GA3P_g; DHAP_g, GA3P_g, Rate Law: TPI_g_Vmax*DHAP_g*(1-GA3P_g/(TPI_g_Keq*DHAP_g))/(TPI_g_KmDHAP*(1+DHAP_g/TPI_g_KmDHAP+GA3P_g/TPI_g_KmGA3P))
FruT_g_k1=250000.0; FruT_g_k2=250000.0Reaction: Fru_c => Fru_g; Fru_c, Fru_g, Rate Law: FruT_g_k1*Fru_c-FruT_g_k2*Fru_g
GlcT_c_Vmax=111.7; GlcT_c_KmGlc=1.0; GlcT_c_alpha=0.75Reaction: Glc_e => Glc_c; Glc_e, Glc_c, Rate Law: GlcT_c_Vmax*(Glc_e-Glc_c)/(GlcT_c_KmGlc+Glc_e+Glc_c+GlcT_c_alpha*Glc_e*Glc_c/GlcT_c_KmGlc)
NADPHu_g_k=2.0Reaction: NADPH_g => NADP_g; NADPH_g, Rate Law: NADPHu_g_k*NADPH_g
PGAM_c_Km3PGA=0.15; PGAM_c_Vmax=225.0; PGAM_c_Km2PGA=0.16; PGAM_c_Keq=0.17Reaction: _3PGA_c => _2PGA_c; _3PGA_c, _2PGA_c, Rate Law: PGAM_c_Vmax*_3PGA_c*(1-_2PGA_c/(PGAM_c_Keq*_3PGA_c))/(PGAM_c_Km3PGA*(1+_3PGA_c/PGAM_c_Km3PGA+_2PGA_c/PGAM_c_Km2PGA))
G3PDH_g_KmDHAP=0.1; G3PDH_g_KmNAD=0.4; G3PDH_g_Vmax=465.0; G3PDH_g_Keq=17085.0; G3PDH_g_KmNADH=0.01; G3PDH_g_KmGly3P=2.0Reaction: NADH_g + DHAP_g => Gly3P_g + NAD_g; DHAP_g, NADH_g, Gly3P_g, NAD_g, Rate Law: G3PDH_g_Vmax*DHAP_g*NADH_g*(1-Gly3P_g*NAD_g/(G3PDH_g_Keq*DHAP_g*NADH_g))/(G3PDH_g_KmDHAP*G3PDH_g_KmNADH*(1+DHAP_g/G3PDH_g_KmDHAP+Gly3P_g/G3PDH_g_KmGly3P)*(1+NADH_g/G3PDH_g_KmNADH+NAD_g/G3PDH_g_KmNAD))
GK_g_Keq=8.37E-4; GK_g_KmATP=0.24; GK_g_KmGly=0.44; GK_g_KmADP=0.56; GK_g_KmGly3P=3.83; GK_g_Vmax=200.0Reaction: Gly3P_g + ADP_g => Gly_e + ATP_g; Gly3P_g, ADP_g, Gly_e, ATP_g, Rate Law: GK_g_Vmax*Gly3P_g*ADP_g*(1-Gly_e*ATP_g/(GK_g_Keq*Gly3P_g*ADP_g))/(GK_g_KmGly3P*GK_g_KmADP*(1+Gly3P_g/GK_g_KmGly3P+Gly_e/GK_g_KmGly)*(1+ADP_g/GK_g_KmADP+ATP_g/GK_g_KmATP))
GlcT_g_k2=250000.0; GlcT_g_k1=250000.0Reaction: Glc_c => Glc_g; Glc_c, Glc_g, Rate Law: GlcT_g_k1*Glc_c-GlcT_g_k2*Glc_g
PGL_c_Km6PG=0.05; PGL_c_k=0.055; PGL_c_Vmax=28.0; PGL_c_Km6PGL=0.05; PGL_c_Keq=20000.0Reaction: _6PGL_c => _6PG_c; _6PGL_c, _6PG_c, Rate Law: PGL_c_k*cytosol*(_6PGL_c-_6PG_c/PGL_c_Keq)+PGL_c_Vmax*_6PGL_c*(1-_6PG_c/(PGL_c_Keq*_6PGL_c))/(PGL_c_Km6PGL*(1+_6PGL_c/PGL_c_Km6PGL+_6PG_c/PGL_c_Km6PG))
HXK_c_KmATP=0.116; HXK_c_KmGlc=0.1; HXK_c_KiFru6P=2.7; HXK_c_Vmax=154.32; HXK_c_KmADP=0.126; HXK_c_KiFru=0.35; HXK_c_Keq=759.0; HXK_c_KmGlc6P=2.7Reaction: Glc_c + ATP_c => Glc6P_c + ADP_c; Fru_c, Fru6P_c, Glc_c, ATP_c, Glc6P_c, ADP_c, Fru_c, Fru6P_c, Rate Law: HXK_c_Vmax*Glc_c*ATP_c*(1-Glc6P_c*ADP_c/(HXK_c_Keq*Glc_c*ATP_c))/(HXK_c_KmGlc*HXK_c_KmATP*(1+Glc_c/HXK_c_KmGlc+Glc6P_c/HXK_c_KmGlc6P)*(1+ATP_c/HXK_c_KmATP+ADP_c/HXK_c_KmADP+Fru_c/HXK_c_KiFru+Fru6P_c/HXK_c_KiFru6P))
AK_c_k1=480.0; AK_c_k2=1000.0Reaction: ADP_c => AMP_c + ATP_c; ADP_c, AMP_c, ATP_c, Rate Law: AK_c_k1*ADP_c^2-AMP_c*ATP_c*AK_c_k2
_6PGDH_c_KmNADP=0.001; _6PGDH_c_Keq=47.0; _6PGDH_c_Vmax=10.6; _6PGDH_c_KmNADPH=6.0E-4; _6PGDH_c_Km6PG=0.0035; _6PGDH_c_KmRul5P=0.03Reaction: NADP_c + _6PG_c => CO2_c + NADPH_c + Rul5P_c; _6PG_c, NADP_c, Rul5P_c, NADPH_c, Rate Law: _6PGDH_c_Vmax*_6PG_c*NADP_c*(1-Rul5P_c*NADPH_c/(_6PGDH_c_Keq*_6PG_c*NADP_c))/(_6PGDH_c_Km6PG*_6PGDH_c_KmNADP*(1+_6PG_c/_6PGDH_c_Km6PG+Rul5P_c/_6PGDH_c_KmRul5P)*(1+NADP_c/_6PGDH_c_KmNADP+NADPH_c/_6PGDH_c_KmNADPH))
PPI_g_KmRul5P=1.4; PPI_g_Vmax=72.0; PPI_g_Keq=5.6; PPI_g_KmRib5P=4.0Reaction: Rul5P_g => Rib5P_g; Rul5P_g, Rib5P_g, Rate Law: PPI_g_Vmax*Rul5P_g*(1-Rib5P_g/(PPI_g_Keq*Rul5P_g))/(PPI_g_KmRul5P*(1+Rul5P_g/PPI_g_KmRul5P+Rib5P_g/PPI_g_KmRib5P))
HXKfru_c_KmFru=0.35; HXKfru_c_KiGlc6P=2.7; HXKfru_c_KmADP=0.126; HXKfru_c_Keq=631.0; HXKfru_c_KmATP=0.116; HXKfru_c_KmFru6P=2.7; HXKfru_c_Vmax=154.32; HXKfru_c_KiGlc=0.1Reaction: Fru_c + ATP_c => ADP_c + Fru6P_c; Glc_c, Glc6P_c, Fru_c, ATP_c, Fru6P_c, ADP_c, Glc_c, Glc6P_c, Rate Law: HXKfru_c_Vmax*Fru_c*ATP_c*(1-Fru6P_c*ADP_c/(HXKfru_c_Keq*Fru_c*ATP_c))/(HXKfru_c_KmFru*HXKfru_c_KmATP*(1+Fru_c/HXKfru_c_KmFru+Fru6P_c/HXKfru_c_KmFru6P)*(1+ATP_c/HXKfru_c_KmATP+ADP_c/HXKfru_c_KmADP+Glc_c/HXKfru_c_KiGlc+Glc6P_c/HXKfru_c_KiGlc6P))
PGK_g_Km13BPGA=0.003; PGK_g_Vmax=2862.0; PGK_g_KmADP=0.1; PGK_g_Km3PGA=1.62; PGK_g_KmATP=0.29; PGK_g_Keq=3377.0Reaction: _13BPGA_g + ADP_g => _3PGA_g + ATP_g; _13BPGA_g, ADP_g, _3PGA_g, ATP_g, Rate Law: PGK_g_Vmax*_13BPGA_g*ADP_g*(1-_3PGA_g*ATP_g/(PGK_g_Keq*_13BPGA_g*ADP_g))/(PGK_g_Km13BPGA*PGK_g_KmADP*(1+_13BPGA_g/PGK_g_Km13BPGA+_3PGA_g/PGK_g_Km3PGA)*(1+ADP_g/PGK_g_KmADP+ATP_g/PGK_g_KmATP))
HXKfru_g_Keq=631.0; HXKfru_g_KmATP=0.116; HXKfru_g_KmFru6P=2.7; HXKfru_g_KiGlc6P=2.7; HXKfru_g_KmADP=0.126; HXKfru_g_KiGlc=0.1; HXKfru_g_Vmax=1774.68; HXKfru_g_KmFru=0.35Reaction: Fru_g + ATP_g => ADP_g + Fru6P_g; Glc_g, Glc6P_g, Fru_g, ATP_g, Fru6P_g, ADP_g, Glc_g, Glc6P_g, Rate Law: HXKfru_g_Vmax*Fru_g*ATP_g*(1-Fru6P_g*ADP_g/(HXKfru_g_Keq*Fru_g*ATP_g))/(HXKfru_g_KmFru*HXKfru_g_KmATP*(1+Fru_g/HXKfru_g_KmFru+Fru6P_g/HXKfru_g_KmFru6P)*(1+ATP_g/HXKfru_g_KmATP+ADP_g/HXKfru_g_KmADP+Glc_g/HXKfru_g_KiGlc+Glc6P_g/HXKfru_g_KiGlc6P))

States:

NameDescription
6PG g[6-phospho-D-gluconic acid]
6PG c[6-phospho-D-gluconic acid]
Rul5P g[D-ribulose 5-phosphate(2-)]
PEP c[phosphoenolpyruvate]
TS2 c[trypanothione]
CO2 g[carbon dioxide]
ATP g[ATP]
Glc c[glucose]
GA3P g[glyceraldehyde 3-phosphate]
Fru16BP g[alpha-D-fructofuranose 1,6-bisphosphate]
Fru g[fructose]
Glc g[glucose]
Glc e[glucose]
TSH2 c[BTO:35490; trypanothione disulfide]
Pyr e[pyruvate]
ADP g[ADP]
13BPGA g[683]
NADP c[NADP(+)]
Fru c[fructose]
DHAP g[glycerone phosphate(2-)]
NAD g[NAD]
NADH g[NADH]
Pyr c[pyruvate]
Fru e[fructose]
CO2 c[carbon dioxide]
6PGL g_6PGL_g
Fru6P g[444848]
2PGA c[59]
Gly3P c[glycerol 1-phosphate]
3PGA c[3-phospho-D-glyceric acid]
NADP g[NADP(+)]
Rib5P g[aldehydo-D-ribose 5-phosphate(2-)]
Pi g[phosphatidylinositol]
Glc6P g[D-glucopyranose 6-phosphate]
ATP c[ATP]
NADPH c[NADPH]
Gly e[glycerol]
6PGL c[6-phosphogluconolactonase3.1.1.17]
Gly3P g[glycerol 1-phosphate]
ADP c[ADP]
AMP g[AMP]
Rul5P c[D-ribulose 5-phosphate(2-)]
3PGA g[3-phospho-D-glyceric acid]
AMP c[AMP]
NADPH g[NADPH]

Kerkhoven2013 - Glycolysis in T.brucei - MODEL A: BIOMD0000000513v0.0.1

Kerkhoven2013 - Glycolysis in T.brucei - MODEL AThere are six models (Model A, B, C, C-fruc, D, D-fruc) described in the…

Details

Dynamic models of metabolism can be useful in identifying potential drug targets, especially in unicellular organisms. A model of glycolysis in the causative agent of human African trypanosomiasis, Trypanosoma brucei, has already shown the utility of this approach. Here we add the pentose phosphate pathway (PPP) of T. brucei to the glycolytic model. The PPP is localized to both the cytosol and the glycosome and adding it to the glycolytic model without further adjustments leads to a draining of the essential bound-phosphate moiety within the glycosome. This phosphate "leak" must be resolved for the model to be a reasonable representation of parasite physiology. Two main types of theoretical solution to the problem could be identified: (i) including additional enzymatic reactions in the glycosome, or (ii) adding a mechanism to transfer bound phosphates between cytosol and glycosome. One example of the first type of solution would be the presence of a glycosomal ribokinase to regenerate ATP from ribose 5-phosphate and ADP. Experimental characterization of ribokinase in T. brucei showed that very low enzyme levels are sufficient for parasite survival, indicating that other mechanisms are required in controlling the phosphate leak. Examples of the second type would involve the presence of an ATP:ADP exchanger or recently described permeability pores in the glycosomal membrane, although the current absence of identified genes encoding such molecules impedes experimental testing by genetic manipulation. Confronted with this uncertainty, we present a modeling strategy that identifies robust predictions in the context of incomplete system characterization. We illustrate this strategy by exploring the mechanism underlying the essential function of one of the PPP enzymes, and validate it by confirming the model predictions experimentally. link: http://identifiers.org/pubmed/24339766

Parameters:

NameDescription
_3PGAT_g_k=250.0Reaction: _3PGA_g => _3PGA_c; _3PGA_g, _3PGA_c, _3PGA_g, _3PGA_c, Rate Law: _3PGAT_g_k*_3PGA_g-_3PGAT_g_k*_3PGA_c
GPO_c_Vmax=368.0; GPO_c_KmGly3P=1.7Reaction: Gly3P_c => DHAP_c; Gly3P_c, Gly3P_c, Rate Law: GPO_c_Vmax*Gly3P_c/(GPO_c_KmGly3P*(1+Gly3P_c/GPO_c_KmGly3P))
GDA_g_k=600.0Reaction: Gly3P_g + DHAP_c => Gly3P_c + DHAP_g; Gly3P_g, DHAP_c, Gly3P_c, DHAP_g, Gly3P_g, DHAP_c, Gly3P_c, DHAP_g, Rate Law: Gly3P_g*GDA_g_k*DHAP_c-Gly3P_c*GDA_g_k*DHAP_g
ENO_c_Vmax=598.0; ENO_c_Km2PGA=0.054; ENO_c_Keq=4.17; ENO_c_KmPEP=0.24Reaction: _2PGA_c => PEP_c; _2PGA_c, PEP_c, _2PGA_c, PEP_c, Rate Law: ENO_c_Vmax*_2PGA_c*(1-PEP_c/(ENO_c_Keq*_2PGA_c))/(ENO_c_Km2PGA*(1+_2PGA_c/ENO_c_Km2PGA+PEP_c/ENO_c_KmPEP))
PFK_g_Vmax=1708.0; PFK_g_KsATP=0.0393; PFK_g_KmFru6P=0.999; PFK_g_KmADP=1.0; PFK_g_KmATP=0.0648; PFK_g_Ki2=10.7; PFK_g_Ki1=15.8; PFK_g_Keq=1035.0Reaction: ATP_g + Fru6P_g => Fru16BP_g + ADP_g; Fru6P_g, ATP_g, Fru16BP_g, ADP_g, Fru6P_g, ATP_g, Fru16BP_g, ADP_g, Rate Law: PFK_g_Vmax*PFK_g_Ki1*Fru6P_g*ATP_g*(1-Fru16BP_g*ADP_g/(PFK_g_Keq*Fru6P_g*ATP_g))/(PFK_g_KmFru6P*PFK_g_KmATP*(Fru16BP_g+PFK_g_Ki1)*(PFK_g_KsATP/PFK_g_KmATP+Fru6P_g/PFK_g_KmFru6P+ATP_g/PFK_g_KmATP+ADP_g/PFK_g_KmADP+Fru16BP_g*ADP_g/(PFK_g_KmADP*PFK_g_Ki2)+Fru6P_g*ATP_g/(PFK_g_KmFru6P*PFK_g_KmATP)))
GlcT_c_Vmax=111.7; GlcT_c_KmGlc=1.0; GlcT_c_alpha=0.75Reaction: Glc_e => Glc_c; Glc_e, Glc_c, Glc_e, Glc_c, Rate Law: GlcT_c_Vmax*(Glc_e-Glc_c)/(GlcT_c_KmGlc+Glc_e+Glc_c+GlcT_c_alpha*Glc_e*Glc_c/GlcT_c_KmGlc)
TPI_g_Keq=0.046; TPI_g_KmDHAP=1.2; TPI_g_KmGA3P=0.25; TPI_g_Vmax=999.3Reaction: DHAP_g => GA3P_g; DHAP_g, GA3P_g, DHAP_g, GA3P_g, Rate Law: TPI_g_Vmax*DHAP_g*(1-GA3P_g/(TPI_g_Keq*DHAP_g))/(TPI_g_KmDHAP*(1+DHAP_g/TPI_g_KmDHAP+GA3P_g/TPI_g_KmGA3P))
ALD_g_KmDHAP=0.015; ALD_g_KiGA3P=0.098; ALD_g_KmGA3P=0.067; ALD_g_Vmax=560.0; ALD_g_KmFru16BP=0.009; ALD_g_KiADP=1.51; ALD_g_KiAMP=3.65; ALD_g_Keq=0.084; ALD_g_KiATP=0.68Reaction: Fru16BP_g => GA3P_g + DHAP_g; ATP_g, ADP_g, AMP_g, Fru16BP_g, GA3P_g, DHAP_g, ATP_g, ADP_g, AMP_g, Fru16BP_g, GA3P_g, DHAP_g, ATP_g, ADP_g, AMP_g, Rate Law: ALD_g_Vmax*Fru16BP_g*(1-GA3P_g*DHAP_g/(Fru16BP_g*ALD_g_Keq))/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP)*(1+GA3P_g/ALD_g_KmGA3P+DHAP_g/ALD_g_KmDHAP+Fru16BP_g/(ALD_g_KmFru16BP*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))+GA3P_g*DHAP_g/(ALD_g_KmGA3P*ALD_g_KmDHAP)+Fru16BP_g*GA3P_g/(ALD_g_KmFru16BP*ALD_g_KiGA3P*(1+ATP_g/ALD_g_KiATP+ADP_g/ALD_g_KiADP+AMP_g/ALD_g_KiAMP))))
ATPu_c_k=50.0Reaction: ATP_c => ADP_c; ATP_c, ADP_c, ATP_c, ADP_c, Rate Law: ATPu_c_k*ATP_c/ADP_c
HXK_g_Vmax=1774.68; HXK_g_KmGlc6P=12.0; HXK_g_KmADP=0.126; HXK_g_Keq=759.0; HXK_g_KmATP=0.116; HXK_g_KmGlc=0.1Reaction: ATP_g + Glc_g => Glc6P_g + ADP_g; Glc_g, ATP_g, Glc6P_g, ADP_g, Glc_g, ATP_g, Glc6P_g, ADP_g, Rate Law: HXK_g_Vmax*Glc_g*ATP_g*(1-Glc6P_g*ADP_g/(HXK_g_Keq*Glc_g*ATP_g))/(HXK_g_KmGlc*HXK_g_KmATP*(1+Glc_g/HXK_g_KmGlc+Glc6P_g/HXK_g_KmGlc6P)*(1+ATP_g/HXK_g_KmATP+ADP_g/HXK_g_KmADP))
PYK_c_KmPyr=50.0; PYK_c_KiADP=0.64; PYK_c_Vmax=1020.0; PYK_c_KmADP=0.114; PYK_c_Keq=10800.0; PYK_c_KiATP=0.57; PYK_c_KmATP=15.0; PYK_c_KmPEP=0.34; PYK_c_n=2.5Reaction: PEP_c + ADP_c => Pyr_c + ATP_c; ADP_c, Pyr_c, ATP_c, PEP_c, ADP_c, Pyr_c, ATP_c, PEP_c, Rate Law: PYK_c_Vmax*ADP_c*(1-Pyr_c*ATP_c/(PYK_c_Keq*PEP_c*ADP_c))*(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n/(PYK_c_KmADP*(1+(PEP_c/(PYK_c_KmPEP*(1+ADP_c/PYK_c_KiADP+ATP_c/PYK_c_KiATP)))^PYK_c_n+Pyr_c/PYK_c_KmPyr)*(1+ADP_c/PYK_c_KmADP+ATP_c/PYK_c_KmATP))
PGAM_c_Km3PGA=0.15; PGAM_c_Vmax=225.0; PGAM_c_Km2PGA=0.16; PGAM_c_Keq=0.17Reaction: _3PGA_c => _2PGA_c; _3PGA_c, _2PGA_c, _3PGA_c, _2PGA_c, Rate Law: PGAM_c_Vmax*_3PGA_c*(1-_2PGA_c/(PGAM_c_Keq*_3PGA_c))/(PGAM_c_Km3PGA*(1+_3PGA_c/PGAM_c_Km3PGA+_2PGA_c/PGAM_c_Km2PGA))
PGI_g_Vmax=1305.0; PGI_g_Ki6PG=0.14; PGI_g_KmGlc6P=0.4; PGI_g_Keq=0.457; _6PG_g=0.0841958; PGI_g_KmFru6P=0.12Reaction: Glc6P_g => Fru6P_g; Glc6P_g, Fru6P_g, Glc6P_g, Fru6P_g, Rate Law: PGI_g_Vmax*Glc6P_g*(1-Fru6P_g/(PGI_g_Keq*Glc6P_g))/(PGI_g_KmGlc6P*(1+Glc6P_g/PGI_g_KmGlc6P+Fru6P_g/PGI_g_KmFru6P+_6PG_g/PGI_g_Ki6PG))
G3PDH_g_KmDHAP=0.1; G3PDH_g_KmNAD=0.4; G3PDH_g_Vmax=465.0; G3PDH_g_Keq=17085.0; G3PDH_g_KmNADH=0.01; G3PDH_g_KmGly3P=2.0Reaction: NADH_g + DHAP_g => Gly3P_g + NAD_g; DHAP_g, NADH_g, Gly3P_g, NAD_g, DHAP_g, NADH_g, Gly3P_g, NAD_g, Rate Law: G3PDH_g_Vmax*DHAP_g*NADH_g*(1-Gly3P_g*NAD_g/(G3PDH_g_Keq*DHAP_g*NADH_g))/(G3PDH_g_KmDHAP*G3PDH_g_KmNADH*(1+DHAP_g/G3PDH_g_KmDHAP+Gly3P_g/G3PDH_g_KmGly3P)*(1+NADH_g/G3PDH_g_KmNADH+NAD_g/G3PDH_g_KmNAD))
GK_g_Keq=8.37E-4; GK_g_KmATP=0.24; GK_g_KmGly=0.44; GK_g_KmADP=0.56; GK_g_KmGly3P=3.83; GK_g_Vmax=200.0Reaction: Gly3P_g + ADP_g => Gly_e + ATP_g; Gly3P_g, ADP_g, Gly_e, ATP_g, Gly3P_g, ADP_g, Gly_e, ATP_g, Rate Law: GK_g_Vmax*Gly3P_g*ADP_g*(1-Gly_e*ATP_g/(GK_g_Keq*Gly3P_g*ADP_g))/(GK_g_KmGly3P*GK_g_KmADP*(1+Gly3P_g/GK_g_KmGly3P+Gly_e/GK_g_KmGly)*(1+ADP_g/GK_g_KmADP+ATP_g/GK_g_KmATP))
GlcT_g_k2=250000.0; GlcT_g_k1=250000.0Reaction: Glc_c => Glc_g; Glc_c, Glc_g, Glc_c, Glc_g, Rate Law: GlcT_g_k1*Glc_c-GlcT_g_k2*Glc_g
AK_g_k2=1000.0; AK_g_k1=480.0Reaction: ADP_g => AMP_g + ATP_g; ADP_g, AMP_g, ATP_g, ADP_g, AMP_g, ATP_g, Rate Law: AK_g_k1*ADP_g^2-AMP_g*ATP_g*AK_g_k2
PyrT_c_Vmax=230.0; PyrT_c_KmPyr=1.96Reaction: Pyr_c => Pyr_e; Pyr_c, Pyr_c, Rate Law: PyrT_c_Vmax*Pyr_c/(PyrT_c_KmPyr*(1+Pyr_c/PyrT_c_KmPyr))
AK_c_k1=480.0; AK_c_k2=1000.0Reaction: ADP_c => AMP_c + ATP_c; ADP_c, AMP_c, ATP_c, ADP_c, AMP_c, ATP_c, Rate Law: AK_c_k1*ADP_c^2-AMP_c*ATP_c*AK_c_k2
PGK_g_Km13BPGA=0.003; PGK_g_Vmax=2862.0; PGK_g_KmADP=0.1; PGK_g_Km3PGA=1.62; PGK_g_KmATP=0.29; PGK_g_Keq=3377.0Reaction: _13BPGA_g + ADP_g => _3PGA_g + ATP_g; _13BPGA_g, ADP_g, _3PGA_g, ATP_g, _13BPGA_g, ADP_g, _3PGA_g, ATP_g, Rate Law: PGK_g_Vmax*_13BPGA_g*ADP_g*(1-_3PGA_g*ATP_g/(PGK_g_Keq*_13BPGA_g*ADP_g))/(PGK_g_Km13BPGA*PGK_g_KmADP*(1+_13BPGA_g/PGK_g_Km13BPGA+_3PGA_g/PGK_g_Km3PGA)*(1+ADP_g/PGK_g_KmADP+ATP_g/PGK_g_KmATP))
GAPDH_g_Vmax=720.9; GAPDH_g_Km13BPGA=0.1; GAPDH_g_KmNAD=0.45; GAPDH_g_KmNADH=0.02; GAPDH_g_KmGA3P=0.15; GAPDH_g_Keq=0.066Reaction: GA3P_g + NAD_g + Pi_g => NADH_g + _13BPGA_g; GA3P_g, NAD_g, _13BPGA_g, NADH_g, GA3P_g, NAD_g, _13BPGA_g, NADH_g, Rate Law: GAPDH_g_Vmax*GA3P_g*NAD_g*(1-_13BPGA_g*NADH_g/(GAPDH_g_Keq*GA3P_g*NAD_g))/(GAPDH_g_KmGA3P*GAPDH_g_KmNAD*(1+GA3P_g/GAPDH_g_KmGA3P+_13BPGA_g/GAPDH_g_Km13BPGA)*(1+NAD_g/GAPDH_g_KmNAD+NADH_g/GAPDH_g_KmNADH))

States:

NameDescription
Fru6P g[444848]
PEP c[phosphoenolpyruvate]
2PGA c[59]
ATP g[ATP]
Gly3P c[glycerol 1-phosphate]
Fru16BP g[alpha-D-fructofuranose 1,6-bisphosphate]
GA3P g[glyceraldehyde 3-phosphate]
3PGA c[3-phospho-D-glyceric acid]
Glc c[glucose]
Glc g[glucose]
Glc e[glucose]
Pi g[phosphatidylinositol]
Glc6P g[D-glucopyranose 6-phosphate]
ATP c[ATP]
Pyr e[pyruvate]
DHAP c[glycerone phosphate(2-)]
13BPGA g[683]
ADP g[ADP]
DHAP g[glycerone phosphate(2-)]
Gly e[glycerol]
NAD g[NAD]
Gly3P g[glycerol 1-phosphate]
ADP c[ADP]
AMP g[AMP]
NADH g[NADH]
Pyr c[pyruvate]
3PGA g[3-phospho-D-glyceric acid]
AMP c[AMP]

Kervizic2008_Cholesterol_SREBP: MODEL0568648427v0.0.1

# Model of cholesterol regulation (with Boolean Formulae) (2008) This model is described in **Dynamical modeling of th…

Details

BACKGROUND: Qualitative dynamics of small gene regulatory networks have been studied in quite some details both with synchronous and asynchronous analysis. However, both methods have their drawbacks: synchronous analysis leads to spurious attractors and asynchronous analysis lacks computational efficiency, which is a problem to simulate large networks. We addressed this question through the analysis of a major biosynthesis pathway. Indeed the cholesterol synthesis pathway plays a pivotal role in dislypidemia and, ultimately, in cancer through intermediates such as mevalonate, farnesyl pyrophosphate and geranyl geranyl pyrophosphate, but no dynamic model of this pathway has been proposed until now. RESULTS: We set up a computational framework to dynamically analyze large biological networks. This framework associates a classical and computationally efficient synchronous Boolean analysis with a newly introduced method based on Markov chains, which identifies spurious cycles among the results of the synchronous simulation. Based on this method, we present here the results of the analysis of the cholesterol biosynthesis pathway and its physiological regulation by the Sterol Response Element Binding Proteins (SREBPs), as well as the modeling of the action of statins, inhibitor drugs, on this pathway. The in silico experiments show the blockade of the cholesterol endogenous synthesis by statins and its regulation by SREPBs, in full agreement with the known biochemical features of the pathway. CONCLUSION: We believe that the method described here to identify spurious cycles opens new routes to compute large and biologically relevant models, thanks to the computational efficiency of synchronous simulation. Furthermore, to the best of our knowledge, we present here the first dynamic systems biology model of the human cholesterol pathway and several of its key regulatory control elements, hoping it would provide a good basis to perform in silico experiments and confront the resulting properties with published and experimental data. The model of the cholesterol pathway and its regulation, along with Boolean formulae used for simulation are available on our web site http://Bioinformaticsu613.free.fr. Graphical results of the simulation are also shown online. The SBML model is available in the BioModels database http://www.ebi.ac.uk/biomodels/ with submission ID: MODEL0568648427. link: http://identifiers.org/pubmed/19025648

Khajanchi2015 - The combined effects of optimal control in cancer remission: BIOMD0000000897v0.0.1

The combined effects of optimal control in cancer remission SubhasKhajanchi DibakarGhosh Abstract We investigate a math…

Details

We investigate a mathematical model depicting the nonlinear dynamics of immunogenic tumors as envisioned by Kuznetsov et al. [1]. To understand the dynamics under what circumstances the cancer cells can be eliminated, we implement the theory of optimal control. We design two types of external treatment strategies, one is Adoptive Cellular Immunotherapy and another is interleukin-2. Our aim is to establish the treatment regimens that maximize the effector cell count and minimize the tumor cell burden and the deleterious effects of the total amount of drugs. We derive the existence of an optimal control by using the boundedness of solutions. We characterize the optimality system, in which the state system is coupled with co-states. The uniqueness of an optimal control of our problem is also analyzed. Finally, we demonstrate the numerical illustrations that the optimal regimens reduce the tumor burden under different scenarios. link: http://identifiers.org/doi/10.1016/j.amc.2015.09.012

Parameters:

NameDescription
s = 13000.0; e1 = 1.0; p = 0.1245; g = 2.019E7Reaction: => E; T, Rate Law: compartment*(s*e1+p*E*T/(g+T))
n = 1.101E-7; e2 = 0.0Reaction: T => ; E, Rate Law: compartment*(n*E*T+e2*T)
m = 3.422E-10; d = 0.0412Reaction: E => ; T, Rate Law: compartment*(m*E*T+d*E)
b = 2.0E-9; a = 0.18Reaction: => T, Rate Law: compartment*a*T*(1-b*T)

States:

NameDescription
T[Neoplastic Cell]
E[Effector Immune Cell]

Khajanchi2017 - Uniform Persistence and Global Stability for a Brain Tumor and Immune System Interaction: BIOMD0000000921v0.0.1

This paper describes the synergistic interaction between the growth of malignant gliomas and the immune system interacti…

Details

This paper describes the synergistic interaction between the growth of malignant gliomas and the immune system interactions using a system of coupled ordinary di®erential equations (ODEs). The proposed mathematical model comprises the interaction of glioma cells, macrophages, activated Cytotoxic T-Lymphocytes (CTLs), the immunosuppressive factor TGF- and the immuno-stimulatory factor IFN-. The dynamical behavior of the proposed system both analytically and numerically is investigated from the point of view of stability. By constructing Lyapunov functions, the global behavior of the glioma-free and the interior equilibrium point have been analyzed under some assumptions. Finally, we perform numerical simulations in order to illustrate our analytical ¯ndings by varying the system parameters. link: http://identifiers.org/doi/10.1142/S1793048017500114

Parameters:

NameDescription
s1 = 63305.0Reaction: => T_beta, Rate Law: compartment*s1
alpha4 = 0.1694; k3 = 334450.0Reaction: C_T => ; G, Rate Law: compartment*alpha4*G/(G+k3)*C_T
mu2 = 6.93Reaction: T_beta =>, Rate Law: compartment*mu2*T_beta
b2 = 1.02E-4Reaction: => I_gamma; C_T, Rate Law: compartment*b2*C_T
alpha3 = 0.0194; k2 = 27000.0Reaction: M => ; G, Rate Law: compartment*alpha3*G/(G+k2)*M
alpha2 = 0.12; k1 = 27000.0; alpha1 = 1.5; e1 = 10000.0Reaction: G => ; T_beta, M, C_T, Rate Law: compartment*1/(T_beta+e1)*(alpha1*M+alpha2*C_T)*G/(G+k1)
k5 = 2000.0; a2 = 0.0Reaction: => C_T; G, T_beta, Rate Law: compartment*a2*G/(k5+T_beta)
a1 = 0.1163; e2 = 10000.0; k4 = 10500.0Reaction: => M; I_gamma, T_beta, Rate Law: compartment*a1*I_gamma/(k4+I_gamma)*1/(T_beta+e2)
mu1 = 0.007Reaction: C_T =>, Rate Law: compartment*mu1*C_T
r1 = 0.01; G_max = 882650.0Reaction: => G, Rate Law: compartment*r1*G*(1-G/G_max)
M_max = 1.0; r2 = 0.3307Reaction: => M, Rate Law: compartment*r2*M*(1-M/M_max)
mu3 = 0.102Reaction: I_gamma =>, Rate Law: compartment*mu3*I_gamma
b1 = 5.75E-6Reaction: => T_beta; G, Rate Law: compartment*b1*G

States:

NameDescription
C T[C12543]
M[macrophage]
T beta[C30098]
G[glioma cell]
I gamma[Interferon Gamma]