SBMLBioModels: Q - S

Q


Qosa2014 - Mechanistic modeling that describes amyloid-Beta clearance across BBB: MODEL1409240002v0.0.1

Qosa2014 - Mechanistic modeling that describes Aβ clearance across BBBQosa2014 - Mechanistic modeling that describes Aβ…

Details

Alzheimer's disease (AD) has a characteristic hallmark of amyloid-β (Aβ) accumulation in the brain. This accumulation of Aβ has been related to its faulty cerebral clearance. Indeed, preclinical studies that used mice to investigate Aβ clearance showed that efflux across blood-brain barrier (BBB) and brain degradation mediate efficient Aβ clearance. However, the contribution of each process to Aβ clearance remains unclear. Moreover, it is still uncertain how species differences between mouse and human could affect Aβ clearance. Here, a modified form of the brain efflux index method was used to estimate the contribution of BBB and brain degradation to Aβ clearance from the brain of wild type mice. We estimated that 62% of intracerebrally injected (125)I-Aβ40 is cleared across BBB while 38% is cleared by brain degradation. Furthermore, in vitro and in silico studies were performed to compare Aβ clearance between mouse and human BBB models. Kinetic studies for Aβ40 disposition in bEnd3 and hCMEC/D3 cells, representative in vitro mouse and human BBB models, respectively, demonstrated 30-fold higher rate of (125)I-Aβ40 uptake and 15-fold higher rate of degradation by bEnd3 compared to hCMEC/D3 cells. Expression studies showed both cells to express different levels of P-glycoprotein and RAGE, while LRP1 levels were comparable. Finally, we established a mechanistic model, which could successfully predict cellular levels of (125)I-Aβ40 and the rate of each process. Established mechanistic model suggested significantly higher rates of Aβ uptake and degradation in bEnd3 cells as rationale for the observed differences in (125)I-Aβ40 disposition between mouse and human BBB models. In conclusion, current study demonstrates the important role of BBB in the clearance of Aβ from the brain. Moreover, it provides insight into the differences between mouse and human BBB with regards to Aβ clearance and offer, for the first time, a mathematical model that describes Aβ clearance across BBB. link: http://identifiers.org/pubmed/24467845

Qu2003_CellCycle: BIOMD0000000110v0.0.1

This model is from the article: Dynamics of the cell cycle: checkpoints, sizers, and timers. Qu Z, MacLellan WR…

Details

We have developed a generic mathematical model of a cell cycle signaling network in higher eukaryotes that can be used to simulate both the G1/S and G2/M transitions. In our model, the positive feedback facilitated by CDC25 and wee1 causes bistability in cyclin-dependent kinase activity, whereas the negative feedback facilitated by SKP2 or anaphase-promoting-complex turns this bistable behavior into limit cycle behavior. The cell cycle checkpoint is a Hopf bifurcation point. These behaviors are coordinated by growth and division to maintain normal cell cycle and size homeostasis. This model successfully reproduces sizer, timer, and the restriction point features of the eukaryotic cell cycle, in addition to other experimental findings. link: http://identifiers.org/pubmed/14645053

Parameters:

NameDescription
k4 = 30.0; k3 = 30.0Reaction: y => x1; c, Rate Law: cell*(k3*c*y-x1*k4)
k16u = 25.0; k16 = 2.0Reaction: ixp => x, Rate Law: cell*k16*k16u*ixp
bi = 0.1; ci = 1.0; ai = 10.0Reaction: ix => ixp; x, Rate Law: cell*((bi+ci*x)*ix-ai*ixp)
k1 = 300.0Reaction: => y, Rate Law: k1*cell
k11 = 1.0Reaction: w0 =>, Rate Law: cell*w0*k11
k2 = 5.0; k2u = 50.0Reaction: y => ; u, Rate Law: cell*(k2+k2u*u)*y
k10 = 10.0Reaction: => w0, Rate Law: k10*cell
bw = 0.1; cw = 1.0; aw = 10.0Reaction: w0 => w1; x, Rate Law: cell*((bw+cw*x)*w0-aw*w1)
a = 4.0; Tau = 25.0Reaction: => u; x, Rate Law: cell*x^2/(a^2+x^2)/Tau
cz = 1.0; bz = 0.1; az = 10.0Reaction: z0 => z1; x, Rate Law: cell*((bz+cz*x)*z0-z1*az)
k7u = 0.0; k7 = 10.0Reaction: x => ; u, Rate Law: cell*(k7+k7u*u)*x
Tau = 25.0Reaction: u =>, Rate Law: cell*u/Tau
k14 = 1.0; k15 = 1.0Reaction: i + x => ix, Rate Law: (k14*x*i-k15*ix)*cell
k5 = 0.1; k6 = 1.0Reaction: x => x1; z2, w0, Rate Law: cell*((k6+w0)*x-(k5+z2)*x1)
k13 = 1.0Reaction: i =>, Rate Law: cell*k13*i
k12 = 0.0Reaction: => i, Rate Law: k12*cell
k9 = 1.0Reaction: z0 =>, Rate Law: cell*k9*z0
k8 = 100.0Reaction: => z0, Rate Law: cell*k8

States:

NameDescription
ix[IPR003175; IPR006670; cyclin-dependent protein kinase holoenzyme complex]
i[IPR003175]
c[cyclin-dependent protein kinase holoenzyme complex]
z1[Cell division control protein 25]
x[IPR006670; cyclin-dependent protein kinase holoenzyme complex]
z0[Cell division control protein 25]
w1[Wee1-like protein kinase]
x1[IPR006670; cyclin-dependent protein kinase holoenzyme complex]
totalCyclin[IPR006670]
ixp[IPR003175; IPR006670; cyclin-dependent protein kinase holoenzyme complex]
u[IPR001810; anaphase-promoting complex]
z2[Cell division control protein 25]
w0[Wee1-like protein kinase]
y[IPR006670]

Quek2008 - Genome-scale metabolic network of Mus musculus: MODEL1507180067v0.0.1

Quek2008 - Genome-scale metabolic network of Mus musculusThis model is described in the article: [On the reconstruction…

Details

Genome-scale metabolic modeling is a systems-based approach that attempts to capture the metabolic complexity of the whole cell, for the purpose of gaining insight into metabolic function and regulation. This is achieved by organizing the metabolic components and their corresponding interactions into a single context. The reconstruction process is a challenging and laborious task, especially during the stage of manual curation. For the mouse genome-scale metabolic model, however, we were able to rapidly reconstruct a compartmentalized model from well-curated metabolic databases online. The prototype model was comprehensive. Apart from minor compound naming and compartmentalization issues, only nine additional reactions without gene associations were added during model curation before the model was able to simulate growth in silico. Further curation led to a metabolic model that consists of 1399 genes mapped to 1757 reactions, with a total of 2037 reactions compartmentalized into the cytoplasm and mitochondria, capable of reproducing metabolic functions inferred from literatures. The reconstruction is made more tractable by developing a formal system to update the model against online databases. Effectively, we can focus our curation efforts into establishing better model annotations and gene-protein-reaction associations within the core metabolism, while relying on genome and proteome databases to build new annotations for peripheral pathways, which may bear less relevance to our modeling interest. link: http://identifiers.org/pubmed/19425150

Quek2014 - Metabolic flux analysis of HEK cell culture using Recon 2 (reduced version of Recon 2): MODEL1504080000v0.0.1

Quek2014 - Metabolic flux analysis of HEK cell culture using Recon 2 (reduced version of Recon 2)This model is described…

Details

A representative stoichiometric model is essential to perform metabolic flux analysis (MFA) using experimentally measured consumption (or production) rates as constraints. For Human Embryonic Kidney (HEK) cell culture, there is the opportunity to use an extremely well-curated and annotated human genome-scale model Recon 2 for MFA. Performing MFA using Recon 2 without any modification would have implied that cells have access to all functionality encoded by the genome, which is not realistic. The majority of intracellular fluxes are poorly determined as only extracellular exchange rates are measured. This is compounded by the fact that there is no suitable metabolic objective function to suppress non-specific fluxes. We devised a heuristic to systematically reduce Recon 2 to emphasize flux through core metabolic reactions. This implies that cells would engage these dominant metabolic pathways to grow, and any significant changes in gross metabolic phenotypes would have invoked changes in these pathways. The reduced metabolic model becomes a functionalized version of Recon 2 used for identifying significant metabolic changes in cells by flux analysis. link: http://identifiers.org/pubmed/24907410

Queralt2006 - Initiation of mitotic exit by downregulation of PP2A in budding yeast: BIOMD0000000953v0.0.1

Mathematical model of mitotic exit in budding yeast.

Details

After anaphase, the high mitotic cyclin-dependent kinase (Cdk) activity is downregulated to promote exit from mitosis. To this end, in the budding yeast S. cerevisiae, the Cdk counteracting phosphatase Cdc14 is activated. In metaphase, Cdc14 is kept inactive in the nucleolus by its inhibitor Net1. During anaphase, Cdk- and Polo-dependent phosphorylation of Net1 is thought to release active Cdc14. How Net1 is phosphorylated specifically in anaphase, when mitotic kinase activity starts to decline, has remained unexplained. Here, we show that PP2A(Cdc55) phosphatase keeps Net1 underphosphorylated in metaphase. The sister chromatid-separating protease separase, activated at anaphase onset, interacts with and downregulates PP2A(Cdc55), thereby facilitating Cdk-dependent Net1 phosphorylation. PP2A(Cdc55) downregulation also promotes phosphorylation of Bfa1, contributing to activation of the "mitotic exit network" that sustains Cdc14 as Cdk activity declines. These findings allow us to present a new quantitative model for mitotic exit in budding yeast. link: http://identifiers.org/pubmed/16713564

Queralt2006_MitoticExit_Cdc55DownregulationBySeparase: BIOMD0000000409v0.0.1

This model is from the article: Downregulation of PP2A(Cdc55) phosphatase by separase initiates mitotic exit in buddin…

Details

After anaphase, the high mitotic cyclin-dependent kinase (Cdk) activity is downregulated to promote exit from mitosis. To this end, in the budding yeast S. cerevisiae, the Cdk counteracting phosphatase Cdc14 is activated. In metaphase, Cdc14 is kept inactive in the nucleolus by its inhibitor Net1. During anaphase, Cdk- and Polo-dependent phosphorylation of Net1 is thought to release active Cdc14. How Net1 is phosphorylated specifically in anaphase, when mitotic kinase activity starts to decline, has remained unexplained. Here, we show that PP2A(Cdc55) phosphatase keeps Net1 underphosphorylated in metaphase. The sister chromatid-separating protease separase, activated at anaphase onset, interacts with and downregulates PP2A(Cdc55), thereby facilitating Cdk-dependent Net1 phosphorylation. PP2A(Cdc55) downregulation also promotes phosphorylation of Bfa1, contributing to activation of the "mitotic exit network" that sustains Cdc14 as Cdk activity declines. These findings allow us to present a new quantitative model for mitotic exit in budding yeast. link: http://identifiers.org/pubmed/16713564

Parameters:

NameDescription
kd = 0.45; Jnet = 0.2; kad = 0.1Reaction: Net1P => Net1; Cdc14, Clb2, PP2A, Rate Law: (kad*Cdc14+kd*PP2A)*Net1P/(Jnet+Net1P)
kssecurin = 0.03Reaction: AA => securinT + securin, Rate Law: kssecurin
ldnet = 1.0Reaction: Net1Cdc14 => Net1, Rate Law: ldnet*Net1Cdc14
kadpolo = 0.25; kdpolo = 0.01Reaction: Polo => degr; Cdh1, Rate Law: (kdpolo+kadpolo*Cdh1)*Polo
Jpolo = 0.25; kipolo = 0.1Reaction: Polo => Polo_i, Rate Law: kipolo*Polo/(Jpolo+Polo)
PP2AT = 1.0; kpp = 0.1; ki = 20.0Reaction: PP2A = (1+kpp*ki*separase)/(1+ki*separase)*PP2AT, Rate Law: missing
kdsecurin = 0.05; kadsecurin = 2.0Reaction: securinT + securin => degr; Cdc20, Rate Law: (kdsecurin+kadsecurin*Cdc20)*securinT
kaicdc15 = 0.2; kicdc15 = 0.0; Jcdc15 = 0.2; Cdk = NaNReaction: Cdc15 => Cdc15_i, Rate Law: (kicdc15+kaicdc15*Cdk)*Cdc15/(Jcdc15+Cdc15)
Cdh1T = 1.0; Jcdh = 0.0015; kadcdh = 1.0; kdcdh = 0.01Reaction: Cdh1_i => Cdh1; Cdc14, Rate Law: (kdcdh+kadcdh*Cdc14)*(Cdh1T-Cdh1)/((Jcdh+Cdh1T)-Cdh1)
kitem = 0.1; kaitem = 1.0; Jtem1 = 0.005Reaction: Tem1 => Tem1_i; PP2A, Rate Law: (kitem+kaitem*PP2A)*Tem1/(Jtem1+Tem1)
kdcdc20 = 0.05; kadcdc20 = 2.0Reaction: Cdc20 => degr; Cdh1, Rate Law: (kdcdc20+kadcdc20*Cdh1)*Cdc20
lamen = 10.0Reaction: AA => MEN; Tem1, Cdc15, Rate Law: lamen*(Tem1-MEN)*(Cdc15-MEN)
kp = 0.4; Jnet = 0.2; kap = 2.0; Cdk = NaNReaction: Net1Cdc14 => Net1P; MEN, Net1, Clb2, Rate Law: (kp*Cdk+kap*MEN)*Net1Cdc14/(Jnet+Net1+Net1Cdc14)
Cdc14T = 0.5Reaction: Cdc14 = Cdc14T-Net1Cdc14, Rate Law: missing
ksclb2 = 0.03Reaction: AA => Clb2, Rate Law: ksclb2
Net1T = 1.0Reaction: Net1P = (Net1T-Net1)-Net1Cdc14, Rate Law: missing
Tem1T = 1.0Reaction: Tem1_i = Tem1T-Tem1, Rate Law: missing
kaacdc15 = 0.5; Jcdc15 = 0.2; kacdc15 = 0.02; Cdc15T = 1.0Reaction: Cdc15_i => Cdc15; Cdc14, Rate Law: (kacdc15+kaacdc15*Cdc14)*(Cdc15T-Cdc15)/((Jcdc15+Cdc15T)-Cdc15)
kadclb2 = 0.2; kaadclb2 = 2.0; kdclb2 = 0.03Reaction: Clb2 => degr; Cdc20, Cdh1, Rate Law: (kdclb2+kadclb2*Cdc20+kaadclb2*Cdh1)*Clb2
kscdc20 = 0.015Reaction: AA => Cdc20, Rate Law: kscdc20
ksseparase = 0.001Reaction: AA => separaseT + separase, Rate Law: ksseparase
lanet = 500.0Reaction: Net1 => Net1Cdc14; Cdc14, Rate Law: lanet*Net1*Cdc14
ldmen = 0.1Reaction: MEN => degr, Rate Law: ldmen*MEN
kaapolo = 0.5; Jpolo = 0.25; kapolo = 0.0; Cdk = NaNReaction: Polo_i => Polo; PoloT, Rate Law: (kapolo+kaapolo*Cdk)*(PoloT-Polo)/((Jpolo+PoloT)-Polo)
kspolo = 0.01Reaction: AA => PoloT + Polo_i, Rate Law: kspolo
Jcdh = 0.0015; Cdk = NaN; kapcdh = 1.0Reaction: Cdh1 => Cdh1_i, Rate Law: kapcdh*Cdk*Cdh1/(Jcdh+Cdh1)
kaatem = 0.5; katem = 0.0; Tem1T = 1.0; Jtem1 = 0.005Reaction: Tem1_i => Tem1; Polo, Rate Law: (katem+kaatem*Polo)*(Tem1T-Tem1)/((Jtem1+Tem1T)-Tem1)
ldsecurin = 1.0; lasecurin = 500.0Reaction: securin + separase => securinseparase, Rate Law: lasecurin*securin*separase-ldsecurin*securinseparase
Cdc15T = 1.0Reaction: Cdc15_i = Cdc15T-Cdc15, Rate Law: missing
kdseparase = 0.004Reaction: separaseT + separase => degr, Rate Law: kdseparase*separaseT
Cdh1T = 1.0Reaction: Cdh1_i = Cdh1T-Cdh1, Rate Law: missing

States:

NameDescription
Polo[Cell cycle serine/threonine-protein kinase CDC5/MSD2]
securinT[Securin]
Polo i[Cell cycle serine/threonine-protein kinase CDC5/MSD2]
MEN[Protein TEM1; Cell division control protein 15]
Tem1[Protein TEM1]
Tem1 i[Protein TEM1]
Net1[Nucleolar protein NET1]
degrdegr
Cdh1 i[APC/C activator protein CDH1]
separaseT[Separin]
Clb2[G2/mitotic-specific cyclin-2]
separase[Separin]
Cdc15[Cell division control protein 15]
Cdc14[Tyrosine-protein phosphatase CDC14]
securinseparase[Securin; Separin]
Net1Cdc14[Nucleolar protein NET1; Tyrosine-protein phosphatase CDC14]
securin[Securin]
PP2A[Protein phosphatase PP2A regulatory subunit B]
PoloT[Cell cycle serine/threonine-protein kinase CDC5/MSD2]
Net1P[Nucleolar protein NET1; Phosphoprotein]
AA[alpha-amino acid]
Cdh1[APC/C activator protein CDH1]
Cdc20[APC/C activator protein CDC20]
Cdc15 i[Cell division control protein 15]

R


Radosavljevic2009_BioterroristAttack_PanicProtection_1: BIOMD0000000836v0.0.1

This model is from the article: Epidemics of panic during a bioterrorist attack--a mathematical model. Radosavljevic…

Details

A bioterrorist attacks usually cause epidemics of panic in a targeted population. We have presented epidemiologic aspect of this phenomenon as a three-component model–host, information on an attack and social network. We have proposed a mathematical model of panic and counter-measures as the function of time in a population exposed to a bioterrorist attack. The model comprises ordinary differential equations and graphically presented combinations of the equations parameters. Clinically, we have presented a model through a sequence of psychic conditions and disorders initiated by an act of bioterrorism. This model might be helpful for an attacked community to timely and properly apply counter-measures and to minimize human mental suffering during a bioterrorist attack. link: http://identifiers.org/pubmed/19423234

Parameters:

NameDescription
delta = 1.0; gamma = 0.0Reaction: P = ((-gamma)+delta*S)*P, Rate Law: ((-gamma)+delta*S)*P
alpha = 6.0; C = 10.0; beta = 2.8Reaction: S = (alpha*(1-S/C)-beta*P)*S, Rate Law: (alpha*(1-S/C)-beta*P)*S

States:

NameDescription
Spanic_intensity
Pprotection+prevention_intensity

Radulescu2008 - NF-κB hierarchy ℳ(16,34,46): MODEL7743576806v0.0.1

Radulescu2008 - NF-κB hierarchy ℳ(16,34,46)This is a model of NF-κB pathway functioning from hierarchy of models of decr…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Radulescu2008_NFkB_hierarchy_M_5_8_12: MODEL7743212613v0.0.1

# NFkB model M(5,8,12) - minimal model This is a model of NFkB pathway functioning from hierarchy of models of decreasi…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Radulescu2008_NFkB_hierarchy_M_6_10_15: MODEL7743315447v0.0.1

# NFkB model M(6,10,15) This is a model of NFkB pathway functioning from hierarchy of models of decreasing complexity,…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Radulescu2008_NFkB_hierarchy_M_8_12_19: MODEL7743358405v0.0.1

# NFkB model M(8,12,19) This is a model of NFkB pathway functioning from hierarchy of models of decreasing complexity,…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Radulescu2008_NFkB_hierarchy_M_14_25_28_Lipniacky: BIOMD0000000226v0.0.1

# NFkB model M(14,25,28) - Lipniacky's NFkB model This is a model of NFkB pathway functioning from hierarchy of models…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Parameters:

NameDescription
kr13 = 0.0; kf13 = 18.4Reaction: s160 + s161 => s135, Rate Law: kf13*s161*s160-kr13*s135
k8 = 0.1Reaction: s139 => s132, Rate Law: k8*s139
kf28 = 0.01; kr28 = 0.0Reaction: s159 => s135, Rate Law: kf28*s159-kr28*s135
k3 = 2.5E-6Reaction: s150 => s133, Rate Law: k3
k10 = 0.1Reaction: s152 => s161 + s132, Rate Law: k10*s152
k19 = 0.0; k20 = 5.0E-7Reaction: s126 => s127; s164, Rate Law: k19+k20*s164
k6 = 1.25E-4Reaction: s132 => s134, Rate Law: k6*s132
k21 = 1.0E-4Reaction: s160 => s122, Rate Law: k21*s160
kr23 = 5.0E-4; kf23 = 0.001Reaction: s160 => s167, Rate Law: kf23*s160-kr23*s167
k27 = 4.0E-4Reaction: s125 => s124, Rate Law: k27*s125
k11 = 1.25E-4Reaction: s130 => s129, Rate Law: k11*s130
k9 = 1.0Reaction: s132 + s135 => s152, Rate Law: k9*s132*s135
k17 = 4.0E-4Reaction: s127 => s153, Rate Law: k17*s127
k26 = 5.0E-7Reaction: s121 => s125; s164, Rate Law: k26*s164
k2 = 1.25E-4Reaction: s133 => s131, Rate Law: k2*s133
kr14 = 0.0; kf14 = 18.4Reaction: s164 + s167 => s159, Rate Law: kf14*s164*s167-kr14*s159
k7 = 0.2Reaction: s160 + s132 => s139, Rate Law: k7*s132*s160
k5 = 0.0015; k4 = 0.1Reaction: s132 => s130; s128, Rate Law: k5*s132+k4*s132*s128
k22 = 0.5Reaction: s125 => s160 + s125, Rate Law: k22*s125
k18 = 3.0E-4Reaction: s128 => s154, Rate Law: k18*s128
k1 = 0.0025Reaction: s133 => s132, Rate Law: k1*s133
k16 = 0.5Reaction: s127 => s128 + s127, Rate Law: k16*s127
k12 = 2.0E-5Reaction: s135 => s161; s132, Rate Law: k12*s135
kf15 = 0.0025; kr15 = 0.0Reaction: s161 => s164, Rate Law: kf15*s161-kr15*s164

States:

NameDescription
s150[Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator]
s124sa12_degraded
s135[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha]
s159[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha]
s121[NF-kappa-B inhibitor alpha]
s153sa96_degraded
s122sa13_degraded
s128[Tumor necrosis factor alpha-induced protein 3]
s132[Inhibitor of nuclear factor kappa-B kinase subunit beta; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B essential modulator]
s167[NF-kappa-B inhibitor alpha]
s160[NF-kappa-B inhibitor alpha]
s127[Tnfaip3-201]
s129sa444_degraded
s152[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha; Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator]
s134sa20_degraded
s154sa97_degraded
s130[Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator]
s164[Nuclear factor NF-kappa-B p105 subunit]
s131sa19_degraded
s139[NF-kappa-B inhibitor alpha; Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator]
s133[Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator]
s161[Nuclear factor NF-kappa-B p105 subunit]
s126[Tumor necrosis factor alpha-induced protein 3]
s125[Nfkbia-201]

Radulescu2008_NFkB_hierarchy_M_14_25_33: MODEL7743444866v0.0.1

# NFkB model M(14,25,33) This is a model of NFkB pathway functioning from hierarchy of models of decreasing complexity,…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Radulescu2008_NFkB_hierarchy_M_14_30_41: MODEL7743528808v0.0.1

# NFkB model M(14,30,41) This is a model of NFkB pathway functioning from hierarchy of models of decreasing complexity,…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Radulescu2008_NFkB_hierarchy_M_24_45_62: MODEL7743608569v0.0.1

# NFkB model M(24,45,62) This is a model of NFkB pathway functioning from hierarchy of models of decreasing complexity,…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Radulescu2008_NFkB_hierarchy_M_34_60_82: MODEL7743631122v0.0.1

# NFkB model M(34,60,82) This is a model of NFkB pathway functioning from hierarchy of models of decreasing complexity,…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Radulescu2008_NFkB_hierarchy_M_39_65_90: BIOMD0000000227v0.0.1

# NFkB model M(39,65,90) - most complex model This is a model of NFkB pathway functioning from hierarchy of models of d…

Details

BACKGROUND: Cellular processes such as metabolism, decision making in development and differentiation, signalling, etc., can be modeled as large networks of biochemical reactions. In order to understand the functioning of these systems, there is a strong need for general model reduction techniques allowing to simplify models without loosing their main properties. In systems biology we also need to compare models or to couple them as parts of larger models. In these situations reduction to a common level of complexity is needed. RESULTS: We propose a systematic treatment of model reduction of multiscale biochemical networks. First, we consider linear kinetic models, which appear as "pseudo-monomolecular" subsystems of multiscale nonlinear reaction networks. For such linear models, we propose a reduction algorithm which is based on a generalized theory of the limiting step that we have developed in 1. Second, for non-linear systems we develop an algorithm based on dominant solutions of quasi-stationarity equations. For oscillating systems, quasi-stationarity and averaging are combined to eliminate time scales much faster and much slower than the period of the oscillations. In all cases, we obtain robust simplifications and also identify the critical parameters of the model. The methods are demonstrated for simple examples and for a more complex model of NF-kappaB pathway. CONCLUSION: Our approach allows critical parameter identification and produces hierarchies of models. Hierarchical modeling is important in "middle-out" approaches when there is need to zoom in and out several levels of complexity. Critical parameter identification is an important issue in systems biology with potential applications to biological control and therapeutics. Our approach also deals naturally with the presence of multiple time scales, which is a general property of systems biology models. link: http://identifiers.org/pubmed/18854041

Parameters:

NameDescription
k8 = 0.1Reaction: s191 => s132, Rate Law: k8*s191
k37 = 5.0E-5Reaction: s113 => s112, Rate Law: k37*s113
kf59 = 0.0038; kr59 = 8.0E-13Reaction: s213 => s205 + s192, Rate Law: kf59*s213-kr59*s192*s205
k10 = 0.1Reaction: s189 => s190 + s132, Rate Law: k10*s189
k53 = 2.0E-4Reaction: s190 => s156, Rate Law: k53*s190
kr23 = 5.0E-4; kf23 = 0.001Reaction: s123 => s93, Rate Law: kf23*s123-kr23*s93
k45 = 0.0053Reaction: s117 => s119 + s117, Rate Law: k45*s117
kf52 = 0.003; kr52 = 0.001Reaction: s114 + s119 => s190, Rate Law: kf52*s114*s119-kr52*s190
k7 = 0.24Reaction: s123 + s132 => s191, Rate Law: k7*s132*s123
k39 = 1.3E-4Reaction: s110 => s114, Rate Law: k39*s110
kf57 = 18.4; kr57 = 0.055Reaction: s93 + s212 => s213, Rate Law: kf57*s93*s212-kr57*s213
kr56 = 4.8E-4; kf56 = 0.62Reaction: s195 + s206 => s214, Rate Law: kf56*s195*s206-kr56*s214
kf66 = 18.4; kr66 = 0.055Reaction: s200 + s93 => s201, Rate Law: kf66*s93*s200-kr66*s201
k43 = 0.1; k42 = 5.0E-4Reaction: s160 => s165; s193, s194, Rate Law: k42*s193+k43*s194
k72 = 2.0E-4Reaction: s192 => s158, Rate Law: k72*s192
k5 = 0.0015; k4 = 0.1Reaction: s132 => s130; s128, Rate Law: k5*s132+k4*s132*s128
kf64 = 0.62; kr64 = 4.8E-4Reaction: s199 + s195 => s200, Rate Law: kf64*s195*s199-kr64*s200
k47 = 6.4E-5Reaction: s119 => s120, Rate Law: k47*s119
kr14 = 0.0; kf14 = 0.5Reaction: s195 + s93 => s192, Rate Law: kf14*s195*s93-kr14*s192
k22 = 0.5Reaction: s178 => s123 + s178, Rate Law: k22*s178
k18 = 3.0E-4Reaction: s128 => s154, Rate Law: k18*s128
k61 = 0.06; k49 = 5.0E-4; k50 = 0.02; k62 = 0.6Reaction: s209 => s185; s214, s212, s205, s206, Rate Law: k49*s205+k50*s206+k62*s214+k61*s212
k12 = 2.0E-5Reaction: s188 => s190, Rate Law: k12*s188
k33 = 5.0E-4; k70 = 0.06; k69 = 0.006; k34 = 0.1Reaction: s170 => s173; s200, s199, s198, s196, Rate Law: k33*s198+k34*s199+k69*s196+k70*s200
k36 = 0.0041Reaction: s113 => s110 + s113, Rate Law: k36*s113
k9 = 1.2Reaction: s132 + s188 => s189, Rate Law: k9*s132*s188
k51 = 0.025Reaction: s185 => s178, Rate Law: k51*s185
kf28 = 0.01; kr28 = 0.0Reaction: s192 => s188, Rate Law: kf28*s192-kr28*s188
k19 = 0.0; k20 = 5.0E-7Reaction: s126 => s127; s195, Rate Law: k19+k20*s195
kr13 = 0.0; kf13 = 0.5Reaction: s123 + s190 => s188, Rate Law: kf13*s190*s123-kr13*s188
k6 = 1.25E-4Reaction: s132 => s134, Rate Law: k6*s132
k21 = 1.0E-4Reaction: s123 => s122, Rate Law: k21*s123
k1 = 0.0Reaction: s133 => s132, Rate Law: k1*s133
k71 = 2.0E-4Reaction: s188 => s157, Rate Law: k71*s188
k46 = 5.0E-5Reaction: s117 => s118, Rate Law: k46*s117
k38 = 6.0E-5Reaction: s110 => s109, Rate Law: k38*s110
k27 = 4.0E-4Reaction: s178 => s124, Rate Law: k27*s178
k11 = 1.25E-4Reaction: s130 => s129, Rate Law: k11*s130
kf32 = 10.0; kr32 = 1.0E-4Reaction: s22 + s198 => s199, Rate Law: kf32*s198*s22-kr32*s199
kr58 = 0.055; kf58 = 18.4Reaction: s93 + s214 => s215, Rate Law: kf58*s93*s214-kr58*s215
kr68 = 8.0E-13; kf68 = 0.0038Reaction: s201 => s199 + s192, Rate Law: kf68*s201-kr68*s192*s199
kr65 = 0.055; kf65 = 18.4Reaction: s196 + s93 => s197, Rate Law: kf65*s93*s196-kr65*s197
kr67 = 8.0E-13; kf67 = 0.0038Reaction: s197 => s198 + s192, Rate Law: kf67*s197-kr67*s192*s198
k17 = 4.0E-4Reaction: s127 => s153, Rate Law: k17*s127
k40 = 6.4E-5Reaction: s114 => s111, Rate Law: k40*s114
k2 = 1.25E-4Reaction: s133 => s131, Rate Law: k2*s133
kr55 = 4.8E-4; kf55 = 0.62Reaction: s195 + s205 => s212, Rate Law: kf55*s195*s205-kr55*s212
kr63 = 4.8E-4; kf63 = 0.62Reaction: s195 + s198 => s196, Rate Law: kf63*s195*s198-kr63*s196
kr41 = 1.0E-4; kf41 = 10.0Reaction: s193 + s36 => s194, Rate Law: kf41*s36*s193-kr41*s194
k44 = 0.016Reaction: s165 => s117, Rate Law: k44*s165
kr48 = 1.0E-4; kf48 = 10.0Reaction: s65 + s205 => s206, Rate Law: kf48*s65*s205-kr48*s206
k16 = 0.5Reaction: s127 => s128 + s127, Rate Law: k16*s127
k3 = 1.0E-5Reaction: s150 => s133, Rate Law: k3
k35 = 0.01Reaction: s173 => s113, Rate Law: k35*s173
k54 = 2.0E-4Reaction: s195 => s108, Rate Law: k54*s195

States:

NameDescription
s113[Nfkb1-201]
s213PromIkBa:RNAP3:p50p65:IkBa
s122sa13_degraded
s128[Tumor necrosis factor alpha-induced protein 3]
s36[Transcription factor RelB]
s197Promp105:RNAP1:p50p65:IkBa
s178[Nfkbia-201]
s198Promp105:RNAP
s189[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha; Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Transcription factor p65]
s160InactivePRaseonp65
s127[Tnfaip3-201]
s134sa20_degraded
s129sa444_degraded
s192[Nuclear factor NF-kappa-B p105 subunit; Transcription factor p65; NF-kappa-B inhibitor alpha]
s119[Transcription factor p65]
s118sa8_degraded
s205PromIkBa:RNAP3
s131sa19_degraded
s133[Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator]
s126[Tumor necrosis factor alpha-induced protein 3]
s193Promp65:RNAP2
s111sa438_degraded
s112sa3_degraded
s124sa12_degraded
s156csa21_degraded
s109sa4_degraded
s214IkBa:RNAP3:FTAz:p50p65
s93[NF-kappa-B inhibitor alpha]
s117[Rela-201]
s120sa9_degraded
s165ActivePRaseonp65
s132[Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator]
s206PromIkBa:RNAP3:FTAz
s22[Transcription factor RelB]
s185ActivePRaseonIkB_alpha
s199Promp105:RNAP:FTAX
s170InactivePRaseonp105
s130[Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator]
s215PromIkBa:RNAP3:FTAz:p50p65:IkB_alpha
s195[Nuclear factor NF-kappa-B p105 subunit; Transcription factor p65]
s188[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha; Transcription factor p65]
s108csa17_degraded
s123[NF-kappa-B inhibitor alpha]
s201Promp105:RNAP1:FTAx:p50p65:IkBa
s114[Nuclear factor NF-kappa-B p105 subunit]
s173ActivePRaseonp105
s190[Nuclear factor NF-kappa-B p105 subunit; Transcription factor p65]
s65[Transcription factor RelB]
s110[Nuclear factor NF-kappa-B p105 subunit]
s200Promp105:RNAP1:FTAx:p50p65
s158csa9_degraded

Raghunathan2009 - Genome-scale metabolic network of Salmonella typhimurium (iRR1083): MODEL1507180058v0.0.1

Raghunathan2009 - Genome-scale metabolic network of Salmonella typhimurium (iRR1083)This model is described in the artic…

Details

BACKGROUND: Infections with Salmonella cause significant morbidity and mortality worldwide. Replication of Salmonella typhimurium inside its host cell is a model system for studying the pathogenesis of intracellular bacterial infections. Genome-scale modeling of bacterial metabolic networks provides a powerful tool to identify and analyze pathways required for successful intracellular replication during host-pathogen interaction. RESULTS: We have developed and validated a genome-scale metabolic network of Salmonella typhimurium LT2 (iRR1083). This model accounts for 1,083 genes that encode proteins catalyzing 1,087 unique metabolic and transport reactions in the bacterium. We employed flux balance analysis and in silico gene essentiality analysis to investigate growth under a wide range of conditions that mimic in vitro and host cell environments. Gene expression profiling of S. typhimurium isolated from macrophage cell lines was used to constrain the model to predict metabolic pathways that are likely to be operational during infection. CONCLUSION: Our analysis suggests that there is a robust minimal set of metabolic pathways that is required for successful replication of Salmonella inside the host cell. This model also serves as platform for the integration of high-throughput data. Its computational power allows identification of networked metabolic pathways and generation of hypotheses about metabolism during infection, which might be used for the rational design of novel antibiotics or vaccine strains. link: http://identifiers.org/pubmed/19356237

Raghunathan2010 - Genome-scale metabolic network of Francisella tularensis (iRS605): MODEL1507180003v0.0.1

Raghunathan2010 - Genome-scale metabolic network of Francisella tularensis (iRS605)This model is described in the articl…

Details

BACKGROUND: Francisella tularensis is a prototypic example of a pathogen for which few experimental datasets exist, but for which copious high-throughout data are becoming available because of its re-emerging significance as biothreat agent. The virulence of Francisella tularensis depends on its growth capabilities within a defined environmental niche of the host cell. RESULTS: We reconstructed the metabolism of Francisella as a stoichiometric matrix. This systems biology approach demonstrated that changes in carbohydrate utilization and amino acid metabolism play a pivotal role in growth, acid resistance, and energy homeostasis during infection with Francisella. We also show how varying the expression of certain metabolic genes in different environments efficiently controls the metabolic capacity of F. tularensis. Selective gene-expression analysis showed modulation of sugar catabolism by switching from oxidative metabolism (TCA cycle) in the initial stages of infection to fatty acid oxidation and gluconeogenesis later on. Computational analysis with constraints derived from experimental data revealed a limited set of metabolic genes that are operational during infection. CONCLUSIONS: This integrated systems approach provides an important tool to understand the pathogenesis of an ill-characterized biothreat agent and to identify potential novel drug targets when rapid target identification is required should such microbes be intentionally released or become epidemic. link: http://identifiers.org/pubmed/20731870

Raia2010 - IL13 Signalling MedB1: BIOMD0000000313v0.0.1

This is the model of IL13 induced signalling in MedB-1 cell described in the article: **Dynamic Mathematical Modeling o…

Details

Primary mediastinal B-cell lymphoma (PMBL) and classical Hodgkin lymphoma (cHL) share a frequent constitutive activation of JAK (Janus kinase)/STAT signaling pathway. Because of complex, nonlinear relations within the pathway, key dynamic properties remained to be identified to predict possible strategies for intervention. We report the development of dynamic pathway models based on quantitative data collected on signaling components of JAK/STAT pathway in two lymphoma-derived cell lines, MedB-1 and L1236, representative of PMBL and cHL, respectively. We show that the amounts of STAT5 and STAT6 are higher whereas those of SHP1 are lower in the two lymphoma cell lines than in normal B cells. Distinctively, L1236 cells harbor more JAK2 and less SHP1 molecules per cell than MedB-1 or control cells. In both lymphoma cell lines, we observe interleukin-13 (IL13)-induced activation of IL4 receptor α, JAK2, and STAT5, but not of STAT6. Genome-wide, 11 early and 16 sustained genes are upregulated by IL13 in both lymphoma cell lines. Specifically, the known STAT-inducible negative regulators CISH and SOCS3 are upregulated within 2 hours in MedB-1 but not in L1236 cells. On the basis of this detailed quantitative information, we established two mathematical models, MedB-1 and L1236 model, able to describe the respective experimental data. Most of the model parameters are identifiable and therefore the models are predictive. Sensitivity analysis of the model identifies six possible therapeutic targets able to reduce gene expression levels in L1236 cells and three in MedB-1. We experimentally confirm reduction in target gene expression in response to inhibition of STAT5 phosphorylation, thereby validating one of the predicted targets. link: http://identifiers.org/pubmed/21127196

Parameters:

NameDescription
pSTAT5_dephosphorylation = 3.43392E-4Reaction: pSTAT5 => STAT5; SHP1, Rate Law: pSTAT5_dephosphorylation*pSTAT5*SHP1*cell
Kon_IL13Rec = 0.00341992Reaction: Rec => IL13_Rec; IL13, Rate Law: Kon_IL13Rec*IL13*Rec*cell
STAT5_phosphorylation = 0.0382596Reaction: STAT5 => pSTAT5; pJAK2, Rate Law: STAT5_phosphorylation*STAT5*pJAK2*cell
SOCS3_degradation = 0.0429186Reaction: SOCS3 =>, Rate Law: SOCS3_degradation*SOCS3*cell
DecoyR_binding = 1.24391E-4Reaction: DecoyR => IL13_DecoyR; IL13, Rate Law: DecoyR_binding*IL13*DecoyR*cell
Rec_intern = 0.103346Reaction: Rec => Rec_i, Rate Law: Rec_intern*Rec*cell
CD274mRNA_production = 8.21752E-5Reaction: => CD274mRNA; pSTAT5, Rate Law: pSTAT5*CD274mRNA_production*cell
pJAK2_dephosphorylation = 6.21906E-4Reaction: pJAK2 => JAK2; SHP1, Rate Law: pJAK2_dephosphorylation*pJAK2*SHP1*cell
pRec_degradation = 0.172928Reaction: p_IL13_Rec_i =>, Rate Law: pRec_degradation*p_IL13_Rec_i*cell
SOCS3_accumulation = 3.70803; SOCS3_translation = 11.9086Reaction: => SOCS3; SOCS3mRNA, Rate Law: SOCS3mRNA*SOCS3_translation/(SOCS3_accumulation+SOCS3mRNA)*cell
SOCS3mRNA_production = 0.00215826Reaction: => SOCS3mRNA; pSTAT5, Rate Law: pSTAT5*SOCS3mRNA_production*cell
pRec_intern = 0.15254Reaction: p_IL13_Rec => p_IL13_Rec_i, Rate Law: pRec_intern*p_IL13_Rec*cell
Rec_recycle = 0.00135598Reaction: Rec_i => Rec, Rate Law: Rec_recycle*Rec_i*cell
IL13stimulation = 1.0 ng_per_mlReaction: IL13 = 2.265*IL13stimulation, Rate Law: missing
Rec_phosphorylation = 999.631Reaction: IL13_Rec => p_IL13_Rec; pJAK2, Rate Law: Rec_phosphorylation*IL13_Rec*pJAK2*cell
JAK2_phosphorylation = 0.157057; JAK2_p_inhibition = 0.0168268Reaction: JAK2 => pJAK2; IL13_Rec, SOCS3, Rate Law: JAK2_phosphorylation*IL13_Rec*JAK2/(1+JAK2_p_inhibition*SOCS3)*cell

States:

NameDescription
p IL13 Rec[MOD:00048; Interleukin-13; Interleukin-4 receptor subunit alpha; Interleukin-13 receptor subunit alpha-1; Phosphoprotein; Non-receptor tyrosine-protein kinase TYK2; interleukin-4 receptor complex; urn:miriam:mod:MOD%3A00048]
SOCS3[Suppressor of cytokine signaling 3]
IL13 DecoyR[Interleukin-13; Interleukin-13 receptor subunit alpha-2]
SOCS3mRNA[messenger RNA; RNA; Suppressor of cytokine signaling 3]
pSTAT5[MOD:00048; Signal transducer and activator of transcription 5B; urn:miriam:mod:MOD%3A00048; Signal transducer and activator of transcription 5A; Phosphoprotein]
IL13[Interleukin-13; interleukin-13 receptor binding]
Rec i[Non-receptor tyrosine-protein kinase TYK2; interleukin-4 receptor complex; Interleukin-13 receptor subunit alpha-1; receptor internalization; Interleukin-4 receptor subunit alpha]
CD274mRNA[messenger RNA; RNA; T-cell surface glycoprotein CD3 zeta chain]
STAT5[Signal transducer and activator of transcription 5A; Signal transducer and activator of transcription 5B]
p IL13 Rec i[MOD:00048; Interleukin-13; Non-receptor tyrosine-protein kinase TYK2; urn:miriam:mod:MOD%3A00048; interleukin-4 receptor complex; Interleukin-4 receptor subunit alpha; Interleukin-13 receptor subunit alpha-1; receptor internalization]
IL13 Rec[Interleukin-13; Interleukin-13 receptor subunit alpha-1; Interleukin-4 receptor subunit alpha; Non-receptor tyrosine-protein kinase TYK2; interleukin-4 receptor complex]
DecoyR[Interleukin-13 receptor subunit alpha-2]
Rec[interleukin-4 receptor complex; Non-receptor tyrosine-protein kinase TYK2; Interleukin-4 receptor subunit alpha; Interleukin-13 receptor subunit alpha-1; interleukin-13 binding]
pJAK2[MOD:00048; Tyrosine-protein kinase JAK2; Phosphoprotein; urn:miriam:mod:MOD%3A00048]
JAK2[Tyrosine-protein kinase JAK2]

Raia2011 - IL13 L1236: BIOMD0000000314v0.0.1

This is the model of IL13 induced signalling in L1236 cells described in the article: **Dynamic Mathematical Modeling…

Details

Primary mediastinal B-cell lymphoma (PMBL) and classical Hodgkin lymphoma (cHL) share a frequent constitutive activation of JAK (Janus kinase)/STAT signaling pathway. Because of complex, nonlinear relations within the pathway, key dynamic properties remained to be identified to predict possible strategies for intervention. We report the development of dynamic pathway models based on quantitative data collected on signaling components of JAK/STAT pathway in two lymphoma-derived cell lines, MedB-1 and L1236, representative of PMBL and cHL, respectively. We show that the amounts of STAT5 and STAT6 are higher whereas those of SHP1 are lower in the two lymphoma cell lines than in normal B cells. Distinctively, L1236 cells harbor more JAK2 and less SHP1 molecules per cell than MedB-1 or control cells. In both lymphoma cell lines, we observe interleukin-13 (IL13)-induced activation of IL4 receptor α, JAK2, and STAT5, but not of STAT6. Genome-wide, 11 early and 16 sustained genes are upregulated by IL13 in both lymphoma cell lines. Specifically, the known STAT-inducible negative regulators CISH and SOCS3 are upregulated within 2 hours in MedB-1 but not in L1236 cells. On the basis of this detailed quantitative information, we established two mathematical models, MedB-1 and L1236 model, able to describe the respective experimental data. Most of the model parameters are identifiable and therefore the models are predictive. Sensitivity analysis of the model identifies six possible therapeutic targets able to reduce gene expression levels in L1236 cells and three in MedB-1. We experimentally confirm reduction in target gene expression in response to inhibition of STAT5 phosphorylation, thereby validating one of the predicted targets. link: http://identifiers.org/pubmed/21127196

Parameters:

NameDescription
Rec_phosphorylation = 9.07541Reaction: IL13_Rec => p_IL13_Rec; pJAK2, Rate Law: Rec_phosphorylation*IL13_Rec*pJAK2*cell
pSTAT5_dephosphorylation = 0.0116389Reaction: pSTAT5 => STAT5; SHP1, Rate Law: pSTAT5_dephosphorylation*pSTAT5*SHP1*cell
CD274mRNA_production = 0.0115928Reaction: => CD274mRNA; pSTAT5, Rate Law: pSTAT5*CD274mRNA_production*cell
Kon_IL13Rec = 0.00174087Reaction: Rec => IL13_Rec; IL13, Rate Law: Kon_IL13Rec*IL13*Rec*cell
pRec_degradation = 0.417538Reaction: p_IL13_Rec_i =>, Rate Law: pRec_degradation*p_IL13_Rec_i*cell
pJAK2_dephosphorylation = 0.0981611Reaction: pJAK2 => JAK2; SHP1, Rate Law: pJAK2_dephosphorylation*pJAK2*SHP1*cell
JAK2_phosphorylation = 0.300019Reaction: JAK2 => pJAK2; IL13_Rec, Rate Law: JAK2_phosphorylation*JAK2*IL13_Rec*cell
Rec_recycle = 0.0039243Reaction: Rec_i => Rec, Rate Law: Rec_recycle*Rec_i*cell
pRec_intern = 0.324132Reaction: p_IL13_Rec => p_IL13_Rec_i, Rate Law: pRec_intern*p_IL13_Rec*cell
IL13stimulation = 1.0 ng_per_mlReaction: IL13 = 3.776*IL13stimulation, Rate Law: missing
STAT5_phosphorylation = 0.00426767Reaction: STAT5 => pSTAT5; pJAK2, Rate Law: STAT5_phosphorylation*STAT5*pJAK2*cell
Rec_intern = 0.259686Reaction: Rec => Rec_i, Rate Law: Rec_intern*Rec*cell

States:

NameDescription
p IL13 Rec[Non-receptor tyrosine-protein kinase TYK2; Interleukin-13; interleukin-4 receptor complex; urn:miriam:mod:MOD%3A00048; Phosphoprotein; Interleukin-13 receptor subunit alpha-1; Interleukin-4 receptor subunit alpha]
pSTAT5[Signal transducer and activator of transcription 5A; Signal transducer and activator of transcription 5B; Phosphoprotein; urn:miriam:mod:MOD%3A00048]
IL13[Interleukin-13; interleukin-13 receptor binding]
Rec i[Non-receptor tyrosine-protein kinase TYK2; interleukin-4 receptor complex; Interleukin-4 receptor subunit alpha; Interleukin-13 receptor subunit alpha-1; receptor internalization]
CD274mRNA[messenger RNA; RNA; T-cell surface glycoprotein CD3 zeta chain]
STAT5[Signal transducer and activator of transcription 5B; Signal transducer and activator of transcription 5A]
p IL13 Rec i[urn:miriam:mod:MOD%3A00048; interleukin-4 receptor complex; Interleukin-13 receptor subunit alpha-1; Interleukin-4 receptor subunit alpha; Non-receptor tyrosine-protein kinase TYK2; Interleukin-13; receptor internalization]
IL13 Rec[Non-receptor tyrosine-protein kinase TYK2; Interleukin-13; interleukin-4 receptor complex; Interleukin-4 receptor subunit alpha; Interleukin-13 receptor subunit alpha-1]
Rec[interleukin-4 receptor complex; Non-receptor tyrosine-protein kinase TYK2; Interleukin-13 receptor subunit alpha-1; Interleukin-4 receptor subunit alpha; interleukin-13 binding]
pJAK2[Tyrosine-protein kinase JAK2; Phosphoprotein; urn:miriam:mod:MOD%3A00048]
JAK2[Tyrosine-protein kinase JAK2]

Ralser2007_Carbohydrate_Rerouting_ROS: BIOMD0000000247v0.0.1

This is the model with unfitted parameters described in the article **Dynamic rerouting of the carbohydrate flux is k…

Details

Eukaryotic cells have evolved various response mechanisms to counteract the deleterious consequences of oxidative stress. Among these processes, metabolic alterations seem to play an important role.We recently discovered that yeast cells with reduced activity of the key glycolytic enzyme triosephosphate isomerase exhibit an increased resistance to the thiol-oxidizing reagent diamide. Here we show that this phenotype is conserved in Caenorhabditis elegans and that the underlying mechanism is based on a redirection of the metabolic flux from glycolysis to the pentose phosphate pathway, altering the redox equilibrium of the cytoplasmic NADP(H) pool. Remarkably, another key glycolytic enzyme, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), is known to be inactivated in response to various oxidant treatments, and we show that this provokes a similar redirection of the metabolic flux.The naturally occurring inactivation of GAPDH functions as a metabolic switch for rerouting the carbohydrate flux to counteract oxidative stress. As a consequence, altering the homoeostasis of cytoplasmic metabolites is a fundamental mechanism for balancing the redox state of eukaryotic cells under stress conditions. link: http://identifiers.org/pubmed/18154684

Parameters:

NameDescription
VmALD=322.258 mMpermin; KeqTPI=0.045 dimensionless; KeqALD=0.069 dimensionless; KmALDDHAP=2.4 mM; KmALDGAPi=10.0 mM; KmALDF16P=0.3 mM; KmALDGAP=2.0 mMReaction: F16P => DHAP + GA3P, Rate Law: cytoplasm*VmALD*F16P/KmALDF16P*(1-DHAP*GA3P/(F16P*KeqALD))/(1+F16P/KmALDF16P+DHAP/KmALDDHAP+GA3P/KmALDGAP+F16P*GA3P/(KmALDF16P*KmALDGAPi)+DHAP*GA3P/(KmALDDHAP*KmALDGAP))
KmEry4P=0.09 mM; KmGA3P=2.1 mM; VmTransk2f=3.2 mMpermin; KmXyl5P=0.16 mM; KmF6P=1.1 mM; VmTransk2r=43.0 mMperminReaction: Xyl5P + Erythrose4P => GA3P + F6P, Rate Law: cytoplasm*(VmTransk2f*Erythrose4P*Xyl5P/(KmEry4P*KmXyl5P)-VmTransk2r*F6P*GA3P/(KmF6P*KmGA3P))/((1+Xyl5P/KmXyl5P+GA3P/KmGA3P)*(1+Erythrose4P/KmEry4P+F6P/KmF6P))
VmG6PDH=4.0 mMpermin; KmG6P=0.04 mM; KmNADP=0.02 mM; KiNADPH=0.017 mMReaction: G6P + NADP => D6PGluconoLactone + NADPH; NADPH, Rate Law: cytoplasm*VmG6PDH*G6P*NADP/(KmG6P*KmNADP)/((1+G6P/KmG6P+NADPH/KiNADPH)*(1+NADP/KmNADP))
VmPPIf=3458.0 mMpermin; KmRibu5P=1.6 mM; KmRibo5P=1.6 mM; VmPPIr=3458.0 mMperminReaction: Ribulose5P => Ribose5P, Rate Law: cytoplasm*(VmPPIf*Ribulose5P/KmRibu5P-VmPPIr*Ribose5P/KmRibo5P)/(1+Ribulose5P/KmRibu5P+Ribose5P/KmRibo5P)
KeqENO=6.7 dimensionless; KmENOP2G=0.04 mM; KmENOPEP=0.5 mM; VmENO=365.806 mMperminReaction: P2G => PEP, Rate Law: cytoplasm*VmENO/KmENOP2G*(P2G-PEP/KeqENO)/(1+P2G/KmENOP2G+PEP/KmENOPEP)
KeqAK=0.45 dimensionless; KATPASE=39.5 permin; SUMAXP = 4.1Reaction: P => X, Rate Law: cytoplasm*KATPASE*(((P-4*KeqAK*P)-SUMAXP)+(((P^2-4*KeqAK*P^2)-2*P*SUMAXP)+8*KeqAK*P*SUMAXP+SUMAXP^2)^0.5)/(2-8*KeqAK)
KmPGMP3G=1.2 mM; KeqPGM=0.19 dimensionless; VmPGM=2525.81 mMpermin; KmPGMP2G=0.08 mMReaction: P3G => P2G, Rate Law: cytoplasm*VmPGM/KmPGMP3G*(P3G-P2G/KeqPGM)/(1+P3G/KmPGMP3G+P2G/KmPGMP2G)
VmGAPDHr=6549.68 mMpermin; VmGAPDHf=1184.52 mMpermin; k_rel_GAPDH = 1.0 dimensionless; KeqTPI=0.045 dimensionless; KmGAPDHNAD=0.09 mM; KeqGAPDH=0.005 dimensionless; KmGAPDHBPG=0.0098 mM; KmGAPDHGAP=0.21 mM; KmGAPDHNADH=0.06 mMReaction: GA3P + NAD => BPG + NADH, Rate Law: cytoplasm*k_rel_GAPDH*VmGAPDHf*GA3P*NAD/(KmGAPDHGAP*KmGAPDHNAD)*(1-BPG*NADH/(GA3P*NAD*KeqGAPDH))/((1+GA3P/KmGAPDHGAP+BPG/KmGAPDHBPG)*(1+NAD/KmGAPDHNAD+NADH/KmGAPDHNADH))
KmADHNAD=0.17 mM; KiADHETOH=90.0 mM; KiADHNADH=0.031 mM; KiADHACE=1.1 mM; KmADHETOH=17.0 mM; KeqADH=6.9E-5 dimensionless; KmADHNADH=0.11 mM; KiADHNAD=0.92 mM; VmADH=810.0 mMpermin; KmADHACE=1.11 mMReaction: ACE + NADH => ETOH + NAD, Rate Law: cytoplasm*(-VmADH/(KiADHNAD*KmADHETOH)*(NAD*ETOH-NADH*ACE/KeqADH)/(1+NAD/KiADHNAD+KmADHNAD*ETOH/(KiADHNAD*KmADHETOH)+KmADHNADH*ACE/(KiADHNADH*KmADHACE)+NADH/KiADHNADH+NAD*ETOH/(KiADHNAD*KmADHETOH)+KmADHNADH*NAD*ACE/(KiADHNAD*KiADHNADH*KmADHACE)+KmADHNAD*ETOH*NADH/(KiADHNAD*KmADHETOH*KiADHNADH)+NADH*ACE/(KiADHNADH*KmADHACE)+NAD*ETOH*ACE/(KiADHNAD*KmADHETOH*KiADHACE)+ETOH*NADH*ACE/(KiADHETOH*KiADHNADH*KmADHACE)))
Km6PGL=0.8 mM; Vm6PGL=4.0 mMperminReaction: D6PGluconoLactone => D6PGluconate, Rate Law: cytoplasm*Vm6PGL*D6PGluconoLactone/(Km6PGL+D6PGluconoLactone)
KSUCC=21.4 perminReaction: ACE + NAD => NADH + SUCC, Rate Law: cytoplasm*KSUCC*ACE
kNADPH=2.0 perminReaction: NADPH => NADP, Rate Law: cytoplasm*kNADPH*NADPH
CPFKATP=3.0 dimensionless; CPFKF16BP=0.397 dimensionless; CPFKF26BP=0.0174 dimensionless; VmPFK=182.903 mMpermin; L0=0.66 dimensionless; SUMAXP = 4.1; KmPFKF6P=0.1 mM; KeqAK=0.45 dimensionless; KPFKF26BP=6.82E-4 mM; CPFKAMP=0.0845 dimensionless; KPFKAMP=0.0995 mM; CiPFKATP=100.0 dimensionless; KPFKF16BP=0.111 mM; KmPFKATP=0.71 mM; gR=5.12 dimensionless; KiPFKATP=0.65 mMReaction: F6P + P => F16P; F26BP, Rate Law: cytoplasm*gR*VmPFK*F6P*((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)*(1+F6P/KmPFKF6P+((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((2-8*KeqAK)*KmPFKATP)+gR*F6P*((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((2-8*KeqAK)*KmPFKATP*KmPFKF6P))/((2-8*KeqAK)*KmPFKATP*KmPFKF6P*(L0*(1+CPFKF26BP*F26BP/KPFKF26BP+CPFKF16BP*F16P/KPFKF16BP)^2*(1+2*CPFKAMP*KeqAK*(SUMAXP-(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)^2/((-1+4*KeqAK)*KPFKAMP*(((SUMAXP-P)+4*KeqAK*P)-(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)))^2*(1+CiPFKATP*((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((2-8*KeqAK)*KiPFKATP))^2*(1+CPFKATP*((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((2-8*KeqAK)*KmPFKATP))^2/((1+F26BP/KPFKF26BP+F16P/KPFKF16BP)^2*(1+2*KeqAK*(SUMAXP-(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)^2/((-1+4*KeqAK)*KPFKAMP*(((SUMAXP-P)+4*KeqAK*P)-(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)))^2*(1+((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((2-8*KeqAK)*KiPFKATP))^2)+(1+F6P/KmPFKF6P+((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((2-8*KeqAK)*KmPFKATP)+gR*F6P*((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((2-8*KeqAK)*KmPFKATP*KmPFKF6P))^2))
VmTransaldf=55.0 mMpermin; KmF6P=0.32 mM; KmGA3P=0.22 mM; VmTransaldr=10.0 mMpermin; KmSeduhept=0.18 mM; KmEry4P=0.018 mMReaction: Seduhept7P + GA3P => F6P + Erythrose4P, Rate Law: cytoplasm*(VmTransaldf*GA3P*Seduhept7P/(KmGA3P*KmSeduhept)-VmTransaldr*F6P*Erythrose4P/(KmF6P*KmEry4P))/((1+GA3P/KmGA3P+F6P/KmF6P)*(1+Seduhept7P/KmSeduhept+Erythrose4P/KmEry4P))
KmG3PDHGLY=1.0 mM; KeqTPI=0.045 dimensionless; KeqG3PDH=4300.0 dimensionless; KmG3PDHDHAP=0.4 mM; KmG3PDHNADH=0.023 mM; KmG3PDHNAD=0.93 mM; VmG3PDH=70.15 mMperminReaction: DHAP + NADH => GLY + NAD, Rate Law: cytoplasm*VmG3PDH*((-GLY*NAD/KeqG3PDH)+NADH*DHAP/(1+KeqTPI))/(KmG3PDHDHAP*KmG3PDHNADH*(1+NAD/KmG3PDHNAD+NADH/KmG3PDHNADH)*(1+GLY/KmG3PDHGLY+DHAP/((1+KeqTPI)*KmG3PDHDHAP)))
VmGluDH=4.0 mMpermin; KmGluconate=0.02 mM; KmNADP=0.03 mM; KiNADPH=0.03 mMReaction: D6PGluconate + NADP => Ribulose5P + NADPH; NADPH, Rate Law: cytoplasm*VmGluDH*D6PGluconate*NADP/(KmGluconate*KmNADP)/((1+D6PGluconate/KmGluconate+NADPH/KiNADPH)*(1+NADP/KmNADP))
VmGA3P=555.0 mMpermin; KmDHAP=1.23 mM; k_rel_TPI = 1.0 dimensionless; KmGA3P=1.27 mM; VmDHAP=10900.0 mMperminReaction: GA3P => DHAP, Rate Law: cytoplasm*k_rel_TPI*(VmDHAP*GA3P/KmGA3P-VmGA3P*DHAP/KmDHAP)/(1+GA3P/KmGA3P+DHAP/KmDHAP)
KmXyl=1.5 mM; KmRibu5P=1.5 mM; VmR5PIr=1039.0 mMpermin; VmR5PIf=1039.0 mMperminReaction: Ribulose5P => Xyl5P, Rate Law: cytoplasm*(VmR5PIf*Ribulose5P/KmRibu5P-VmR5PIr*Xyl5P/KmXyl)/(1+Ribulose5P/KmRibu5P+Xyl5P/KmXyl)
VmGLT=97.264 mMpermin; KeqGLT=1.0 mM; KmGLTGLCo=1.1918 mM; KmGLTGLCi=1.1918 mMReaction: GLCo => GLCi, Rate Law: cytoplasm*VmGLT*(GLCo-GLCi/KeqGLT)/(KmGLTGLCo*(1+GLCo/KmGLTGLCo+GLCi/KmGLTGLCi+0.91*GLCo*GLCi/(KmGLTGLCi*KmGLTGLCo)))
KeqAK=0.45 dimensionless; KmPGKBPG=0.003 mM; KeqPGK=3200.0 dimensionless; KmPGKADP=0.2 mM; KmPGKATP=0.3 mM; VmPGK=1306.45 mMpermin; KmPGKP3G=0.53 mM; SUMAXP = 4.1Reaction: BPG => P3G + P, Rate Law: cytoplasm*VmPGK*(KeqPGK*BPG*(SUMAXP-(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/(1-4*KeqAK)-((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)*P3G/(2-8*KeqAK))/(KmPGKATP*KmPGKP3G*(1+(SUMAXP-(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((1-4*KeqAK)*KmPGKADP)+((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((2-8*KeqAK)*KmPGKATP))*(1+BPG/KmPGKBPG+P3G/KmPGKP3G))
VmPYK=1088.71 mMpermin; KeqAK=0.45 dimensionless; KmPYKADP=0.53 mM; KmPYKPEP=0.14 mM; KmPYKATP=1.5 mM; KeqPYK=6500.0 dimensionless; KmPYKPYR=21.0 mM; SUMAXP = 4.1Reaction: PEP => PYR + P, Rate Law: cytoplasm*VmPYK/(KmPYKPEP*KmPYKADP)*(PEP*(SUMAXP-(((P^2-4*KeqAK*P^2)-2*P*SUMAXP)+8*KeqAK*P*SUMAXP+SUMAXP^2)^0.5)/(1-4*KeqAK)-PYR*(((P-4*KeqAK*P)-SUMAXP)+(((P^2-4*KeqAK*P^2)-2*P*SUMAXP)+8*KeqAK*P*SUMAXP+SUMAXP^2)^0.5)/(2-8*KeqAK)/KeqPYK)/((1+PEP/KmPYKPEP+PYR/KmPYKPYR)*(1+(((P-4*KeqAK*P)-SUMAXP)+(((P^2-4*KeqAK*P^2)-2*P*SUMAXP)+8*KeqAK*P*SUMAXP+SUMAXP^2)^0.5)/(2-8*KeqAK)/KmPYKATP+(SUMAXP-(((P^2-4*KeqAK*P^2)-2*P*SUMAXP)+8*KeqAK*P*SUMAXP+SUMAXP^2)^0.5)/(1-4*KeqAK)/KmPYKADP))
VmPDC=174.194 mMpermin; KmPDCPYR=4.33 mM; nPDC=1.9 dimensionlessReaction: PYR => ACE + CO2, Rate Law: cytoplasm*VmPDC*PYR^nPDC/KmPDCPYR^nPDC/(1+PYR^nPDC/KmPDCPYR^nPDC)
KmPGIF6P=0.3 mM; KeqPGI=0.314 dimensionless; VmPGI=339.677 mMpermin; KmPGIG6P=1.4 mMReaction: G6P => F6P, Rate Law: cytoplasm*VmPGI/KmPGIG6P*(G6P-F6P/KeqPGI)/(1+G6P/KmPGIG6P+F6P/KmPGIF6P)
KmSeduhept=0.15 mM; KmXyl5P=0.15 mM; VmTransk1f=4.0 mMpermin; KmRibose5P=0.1 mM; KmGA3P=0.1 mM; VmTransk1r=2.0 mMperminReaction: Ribose5P + Xyl5P => GA3P + Seduhept7P, Rate Law: cytoplasm*(VmTransk1f*Ribose5P*Xyl5P/(KmRibose5P*KmXyl5P)-VmTransk1r*GA3P*Seduhept7P/(KmGA3P*KmSeduhept))/((1+Ribose5P/KmRibose5P+GA3P/KmGA3P)*(1+Xyl5P/KmXyl5P+Seduhept7P/KmSeduhept))
KeqAK=0.45 dimensionless; KmGLKADP=0.23 mM; KmGLKGLCi=0.08 mM; VmGLK=226.452 mMpermin; KmGLKG6P=30.0 mM; KeqGLK=3800.0 dimensionless; KmGLKATP=0.15 mM; SUMAXP = 4.1Reaction: GLCi + P => G6P, Rate Law: cytoplasm*VmGLK*((-G6P*(SUMAXP-(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((1-4*KeqAK)*KeqGLK))+GLCi*((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/(2-8*KeqAK))/(KmGLKATP*KmGLKGLCi*(1+G6P/KmGLKG6P+GLCi/KmGLKGLCi)*(1+(SUMAXP-(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((1-4*KeqAK)*KmGLKADP)+((((-SUMAXP)+P)-4*KeqAK*P)+(((SUMAXP^2-2*SUMAXP*P)+8*KeqAK*SUMAXP*P+P^2)-4*KeqAK*P^2)^0.5)/((2-8*KeqAK)*KmGLKATP)))

States:

NameDescription
Seduhept7P[sedoheptulose 7-phosphate]
P[ADP; ATP; ADP; ADP; ATP]
GLY[glycerol; Glycerol]
DHAP[dihydroxyacetone phosphate]
F16P[keto-D-fructose 1,6-bisphosphate; D-Fructose 1,6-bisphosphate]
NADPH[NADPH]
Xyl5P[D-xylulose 5-phosphate]
GLCi[glucose; C00293]
P2G[2-phospho-D-glyceric acid; 2-Phospho-D-glycerate]
Ribulose5P[D-ribulose 5-phosphate]
P3G[3-phospho-D-glyceric acid; 3-Phospho-D-glycerate]
GLCo[glucose; C00293]
Ribose5P[aldehydo-D-ribose 5-phosphate]
NADH[NADH; NADH]
PYR[pyruvate; Pyruvate]
XX
NADP[NADP(+)]
Erythrose4P[D-erythrose 4-phosphate]
GA3P[glyceraldehyde 3-phosphate]
SUCC[succinate(2-)]
BPG[3-phospho-D-glyceroyl dihydrogen phosphate; 3-Phospho-D-glyceroyl phosphate]
F6P[keto-D-fructose 6-phosphate; beta-D-Fructose 6-phosphate]
CO2[carbon dioxide; CO2]
G6P[alpha-D-glucose 6-phosphate; alpha-D-Glucose 6-phosphate]
D6PGluconoLactone[6-O-phosphono-D-glucono-1,5-lactone]
D6PGluconate[6-phospho-D-gluconate]
PEP[phosphoenolpyruvate; Phosphoenolpyruvate]
NAD[NAD(+); NAD+]
ETOH[ethanol; Ethanol]
ACE[acetaldehyde; Acetaldehyde]

Raman2006_MycolicAcid: MODEL8568434338v0.0.1

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

Details

Mycobacterium tuberculosis is the focus of several investigations for design of newer drugs, as tuberculosis remains a major epidemic despite the availability of several drugs and a vaccine. Mycobacteria owe many of their unique qualities to mycolic acids, which are known to be important for their growth, survival, and pathogenicity. Mycolic acid biosynthesis has therefore been the focus of a number of biochemical and genetic studies. It also turns out to be the pathway inhibited by front-line anti-tubercular drugs such as isoniazid and ethionamide. Recent years have seen the emergence of systems-based methodologies that can be used to study microbial metabolism. Here, we seek to apply insights from flux balance analyses of the mycolic acid pathway (MAP) for the identification of anti-tubercular drug targets. We present a comprehensive model of mycolic acid synthesis in the pathogen M. tuberculosis involving 197 metabolites participating in 219 reactions catalysed by 28 proteins. Flux balance analysis (FBA) has been performed on the MAP model, which has provided insights into the metabolic capabilities of the pathway. In silico systematic gene deletions and inhibition of InhA by isoniazid, studied here, provide clues about proteins essential for the pathway and hence lead to a rational identification of possible drug targets. Feasibility studies using sequence analysis of the M. tuberculosis H37Rv and human proteomes indicate that, apart from the known InhA, potential targets for anti-tubercular drug design are AccD3, Fas, FabH, Pks13, DesA1/2, and DesA3. Proteins identified as essential by FBA correlate well with those previously identified experimentally through transposon site hybridisation mutagenesis. This study demonstrates the application of FBA for rational identification of potential anti-tubercular drug targets, which can indeed be a general strategy in drug design. The targets, chosen based on the critical points in the pathway, form a ready shortlist for experimental testing. link: http://identifiers.org/pubmed/16261191

Rangel-Reyes2017 - Dendritic Immunotherapy Improvement for an Optimal Control Murine Model: MODEL1909160002v0.0.1

This is a mathematical model provides a platform for investigating the efficacy of dendritic cell vaccines during cancer…

Details

Therapeutic protocols in immunotherapy are usually proposed following the intuition and experience of the therapist. In order to deduce such protocols mathematical modeling, optimal control and simulations are used instead of the therapist's experience. Clinical efficacy of dendritic cell (DC) vaccines to cancer treatment is still unclear, since dendritic cells face several obstacles in the host environment, such as immunosuppression and poor transference to the lymph nodes reducing the vaccine effect. In view of that, we have created a mathematical murine model to measure the effects of dendritic cell injections admitting such obstacles. In addition, the model considers a therapy given by bolus injections of small duration as opposed to a continual dose. Doses timing defines the therapeutic protocols, which in turn are improved to minimize the tumor mass by an optimal control algorithm. We intend to supplement therapist's experience and intuition in the protocol's implementation. Experimental results made on mice infected with melanoma with and without therapy agree with the model. It is shown that the dendritic cells' percentage that manages to reach the lymph nodes has a crucial impact on the therapy outcome. This suggests that efforts in finding better methods to deliver DC vaccines should be pursued. link: http://identifiers.org/pubmed/28912828

Rantasalo2016 - Synthetic expresion modulator constitutive STF_B42: MODEL1510230002v0.0.1

Rantasalo2015-Synthetic_expresion_modulator_constitutiveSTF_B42 This model is part of a family of models describing a m…

Details

This work describes the development and characterization of a modular synthetic expression system that provides a broad range of adjustable and predictable expression levels in S. cerevisiae. The system works as a fixed-gain transcription amplifier, where the input signal is transferred via a synthetic transcription factor (sTF) onto a synthetic promoter, containing a defined core promoter, generating a transcription output signal. The system activation is based on the bacterial LexA-DNA-binding domain, a set of modified, modular LexA-binding sites and a selection of transcription activation domains. We show both experimentally and computationally that the tuning of the system is achieved through the selection of three separate modules, each of which enables an adjustable output signal: 1) the transcription-activation domain of the sTF, 2) the binding-site modules in the output promoter, and 3) the core promoter modules which define the transcription initiation site in the output promoter. The system has a novel bidirectional architecture that enables generation of compact, yet versatile expression modules for multiple genes with highly diversified expression levels ranging from negligible to very strong using one synthetic transcription factor. In contrast to most existing modular gene expression regulation systems, the present system is independent from externally added compounds. Furthermore, the established system was minimally affected by the several tested growth conditions. These features suggest that it can be highly useful in large scale biotechnology applications. link: http://identifiers.org/doi/10.1371/journal.pone.0148320

Rantasalo2016 - Synthetic expresion modulator constitutive STF_VP16: MODEL1510230001v0.0.1

Rantasalo2015-Synthetic_expresion_modulator_constitutiveSTF_VP16 This model is part of a family of models describing a…

Details

This work describes the development and characterization of a modular synthetic expression system that provides a broad range of adjustable and predictable expression levels in S. cerevisiae. The system works as a fixed-gain transcription amplifier, where the input signal is transferred via a synthetic transcription factor (sTF) onto a synthetic promoter, containing a defined core promoter, generating a transcription output signal. The system activation is based on the bacterial LexA-DNA-binding domain, a set of modified, modular LexA-binding sites and a selection of transcription activation domains. We show both experimentally and computationally that the tuning of the system is achieved through the selection of three separate modules, each of which enables an adjustable output signal: 1) the transcription-activation domain of the sTF, 2) the binding-site modules in the output promoter, and 3) the core promoter modules which define the transcription initiation site in the output promoter. The system has a novel bidirectional architecture that enables generation of compact, yet versatile expression modules for multiple genes with highly diversified expression levels ranging from negligible to very strong using one synthetic transcription factor. In contrast to most existing modular gene expression regulation systems, the present system is independent from externally added compounds. Furthermore, the established system was minimally affected by the several tested growth conditions. These features suggest that it can be highly useful in large scale biotechnology applications. link: http://identifiers.org/doi/10.1371/journal.pone.0148320

Rantasalo2016 - Synthetic expresion modulator constitutive STF_VP16_pBID2-ED corePromoter: MODEL1510230003v0.0.1

Rantasalo2015-Synthetic_expresion_modulator_constitutiveSTF_VP16_pBID2-EDcorePromoter This model is part of a family of…

Details

This work describes the development and characterization of a modular synthetic expression system that provides a broad range of adjustable and predictable expression levels in S. cerevisiae. The system works as a fixed-gain transcription amplifier, where the input signal is transferred via a synthetic transcription factor (sTF) onto a synthetic promoter, containing a defined core promoter, generating a transcription output signal. The system activation is based on the bacterial LexA-DNA-binding domain, a set of modified, modular LexA-binding sites and a selection of transcription activation domains. We show both experimentally and computationally that the tuning of the system is achieved through the selection of three separate modules, each of which enables an adjustable output signal: 1) the transcription-activation domain of the sTF, 2) the binding-site modules in the output promoter, and 3) the core promoter modules which define the transcription initiation site in the output promoter. The system has a novel bidirectional architecture that enables generation of compact, yet versatile expression modules for multiple genes with highly diversified expression levels ranging from negligible to very strong using one synthetic transcription factor. In contrast to most existing modular gene expression regulation systems, the present system is independent from externally added compounds. Furthermore, the established system was minimally affected by the several tested growth conditions. These features suggest that it can be highly useful in large scale biotechnology applications. link: http://identifiers.org/doi/10.1371/journal.pone.0148320

Rantasalo2016 - Synthetic expresion modulator induced STF_B42: MODEL1510230004v0.0.1

Rantasalo2015-Synthetic_expresion_modulator_inducedSTF_B42 This model is part of a family of models describing a modula…

Details

This work describes the development and characterization of a modular synthetic expression system that provides a broad range of adjustable and predictable expression levels in S. cerevisiae. The system works as a fixed-gain transcription amplifier, where the input signal is transferred via a synthetic transcription factor (sTF) onto a synthetic promoter, containing a defined core promoter, generating a transcription output signal. The system activation is based on the bacterial LexA-DNA-binding domain, a set of modified, modular LexA-binding sites and a selection of transcription activation domains. We show both experimentally and computationally that the tuning of the system is achieved through the selection of three separate modules, each of which enables an adjustable output signal: 1) the transcription-activation domain of the sTF, 2) the binding-site modules in the output promoter, and 3) the core promoter modules which define the transcription initiation site in the output promoter. The system has a novel bidirectional architecture that enables generation of compact, yet versatile expression modules for multiple genes with highly diversified expression levels ranging from negligible to very strong using one synthetic transcription factor. In contrast to most existing modular gene expression regulation systems, the present system is independent from externally added compounds. Furthermore, the established system was minimally affected by the several tested growth conditions. These features suggest that it can be highly useful in large scale biotechnology applications. link: http://identifiers.org/doi/10.1371/journal.pone.0148320

Rantasalo2016 - Synthetic expresion modulator induced STF_VP16: MODEL1510230005v0.0.1

Rantasalo2015-Synthetic_expresion_modulator_inducedSTF_VP16 This model is part of a family of models describing a modul…

Details

This work describes the development and characterization of a modular synthetic expression system that provides a broad range of adjustable and predictable expression levels in S. cerevisiae. The system works as a fixed-gain transcription amplifier, where the input signal is transferred via a synthetic transcription factor (sTF) onto a synthetic promoter, containing a defined core promoter, generating a transcription output signal. The system activation is based on the bacterial LexA-DNA-binding domain, a set of modified, modular LexA-binding sites and a selection of transcription activation domains. We show both experimentally and computationally that the tuning of the system is achieved through the selection of three separate modules, each of which enables an adjustable output signal: 1) the transcription-activation domain of the sTF, 2) the binding-site modules in the output promoter, and 3) the core promoter modules which define the transcription initiation site in the output promoter. The system has a novel bidirectional architecture that enables generation of compact, yet versatile expression modules for multiple genes with highly diversified expression levels ranging from negligible to very strong using one synthetic transcription factor. In contrast to most existing modular gene expression regulation systems, the present system is independent from externally added compounds. Furthermore, the established system was minimally affected by the several tested growth conditions. These features suggest that it can be highly useful in large scale biotechnology applications. link: http://identifiers.org/doi/10.1371/journal.pone.0148320

Rao2014 - Fatty acid beta-oxidation (reduced model): BIOMD0000000835v0.0.1

This represents the reduced version of the "time course model" of Van Eunen et al (2013): Biochemical competition makes…

Details

BACKGROUND: In this paper we propose a model reduction method for biochemical reaction networks governed by a variety of reversible and irreversible enzyme kinetic rate laws, including reversible Michaelis-Menten and Hill kinetics. The method proceeds by a stepwise reduction in the number of complexes, defined as the left and right-hand sides of the reactions in the network. It is based on the Kron reduction of the weighted Laplacian matrix, which describes the graph structure of the complexes and reactions in the network. It does not rely on prior knowledge of the dynamic behaviour of the network and hence can be automated, as we demonstrate. The reduced network has fewer complexes, reactions, variables and parameters as compared to the original network, and yet the behaviour of a preselected set of significant metabolites in the reduced network resembles that of the original network. Moreover the reduced network largely retains the structure and kinetics of the original model. RESULTS: We apply our method to a yeast glycolysis model and a rat liver fatty acid beta-oxidation model. When the number of state variables in the yeast model is reduced from 12 to 7, the difference between metabolite concentrations in the reduced and the full model, averaged over time and species, is only 8%. Likewise, when the number of state variables in the rat-liver beta-oxidation model is reduced from 42 to 29, the difference between the reduced model and the full model is 7.5%. CONCLUSIONS: The method has improved our understanding of the dynamics of the two networks. We found that, contrary to the general disposition, the first few metabolites which were deleted from the network during our stepwise reduction approach, are not those with the shortest convergence times. It shows that our reduction approach performs differently from other approaches that are based on time-scale separation. The method can be used to facilitate fitting of the parameters or to embed a detailed model of interest in a more coarse-grained yet realistic environment. link: http://identifiers.org/pubmed/24885656

Parameters:

NameDescription
Kmcpt2C10AcylCarMAT = 51.0; Keqcpt2 = 2.22; Kmcpt2C12AcylCarMAT = 51.0; Kmcpt2C16AcylCoAMAT = 38.0; Vcpt2 = 0.391; Kmcpt2C12AcylCoAMAT = 38.0; Kmcpt2C10AcylCoAMAT = 38.0; sfcpt2C12=0.95; Kmcpt2C16AcylCarMAT = 51.0; Kmcpt2C14AcylCoAMAT = 38.0; Kmcpt2C14AcylCarMAT = 51.0; Kmcpt2CoAMAT = 30.0; Kmcpt2C6AcylCoAMAT = 1000.0; Kmcpt2C4AcylCoAMAT = 1000000.0; Kmcpt2C8AcylCoAMAT = 38.0; Kmcpt2C8AcylCarMAT = 51.0; Kmcpt2C4AcylCarMAT = 51.0; Kmcpt2C6AcylCarMAT = 51.0; Kmcpt2CarMAT = 350.0Reaction: C12AcylCarMAT => C12AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C12AcylCarMAT, C12AcylCoAMAT, Rate Law: VMAT*sfcpt2C12*Vcpt2*(C12AcylCarMAT*CoAMAT/(Kmcpt2C12AcylCarMAT*Kmcpt2CoAMAT)-C12AcylCoAMAT*CarMAT/(Kmcpt2C12AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))/VMAT
KmlcadC10EnoylCoAMAT = 1.08; KmlcadC14AcylCoAMAT = 7.4; Keqlcad = 6.0; KmlcadFADH = 24.2; sflcadC12=0.9; KmlcadC12AcylCoAMAT = 9.0; KmlcadFAD = 0.12; KmlcadC12EnoylCoAMAT = 1.08; KmlcadC10AcylCoAMAT = 24.3; KmlcadC16EnoylCoAMAT = 1.08; Vlcad = 0.01; KmlcadC16AcylCoAMAT = 2.5; KmlcadC8AcylCoAMAT = 123.0; KmlcadC8EnoylCoAMAT = 1.08; KmlcadC14EnoylCoAMAT = 1.08Reaction: C12AcylCoAMAT => C12EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C12AcylCoAMAT, FADHMAT, Rate Law: VMAT*sflcadC12*Vlcad*(C12AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC12AcylCoAMAT*KmlcadFAD)-C14EnoylCoAMAT*FADHMAT/(KmlcadC12AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C14EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))/VMAT
Keqmckat = 1051.0; KmmckatC4AcylCoAMAT = 13.83; Vmckat = 0.377; KmmckatC8KetoacylCoAMAT = 3.2; KmmckatCoAMAT = 26.6; KmmckatC16KetoacylCoAMAT = 1.1; KmmckatC6KetoacylCoAMAT = 6.7; KmmckatC16AcylCoAMAT = 13.83; KmmckatC10AcylCoAMAT = 13.83; KmmckatC8AcylCoAMAT = 13.83; KmmckatC14KetoacylCoAMAT = 1.2; KmmckatC12KetoacylCoAMAT = 1.3; sfmckatC4=0.49; KmmckatAcetylCoAMAT = 30.0; KmmckatC12AcylCoAMAT = 13.83; KmmckatC6AcylCoAMAT = 13.83; KmmckatC10KetoacylCoAMAT = 2.1; KmmckatC4AcetoacylCoAMAT = 12.4; KmmckatC14AcylCoAMAT = 13.83Reaction: C4AcetoacylCoAMAT => AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, CoAMAT, C4AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: VMAT*sfmckatC4*Vmckat*(C4AcetoacylCoAMAT*CoAMAT/(KmmckatC4AcetoacylCoAMAT*KmmckatCoAMAT)-AcetylCoAMAT*AcetylCoAMAT/(KmmckatC4AcetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))/VMAT
KmmtpC6AcylCoAMAT = 13.83; sfmtpC12=0.81; Keqmtp = 0.71; KmmtpC14EnoylCoAMAT = 25.0; KmmtpC10AcylCoAMAT = 13.83; KmmtpC12AcylCoAMAT = 13.83; KmmtpAcetylCoAMAT = 30.0; KmmtpC8AcylCoAMAT = 13.83; KmmtpC16EnoylCoAMAT = 25.0; KmmtpC14AcylCoAMAT = 13.83; KmmtpC10EnoylCoAMAT = 25.0; KicrotC4AcetoacylCoA = 1.6; KmmtpCoAMAT = 30.0; Vmtp = 2.84; KmmtpC12EnoylCoAMAT = 25.0; KmmtpNADMAT = 60.0; KmmtpC16AcylCoAMAT = 13.83; KmmtpC8EnoylCoAMAT = 25.0; KmmtpNADHMAT = 50.0Reaction: C12EnoylCoAMAT => C10AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, AcetylCoAMAT, C10AcylCoAMAT, C12EnoylCoAMAT, NADHMAT, Rate Law: VMAT*sfmtpC12*Vmtp*(C12EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC12EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C10AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC12EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))/VMAT
KmmcadC12EnoylCoAMAT = 1.08; KmmcadC8AcylCoAMAT = 4.0; KmmcadC6EnoylCoAMAT = 1.08; KmmcadC12AcylCoAMAT = 5.7; KmmcadC6AcylCoAMAT = 9.4; KmmcadC4AcylCoAMAT = 135.0; Vmcad = 0.081; Keqmcad = 6.0; KmmcadFADH = 24.2; KmmcadC10AcylCoAMAT = 5.4; KmmcadFAD = 0.12; KmmcadC10EnoylCoAMAT = 1.08; KmmcadC4EnoylCoAMAT = 1.08; sfmcadC10=0.8; KmmcadC8EnoylCoAMAT = 1.08Reaction: C10AcylCoAMAT => C10EnoylCoAMAT + FADHMAT; C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C10AcylCoAMAT, C10EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sfmcadC10*Vmcad*(C10AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC10AcylCoAMAT*KmmcadFAD)-C10EnoylCoAMAT*FADHMAT/(KmmcadC10AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))/VMAT
KmmtpC6AcylCoAMAT = 13.83; Keqmtp = 0.71; KmmtpC14EnoylCoAMAT = 25.0; KmmtpC10AcylCoAMAT = 13.83; sfmtpC8=0.34; KmmtpC12AcylCoAMAT = 13.83; KmmtpAcetylCoAMAT = 30.0; KmmtpC8AcylCoAMAT = 13.83; KmmtpC16EnoylCoAMAT = 25.0; KmmtpC14AcylCoAMAT = 13.83; KmmtpC10EnoylCoAMAT = 25.0; KicrotC4AcetoacylCoA = 1.6; KmmtpCoAMAT = 30.0; Vmtp = 2.84; KmmtpC12EnoylCoAMAT = 25.0; KmmtpNADMAT = 60.0; KmmtpC16AcylCoAMAT = 13.83; KmmtpC8EnoylCoAMAT = 25.0; KmmtpNADHMAT = 50.0Reaction: C8EnoylCoAMAT => C6AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcetoacylCoAMAT, AcetylCoAMAT, C6AcylCoAMAT, C8EnoylCoAMAT, NADHMAT, Rate Law: VMAT*sfmtpC8*Vmtp*(C8EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC8EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C6AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC8EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))/VMAT
Kmcpt2C10AcylCarMAT = 51.0; Keqcpt2 = 2.22; Kmcpt2C12AcylCarMAT = 51.0; Kmcpt2C16AcylCoAMAT = 38.0; Vcpt2 = 0.391; Kmcpt2C12AcylCoAMAT = 38.0; Kmcpt2C10AcylCoAMAT = 38.0; Kmcpt2C16AcylCarMAT = 51.0; Kmcpt2C14AcylCoAMAT = 38.0; Kmcpt2C14AcylCarMAT = 51.0; Kmcpt2CoAMAT = 30.0; Kmcpt2C6AcylCoAMAT = 1000.0; Kmcpt2C4AcylCoAMAT = 1000000.0; Kmcpt2C8AcylCoAMAT = 38.0; sfcpt2C8=0.35; Kmcpt2C8AcylCarMAT = 51.0; Kmcpt2C4AcylCarMAT = 51.0; Kmcpt2C6AcylCarMAT = 51.0; Kmcpt2CarMAT = 350.0Reaction: C8AcylCarMAT => C8AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C8AcylCarMAT, C8AcylCoAMAT, Rate Law: VMAT*sfcpt2C8*Vcpt2*(C8AcylCarMAT*CoAMAT/(Kmcpt2C8AcylCarMAT*Kmcpt2CoAMAT)-C8AcylCoAMAT*CarMAT/(Kmcpt2C8AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))/VMAT
KmcactCarMAT = 130.0; KmcactCarCYT = 130.0; KicactC16AcylCarCYT=56.0; KmcactC16AcylCarMAT=15.0; KmcactC16AcylCarCYT=15.0; Vfcact = 0.42; Keqcact = 1.0; KicactCarCYT = 200.0; Vrcact = 0.42Reaction: C16AcylCarCYT => C16AcylCarMAT; CarMAT, CarCYT, C16AcylCarCYT, C16AcylCarMAT, Rate Law: Vfcact*(C16AcylCarCYT*CarMAT-C16AcylCarMAT*CarCYT/Keqcact)/(C16AcylCarCYT*CarMAT+KmcactCarMAT*C16AcylCarCYT+KmcactC16AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C16AcylCarMAT*(1+C16AcylCarCYT/KicactC16AcylCarCYT)+CarCYT*(KmcactC16AcylCarMAT+C16AcylCarMAT)))
Kmcpt2C10AcylCarMAT = 51.0; Keqcpt2 = 2.22; Kmcpt2C12AcylCarMAT = 51.0; Kmcpt2C16AcylCoAMAT = 38.0; Vcpt2 = 0.391; Kmcpt2C12AcylCoAMAT = 38.0; Kmcpt2C10AcylCoAMAT = 38.0; Kmcpt2C16AcylCarMAT = 51.0; Kmcpt2C14AcylCoAMAT = 38.0; Kmcpt2C14AcylCarMAT = 51.0; Kmcpt2CoAMAT = 30.0; Kmcpt2C6AcylCoAMAT = 1000.0; sfcpt2C16=0.85; Kmcpt2C4AcylCoAMAT = 1000000.0; Kmcpt2C8AcylCoAMAT = 38.0; Kmcpt2C8AcylCarMAT = 51.0; Kmcpt2C4AcylCarMAT = 51.0; Kmcpt2C6AcylCarMAT = 51.0; Kmcpt2CarMAT = 350.0Reaction: C16AcylCarMAT => C16AcylCoAMAT; C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C16AcylCarMAT, C16AcylCoAMAT, Rate Law: VMAT*sfcpt2C16*Vcpt2*(C16AcylCarMAT*CoAMAT/(Kmcpt2C16AcylCarMAT*Kmcpt2CoAMAT)-C16AcylCoAMAT*CarMAT/(Kmcpt2C16AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))/VMAT
sfmtpC14=0.9; KmmtpC6AcylCoAMAT = 13.83; Keqmtp = 0.71; KmmtpC14EnoylCoAMAT = 25.0; KmmtpC10AcylCoAMAT = 13.83; KmmtpC12AcylCoAMAT = 13.83; KmmtpAcetylCoAMAT = 30.0; KmmtpC8AcylCoAMAT = 13.83; KmmtpC16EnoylCoAMAT = 25.0; KmmtpC14AcylCoAMAT = 13.83; KmmtpC10EnoylCoAMAT = 25.0; KicrotC4AcetoacylCoA = 1.6; KmmtpCoAMAT = 30.0; Vmtp = 2.84; KmmtpC12EnoylCoAMAT = 25.0; KmmtpNADMAT = 60.0; KmmtpC16AcylCoAMAT = 13.83; KmmtpC8EnoylCoAMAT = 25.0; KmmtpNADHMAT = 50.0Reaction: C14EnoylCoAMAT => C12AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, AcetylCoAMAT, C12AcylCoAMAT, C14EnoylCoAMAT, NADHMAT, Rate Law: VMAT*sfmtpC14*Vmtp*(C14EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC14EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C12AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC14EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))/VMAT
KmlcadC10EnoylCoAMAT = 1.08; KmlcadC14AcylCoAMAT = 7.4; Keqlcad = 6.0; sflcadC10=0.75; KmlcadFADH = 24.2; KmlcadC12AcylCoAMAT = 9.0; KmlcadFAD = 0.12; KmlcadC12EnoylCoAMAT = 1.08; KmlcadC10AcylCoAMAT = 24.3; KmlcadC16EnoylCoAMAT = 1.08; Vlcad = 0.01; KmlcadC16AcylCoAMAT = 2.5; KmlcadC8AcylCoAMAT = 123.0; KmlcadC8EnoylCoAMAT = 1.08; KmlcadC14EnoylCoAMAT = 1.08Reaction: C10AcylCoAMAT => C10EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C10AcylCoAMAT, C10EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sflcadC10*Vlcad*(C10AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC10AcylCoAMAT*KmlcadFAD)-C10EnoylCoAMAT*FADHMAT/(KmlcadC10AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))/VMAT
Kmcpt2C10AcylCarMAT = 51.0; Keqcpt2 = 2.22; sfcpt2C10=0.95; Kmcpt2C12AcylCarMAT = 51.0; Kmcpt2C16AcylCoAMAT = 38.0; Vcpt2 = 0.391; Kmcpt2C12AcylCoAMAT = 38.0; Kmcpt2C10AcylCoAMAT = 38.0; Kmcpt2C16AcylCarMAT = 51.0; Kmcpt2C14AcylCoAMAT = 38.0; Kmcpt2C14AcylCarMAT = 51.0; Kmcpt2CoAMAT = 30.0; Kmcpt2C6AcylCoAMAT = 1000.0; Kmcpt2C4AcylCoAMAT = 1000000.0; Kmcpt2C8AcylCoAMAT = 38.0; Kmcpt2C8AcylCarMAT = 51.0; Kmcpt2C4AcylCarMAT = 51.0; Kmcpt2C6AcylCarMAT = 51.0; Kmcpt2CarMAT = 350.0Reaction: C10AcylCarMAT => C10AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C10AcylCarMAT, C10AcylCoAMAT, Rate Law: VMAT*sfcpt2C10*Vcpt2*(C10AcylCarMAT*CoAMAT/(Kmcpt2C10AcylCarMAT*Kmcpt2CoAMAT)-C10AcylCoAMAT*CarMAT/(Kmcpt2C10AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))/VMAT
KmcactC4AcylCarMAT=15.0; KmcactCarMAT = 130.0; KicactC4AcylCarCYT=56.0; KmcactCarCYT = 130.0; Vfcact = 0.42; Keqcact = 1.0; KicactCarCYT = 200.0; Vrcact = 0.42; KmcactC4AcylCarCYT=15.0Reaction: C4AcylCarCYT => C4AcylCarMAT; CarMAT, CarCYT, C4AcylCarCYT, C4AcylCarMAT, Rate Law: Vfcact*(C4AcylCarCYT*CarMAT-C4AcylCarMAT*CarCYT/Keqcact)/(C4AcylCarCYT*CarMAT+KmcactCarMAT*C4AcylCarCYT+KmcactC4AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C4AcylCarMAT*(1+C4AcylCarCYT/KicactC4AcylCarCYT)+CarCYT*(KmcactC4AcylCarMAT+C4AcylCarMAT)))
KmcactCarMAT = 130.0; KmcactCarCYT = 130.0; Vfcact = 0.42; Keqcact = 1.0; KicactCarCYT = 200.0; Vrcact = 0.42; KicactC6AcylCarCYT=56.0; KmcactC6AcylCarMAT=15.0; KmcactC6AcylCarCYT=15.0Reaction: C6AcylCarCYT => C6AcylCarMAT; CarMAT, CarCYT, C6AcylCarCYT, C6AcylCarMAT, Rate Law: Vfcact*(C6AcylCarCYT*CarMAT-C6AcylCarMAT*CarCYT/Keqcact)/(C6AcylCarCYT*CarMAT+KmcactCarMAT*C6AcylCarCYT+KmcactC6AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C6AcylCarMAT*(1+C6AcylCarCYT/KicactC6AcylCarCYT)+CarCYT*(KmcactC6AcylCarMAT+C6AcylCarMAT)))
KmlcadC10EnoylCoAMAT = 1.08; KmlcadC14AcylCoAMAT = 7.4; Keqlcad = 6.0; sflcadC16=0.9; KmlcadFADH = 24.2; KmlcadC12AcylCoAMAT = 9.0; KmlcadFAD = 0.12; KmlcadC12EnoylCoAMAT = 1.08; KmlcadC10AcylCoAMAT = 24.3; KmlcadC16EnoylCoAMAT = 1.08; Vlcad = 0.01; KmlcadC16AcylCoAMAT = 2.5; KmlcadC8AcylCoAMAT = 123.0; KmlcadC8EnoylCoAMAT = 1.08; KmlcadC14EnoylCoAMAT = 1.08Reaction: C16AcylCoAMAT => C16EnoylCoAMAT + FADHMAT; C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C16AcylCoAMAT, C16EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sflcadC16*Vlcad*(C16AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC16AcylCoAMAT*KmlcadFAD)-C16EnoylCoAMAT*FADHMAT/(KmlcadC16AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))/VMAT
KmvlcadC14AcylCoAMAT = 4.0; sfvlcadC14=0.42; KmvlcadC14EnoylCoAMAT = 1.08; KmvlcadFAD = 0.12; KmvlcadC16AcylCoAMAT = 6.5; KmvlcadFADH = 24.2; KmvlcadC16EnoylCoAMAT = 1.08; Keqvlcad = 6.0; KmvlcadC12AcylCoAMAT = 2.7; Vvlcad = 0.008; KmvlcadC12EnoylCoAMAT = 1.08Reaction: C14AcylCoAMAT => C14EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C14AcylCoAMAT, C14EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sfvlcadC14*Vvlcad*(C14AcylCoAMAT*(FADtMAT-FADHMAT)/(KmvlcadC14AcylCoAMAT*KmvlcadFAD)-C14EnoylCoAMAT*FADHMAT/(KmvlcadC14AcylCoAMAT*KmvlcadFAD*Keqvlcad))/((1+C14AcylCoAMAT/KmvlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmvlcadC14EnoylCoAMAT+C16AcylCoAMAT/KmvlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmvlcadC16EnoylCoAMAT+C12AcylCoAMAT/KmvlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmvlcadC12EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmvlcadFAD+FADHMAT/KmvlcadFADH))/VMAT
KmmtpC6AcylCoAMAT = 13.83; Keqmtp = 0.71; KmmtpC14EnoylCoAMAT = 25.0; KmmtpC10AcylCoAMAT = 13.83; KmmtpC12AcylCoAMAT = 13.83; KmmtpAcetylCoAMAT = 30.0; KmmtpC8AcylCoAMAT = 13.83; KmmtpC16EnoylCoAMAT = 25.0; KmmtpC14AcylCoAMAT = 13.83; KmmtpC10EnoylCoAMAT = 25.0; sfmtpC16=1.0; KicrotC4AcetoacylCoA = 1.6; KmmtpCoAMAT = 30.0; Vmtp = 2.84; KmmtpC12EnoylCoAMAT = 25.0; KmmtpNADMAT = 60.0; KmmtpC16AcylCoAMAT = 13.83; KmmtpC8EnoylCoAMAT = 25.0; KmmtpNADHMAT = 50.0Reaction: C16EnoylCoAMAT => C14AcylCoAMAT + AcetylCoAMAT + NADHMAT; C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, AcetylCoAMAT, C14AcylCoAMAT, C16EnoylCoAMAT, NADHMAT, Rate Law: VMAT*sfmtpC16*Vmtp*(C16EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC16EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C14AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC16EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))/VMAT
KicactC8AcylCarCYT=56.0; KmcactCarMAT = 130.0; KmcactC8AcylCarMAT=15.0; KmcactCarCYT = 130.0; Vfcact = 0.42; Keqcact = 1.0; KicactCarCYT = 200.0; Vrcact = 0.42; KmcactC8AcylCarCYT=15.0Reaction: C8AcylCarCYT => C8AcylCarMAT; CarMAT, CarCYT, C8AcylCarCYT, C8AcylCarMAT, Rate Law: Vfcact*(C8AcylCarCYT*CarMAT-C8AcylCarMAT*CarCYT/Keqcact)/(C8AcylCarCYT*CarMAT+KmcactCarMAT*C8AcylCarCYT+KmcactC8AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C8AcylCarMAT*(1+C8AcylCarCYT/KicactC8AcylCarCYT)+CarCYT*(KmcactC8AcylCarMAT+C8AcylCarMAT)))
KmvlcadC14AcylCoAMAT = 4.0; KmvlcadC14EnoylCoAMAT = 1.08; KmvlcadFAD = 0.12; KmvlcadC16AcylCoAMAT = 6.5; KmvlcadFADH = 24.2; sfvlcadC16=1.0; KmvlcadC16EnoylCoAMAT = 1.08; Keqvlcad = 6.0; KmvlcadC12AcylCoAMAT = 2.7; Vvlcad = 0.008; KmvlcadC12EnoylCoAMAT = 1.08Reaction: C16AcylCoAMAT => C16EnoylCoAMAT + FADHMAT; C14AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C16AcylCoAMAT, C16EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sfvlcadC16*Vvlcad*(C16AcylCoAMAT*(FADtMAT-FADHMAT)/(KmvlcadC16AcylCoAMAT*KmvlcadFAD)-C16EnoylCoAMAT*FADHMAT/(KmvlcadC16AcylCoAMAT*KmvlcadFAD*Keqvlcad))/((1+C16AcylCoAMAT/KmvlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmvlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmvlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmvlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmvlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmvlcadC12EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmvlcadFAD+FADHMAT/KmvlcadFADH))/VMAT
Kmcpt2C10AcylCarMAT = 51.0; Keqcpt2 = 2.22; Kmcpt2C12AcylCarMAT = 51.0; Kmcpt2C16AcylCoAMAT = 38.0; Vcpt2 = 0.391; Kmcpt2C12AcylCoAMAT = 38.0; Kmcpt2C10AcylCoAMAT = 38.0; Kmcpt2C14AcylCoAMAT = 38.0; Kmcpt2C16AcylCarMAT = 51.0; sfcpt2C14=1.0; Kmcpt2C14AcylCarMAT = 51.0; Kmcpt2CoAMAT = 30.0; Kmcpt2C6AcylCoAMAT = 1000.0; Kmcpt2C4AcylCoAMAT = 1000000.0; Kmcpt2C8AcylCoAMAT = 38.0; Kmcpt2C8AcylCarMAT = 51.0; Kmcpt2C6AcylCarMAT = 51.0; Kmcpt2CarMAT = 350.0Reaction: C14AcylCarMAT => C14AcylCoAMAT; C16AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C14AcylCarMAT, C14AcylCoAMAT, Rate Law: VMAT*sfcpt2C14*Vcpt2*(C14AcylCarMAT*CoAMAT/(Kmcpt2C14AcylCarMAT*Kmcpt2CoAMAT)-C14AcylCoAMAT*CarMAT/(Kmcpt2C14AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCoAMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))/VMAT
KmmtpC6AcylCoAMAT = 13.83; Keqmtp = 0.71; KmmtpC14EnoylCoAMAT = 25.0; KmmtpC10AcylCoAMAT = 13.83; KmmtpC12AcylCoAMAT = 13.83; KmmtpAcetylCoAMAT = 30.0; KmmtpC8AcylCoAMAT = 13.83; KmmtpC16EnoylCoAMAT = 25.0; KmmtpC14AcylCoAMAT = 13.83; KmmtpC10EnoylCoAMAT = 25.0; KicrotC4AcetoacylCoA = 1.6; KmmtpCoAMAT = 30.0; Vmtp = 2.84; KmmtpC12EnoylCoAMAT = 25.0; KmmtpNADMAT = 60.0; KmmtpC16AcylCoAMAT = 13.83; sfmtpC10=0.73; KmmtpC8EnoylCoAMAT = 25.0; KmmtpNADHMAT = 50.0Reaction: C10EnoylCoAMAT => C8AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, AcetylCoAMAT, C10EnoylCoAMAT, C8AcylCoAMAT, NADHMAT, Rate Law: VMAT*sfmtpC10*Vmtp*(C10EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC10EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C8AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC10EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))/VMAT
KmcactCarMAT = 130.0; KicactC14AcylCarCYT=56.0; KmcactC14AcylCarMAT=15.0; KmcactCarCYT = 130.0; Vfcact = 0.42; Keqcact = 1.0; KicactCarCYT = 200.0; Vrcact = 0.42; KmcactC14AcylCarCYT=15.0Reaction: C14AcylCarCYT => C14AcylCarMAT; CarMAT, CarCYT, C14AcylCarCYT, C14AcylCarMAT, Rate Law: Vfcact*(C14AcylCarCYT*CarMAT-C14AcylCarMAT*CarCYT/Keqcact)/(C14AcylCarCYT*CarMAT+KmcactCarMAT*C14AcylCarCYT+KmcactC14AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C14AcylCarMAT*(1+C14AcylCarCYT/KicactC14AcylCarCYT)+CarCYT*(KmcactC14AcylCarMAT+C14AcylCarMAT)))
Vcpt1=0.012; Kmcpt1C16AcylCoACYT=13.8; Kmcpt1CoACYT=40.7; Keqcpt1=0.45; ncpt1=2.4799; sfcpt1C16=1.0; Kmcpt1CarCYT=125.0; Kicpt1MalCoACYT=9.1; Kmcpt1C16AcylCarCYT=136.0Reaction: => C16AcylCarCYT; C16AcylCoACYT, CarCYT, CoACYT, MalCoACYT, C16AcylCarCYT, Rate Law: VCYT*sfcpt1C16*Vcpt1*(C16AcylCoACYT*CarCYT/(Kmcpt1C16AcylCoACYT*Kmcpt1CarCYT)-C16AcylCarCYT*CoACYT/(Kmcpt1C16AcylCoACYT*Kmcpt1CarCYT*Keqcpt1))/((1+C16AcylCoACYT/Kmcpt1C16AcylCoACYT+C16AcylCarCYT/Kmcpt1C16AcylCarCYT+(MalCoACYT/Kicpt1MalCoACYT)^ncpt1)*(1+CarCYT/Kmcpt1CarCYT+CoACYT/Kmcpt1CoACYT))/VCYT
Keqmckat = 1051.0; KmmckatC4AcylCoAMAT = 13.83; Vmckat = 0.377; KmmckatC8KetoacylCoAMAT = 3.2; KmmckatCoAMAT = 26.6; KmmckatC16KetoacylCoAMAT = 1.1; KmmckatC6KetoacylCoAMAT = 6.7; KmmckatC16AcylCoAMAT = 13.83; KmmckatC10AcylCoAMAT = 13.83; KmmckatC8AcylCoAMAT = 13.83; KmmckatC14KetoacylCoAMAT = 1.2; KmmckatC12KetoacylCoAMAT = 1.3; KmmckatAcetylCoAMAT = 30.0; KmmckatC12AcylCoAMAT = 13.83; KmmckatC6AcylCoAMAT = 13.83; sfmckatC6=1.0; KmmckatC10KetoacylCoAMAT = 2.1; KmmckatC4AcetoacylCoAMAT = 12.4; KmmckatC14AcylCoAMAT = 13.83Reaction: C6KetoacylCoAMAT => C4AcylCoAMAT + AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, AcetylCoAMAT, C4AcylCoAMAT, C6KetoacylCoAMAT, Rate Law: VMAT*sfmckatC6*Vmckat*(C6KetoacylCoAMAT*CoAMAT/(KmmckatC6KetoacylCoAMAT*KmmckatCoAMAT)-C4AcylCoAMAT*AcetylCoAMAT/(KmmckatC6KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))/VMAT
Kmcpt2C10AcylCarMAT = 51.0; Keqcpt2 = 2.22; Kmcpt2C12AcylCarMAT = 51.0; Kmcpt2C16AcylCoAMAT = 38.0; Vcpt2 = 0.391; Kmcpt2C12AcylCoAMAT = 38.0; sfcpt2C4=0.01; Kmcpt2C10AcylCoAMAT = 38.0; Kmcpt2C16AcylCarMAT = 51.0; Kmcpt2C14AcylCoAMAT = 38.0; Kmcpt2C14AcylCarMAT = 51.0; Kmcpt2CoAMAT = 30.0; Kmcpt2C6AcylCoAMAT = 1000.0; Kmcpt2C4AcylCoAMAT = 1000000.0; Kmcpt2C8AcylCoAMAT = 38.0; Kmcpt2C8AcylCarMAT = 51.0; Kmcpt2C4AcylCarMAT = 51.0; Kmcpt2C6AcylCarMAT = 51.0; Kmcpt2CarMAT = 350.0Reaction: C4AcylCarMAT => C4AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, CarMAT, C4AcylCarMAT, C4AcylCoAMAT, Rate Law: VMAT*sfcpt2C4*Vcpt2*(C4AcylCarMAT*CoAMAT/(Kmcpt2C4AcylCarMAT*Kmcpt2CoAMAT)-C4AcylCoAMAT*CarMAT/(Kmcpt2C4AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))/VMAT
KmscadC4EnoylCoAMAT = 1.08; KmscadC6AcylCoAMAT = 285.0; KmscadC6EnoylCoAMAT = 1.08; KmscadFAD = 0.12; Keqscad = 6.0; Vscad = 0.081; KmscadFADH = 24.2; sfscadC4=1.0; KmscadC4AcylCoAMAT = 10.7Reaction: C4AcylCoAMAT => C4EnoylCoAMAT + FADHMAT; C6AcylCoAMAT, FADtMAT, C6EnoylCoAMAT, C4AcylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sfscadC4*Vscad*(C4AcylCoAMAT*(FADtMAT-FADHMAT)/(KmscadC4AcylCoAMAT*KmscadFAD)-C4EnoylCoAMAT*FADHMAT/(KmscadC4AcylCoAMAT*KmscadFAD*Keqscad))/((1+C4AcylCoAMAT/KmscadC4AcylCoAMAT+C4EnoylCoAMAT/KmscadC4EnoylCoAMAT+C6AcylCoAMAT/KmscadC6AcylCoAMAT+C6EnoylCoAMAT/KmscadC6EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmscadFAD+FADHMAT/KmscadFADH))/VMAT
Ksacesink=6000000.0; K1acesink=30.0Reaction: AcetylCoAMAT => ; AcetylCoAMAT, Rate Law: VMAT*Ksacesink*(AcetylCoAMAT-K1acesink)/VMAT
KmmcadC12EnoylCoAMAT = 1.08; KmmcadC8AcylCoAMAT = 4.0; KmmcadC6EnoylCoAMAT = 1.08; KmmcadC12AcylCoAMAT = 5.7; KmmcadC6AcylCoAMAT = 9.4; KmmcadC4AcylCoAMAT = 135.0; Vmcad = 0.081; Keqmcad = 6.0; KmmcadFADH = 24.2; KmmcadC10AcylCoAMAT = 5.4; KmmcadFAD = 0.12; KmmcadC10EnoylCoAMAT = 1.08; KmmcadC4EnoylCoAMAT = 1.08; sfmcadC12=0.38; KmmcadC8EnoylCoAMAT = 1.08Reaction: C12AcylCoAMAT => C12EnoylCoAMAT + FADHMAT; C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C12AcylCoAMAT, C12EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sfmcadC12*Vmcad*(C12AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC12AcylCoAMAT*KmmcadFAD)-C12EnoylCoAMAT*FADHMAT/(KmmcadC12AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))/VMAT
KmcactC10AcylCarMAT=15.0; KmcactCarMAT = 130.0; KmcactCarCYT = 130.0; Vfcact = 0.42; Keqcact = 1.0; KicactCarCYT = 200.0; Vrcact = 0.42; KmcactC10AcylCarCYT=15.0; KicactC10AcylCarCYT=56.0Reaction: C10AcylCarCYT => C10AcylCarMAT; CarMAT, CarCYT, C10AcylCarCYT, C10AcylCarMAT, Rate Law: Vfcact*(C10AcylCarCYT*CarMAT-C10AcylCarMAT*CarCYT/Keqcact)/(C10AcylCarCYT*CarMAT+KmcactCarMAT*C10AcylCarCYT+KmcactC10AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C10AcylCarMAT*(1+C10AcylCarCYT/KicactC10AcylCarCYT)+CarCYT*(KmcactC10AcylCarMAT+C10AcylCarMAT)))
KicactC12AcylCarCYT=56.0; KmcactCarMAT = 130.0; KmcactCarCYT = 130.0; Vfcact = 0.42; Keqcact = 1.0; KicactCarCYT = 200.0; Vrcact = 0.42; KmcactC12AcylCarMAT=15.0; KmcactC12AcylCarCYT=15.0Reaction: C12AcylCarCYT => C12AcylCarMAT; CarMAT, CarCYT, C12AcylCarCYT, C12AcylCarMAT, Rate Law: Vfcact*(C12AcylCarCYT*CarMAT-C12AcylCarMAT*CarCYT/Keqcact)/(C12AcylCarCYT*CarMAT+KmcactCarMAT*C12AcylCarCYT+KmcactC12AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C12AcylCarMAT*(1+C12AcylCarCYT/KicactC12AcylCarCYT)+CarCYT*(KmcactC12AcylCarMAT+C12AcylCarMAT)))
sfcrotC4=1.0; KmcrotC6EnoylCoAMAT = 25.0; Keqmschad = 2.17E-4; sfmschadC4=0.67; Vcrot = 3.6; Keqcrot = 3.13; KmmschadC4AcetoacylCoAMAT = 16.9; KicrotC4AcetoacylCoA = 1.6; Vmschad = 1.0; KmmschadNADMAT = 58.5; KmmschadC4HydroxyacylCoAMAT = 69.9; KmmschadNADHMAT = 5.4; KmmschadC6KetoacylCoAMAT = 5.8; KmcrotC4EnoylCoAMAT = 40.0Reaction: C4EnoylCoAMAT + species_1 => C4AcetoacylCoAMAT + NADHMAT; C6KetoacylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, species_1, C4AcetoacylCoAMAT, NADHMAT, Rate Law: VMAT*sfcrotC4*Vcrot*sfmschadC4*Vmschad*(C4EnoylCoAMAT*species_1/(KmcrotC4EnoylCoAMAT*KmmschadC4HydroxyacylCoAMAT*KmmschadNADMAT)-C4AcetoacylCoAMAT*NADHMAT/(KmcrotC4EnoylCoAMAT*KmmschadC4HydroxyacylCoAMAT*KmmschadNADMAT*Keqcrot*Keqmschad))/(sfcrotC4*Vcrot*(1+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT)*(1+species_1/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT)/(KmcrotC4EnoylCoAMAT*Keqcrot)+sfmschadC4*Vmschad*species_1*(1+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)/(KmmschadC4HydroxyacylCoAMAT*KmmschadNADMAT))/VMAT
KmmcadC12EnoylCoAMAT = 1.08; KmmcadC8AcylCoAMAT = 4.0; KmmcadC6EnoylCoAMAT = 1.08; KmmcadC12AcylCoAMAT = 5.7; KmmcadC6AcylCoAMAT = 9.4; sfmcadC8=0.87; KmmcadC4AcylCoAMAT = 135.0; Vmcad = 0.081; Keqmcad = 6.0; KmmcadFADH = 24.2; KmmcadC10AcylCoAMAT = 5.4; KmmcadFAD = 0.12; KmmcadC10EnoylCoAMAT = 1.08; KmmcadC4EnoylCoAMAT = 1.08; KmmcadC8EnoylCoAMAT = 1.08Reaction: C8AcylCoAMAT => C8EnoylCoAMAT + FADHMAT; C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C8AcylCoAMAT, C8EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sfmcadC8*Vmcad*(C8AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC8AcylCoAMAT*KmmcadFAD)-C8EnoylCoAMAT*FADHMAT/(KmmcadC8AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))/VMAT
KmvlcadC14AcylCoAMAT = 4.0; KmvlcadC14EnoylCoAMAT = 1.08; KmvlcadFAD = 0.12; KmvlcadC16AcylCoAMAT = 6.5; KmvlcadFADH = 24.2; KmvlcadC16EnoylCoAMAT = 1.08; Keqvlcad = 6.0; KmvlcadC12AcylCoAMAT = 2.7; sfvlcadC12=0.11; Vvlcad = 0.008; KmvlcadC12EnoylCoAMAT = 1.08Reaction: C12AcylCoAMAT => C12EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12AcylCoAMAT, C12EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sfvlcadC12*Vvlcad*(C12AcylCoAMAT*(FADtMAT-FADHMAT)/(KmvlcadC12AcylCoAMAT*KmvlcadFAD)-C12EnoylCoAMAT*FADHMAT/(KmvlcadC12AcylCoAMAT*KmvlcadFAD*Keqvlcad))/((1+C12AcylCoAMAT/KmvlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmvlcadC12EnoylCoAMAT+C16AcylCoAMAT/KmvlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmvlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmvlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmvlcadC14EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmvlcadFAD+FADHMAT/KmvlcadFADH))/VMAT
Kmcpt2C10AcylCarMAT = 51.0; sfcpt2C6=0.15; Keqcpt2 = 2.22; Kmcpt2C12AcylCarMAT = 51.0; Kmcpt2C16AcylCoAMAT = 38.0; Vcpt2 = 0.391; Kmcpt2C12AcylCoAMAT = 38.0; Kmcpt2C10AcylCoAMAT = 38.0; Kmcpt2C16AcylCarMAT = 51.0; Kmcpt2C14AcylCoAMAT = 38.0; Kmcpt2C14AcylCarMAT = 51.0; Kmcpt2CoAMAT = 30.0; Kmcpt2C6AcylCoAMAT = 1000.0; Kmcpt2C4AcylCoAMAT = 1000000.0; Kmcpt2C8AcylCoAMAT = 38.0; Kmcpt2C8AcylCarMAT = 51.0; Kmcpt2C4AcylCarMAT = 51.0; Kmcpt2C6AcylCarMAT = 51.0; Kmcpt2CarMAT = 350.0Reaction: C6AcylCarMAT => C6AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcylCoAMAT, CarMAT, C6AcylCarMAT, C6AcylCoAMAT, Rate Law: VMAT*sfcpt2C6*Vcpt2*(C6AcylCarMAT*CoAMAT/(Kmcpt2C6AcylCarMAT*Kmcpt2CoAMAT)-C6AcylCoAMAT*CarMAT/(Kmcpt2C6AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))/VMAT
KmlcadC10EnoylCoAMAT = 1.08; KmlcadC14AcylCoAMAT = 7.4; Keqlcad = 6.0; KmlcadFADH = 24.2; sflcadC14=1.0; KmlcadC12AcylCoAMAT = 9.0; KmlcadFAD = 0.12; KmlcadC12EnoylCoAMAT = 1.08; KmlcadC10AcylCoAMAT = 24.3; KmlcadC16EnoylCoAMAT = 1.08; Vlcad = 0.01; KmlcadC16AcylCoAMAT = 2.5; KmlcadC8AcylCoAMAT = 123.0; KmlcadC8EnoylCoAMAT = 1.08; KmlcadC14EnoylCoAMAT = 1.08Reaction: C14AcylCoAMAT => C14EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C14AcylCoAMAT, C14EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sflcadC14*Vlcad*(C14AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC14AcylCoAMAT*KmlcadFAD)-C14EnoylCoAMAT*FADHMAT/(KmlcadC14AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))/VMAT
KmmcadC12EnoylCoAMAT = 1.08; KmmcadC8AcylCoAMAT = 4.0; KmmcadC6EnoylCoAMAT = 1.08; sfmcadC6=1.0; KmmcadC6AcylCoAMAT = 9.4; KmmcadC12AcylCoAMAT = 5.7; KmmcadC4AcylCoAMAT = 135.0; Vmcad = 0.081; Keqmcad = 6.0; KmmcadFADH = 24.2; KmmcadC10AcylCoAMAT = 5.4; KmmcadFAD = 0.12; KmmcadC10EnoylCoAMAT = 1.08; KmmcadC4EnoylCoAMAT = 1.08; KmmcadC8EnoylCoAMAT = 1.08Reaction: C6AcylCoAMAT => C6EnoylCoAMAT + FADHMAT; C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C4EnoylCoAMAT, C6AcylCoAMAT, C6EnoylCoAMAT, FADHMAT, Rate Law: VMAT*sfmcadC6*Vmcad*(C6AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC6AcylCoAMAT*KmmcadFAD)-C6EnoylCoAMAT*FADHMAT/(KmmcadC6AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))/VMAT
KmcrotC6EnoylCoAMAT = 25.0; Keqmschad = 2.17E-4; Vcrot = 3.6; KmmschadC6HydroxyacylCoAMAT = 28.6; Keqcrot = 3.13; KmmschadC4AcetoacylCoAMAT = 16.9; KicrotC4AcetoacylCoA = 1.6; Vmschad = 1.0; sfcrotC6=0.83; KmmschadNADMAT = 58.5; KmmschadNADHMAT = 5.4; KmmschadC6KetoacylCoAMAT = 5.8; sfmschadC6=1.0; KmcrotC4EnoylCoAMAT = 40.0Reaction: C6EnoylCoAMAT + species_1 => C6KetoacylCoAMAT + NADHMAT; C4AcetoacylCoAMAT, C4EnoylCoAMAT, C6EnoylCoAMAT, species_1, C6KetoacylCoAMAT, NADHMAT, Rate Law: VMAT*sfcrotC6*Vcrot*sfmschadC6*Vmschad*(C6EnoylCoAMAT*species_1/(KmcrotC6EnoylCoAMAT*KmmschadC6HydroxyacylCoAMAT*KmmschadNADMAT)-C6KetoacylCoAMAT*NADHMAT/(KmcrotC6EnoylCoAMAT*KmmschadC6HydroxyacylCoAMAT*KmmschadNADMAT*Keqcrot*Keqmschad))/(sfcrotC6*Vcrot*(1+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+species_1/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT)/(KmcrotC6EnoylCoAMAT*Keqcrot)+sfmschadC6*Vmschad*species_1*(1+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)/(KmmschadC6HydroxyacylCoAMAT*KmmschadNADMAT))/VMAT

States:

NameDescription
C12AcylCarCYTC12AcylCarCYT
C14AcylCarMATC14AcylCarMAT
C10AcylCarCYTC10AcylCarCYT
species 1NAD
C14EnoylCoAMATC14EnoylCoAMAT
C10EnoylCoAMATC10EnoylCoAMAT
C12AcylCoAMATC12AcylCoAMAT
C6AcylCoAMATC6AcylCoAMAT
C16AcylCarCYTC16AcylCarCYT
C10AcylCarMATC10AcylCarMAT
C6AcylCarMATC6AcylCarMAT
C8AcylCoAMATC8AcylCoAMAT
C4AcylCoAMATC4AcylCoAMAT
C4EnoylCoAMATC4EnoylCoAMAT
C16AcylCarMATC16AcylCarMAT
C16AcylCoACYTC16AcylCoACYT
NADHMATNADHMAT
C8AcylCarCYTC8AcylCarCYT
C6AcylCarCYTC6AcylCarCYT
C14AcylCarCYTC14AcylCarCYT
C16EnoylCoAMATC16EnoylCoAMAT
C10AcylCoAMATC10AcylCoAMAT
FADHMATFADHMAT
C14AcylCoAMATC14AcylCoAMAT
AcetylCoAMATAcetylCoAMAT
C16AcylCoAMATC16AcylCoAMAT
C6KetoacylCoAMATC6KetoacylCoAMAT
C4AcylCarCYTC4AcylCarCYT
CoAMATCoAMAT
C12EnoylCoAMATC12EnoylCoAMAT
C6EnoylCoAMATC6EnoylCoAMAT
C8AcylCarMATC8AcylCarMAT
C12AcylCarMATC12AcylCarMAT
C4AcetoacylCoAMATC4AcetoacylCoAMAT
C4AcylCarMATC4AcylCarMAT
C8EnoylCoAMATC8EnoylCoAMAT

Rao2014 - Yeast glycolysis (reduced model): MODEL1403250002v0.0.1

This is the reduced model corresponding to "glucose upshift" condition described in the paper "Testing Biochemistry Revi…

Details

BACKGROUND: In this paper we propose a model reduction method for biochemical reaction networks governed by a variety of reversible and irreversible enzyme kinetic rate laws, including reversible Michaelis-Menten and Hill kinetics. The method proceeds by a stepwise reduction in the number of complexes, defined as the left and right-hand sides of the reactions in the network. It is based on the Kron reduction of the weighted Laplacian matrix, which describes the graph structure of the complexes and reactions in the network. It does not rely on prior knowledge of the dynamic behaviour of the network and hence can be automated, as we demonstrate. The reduced network has fewer complexes, reactions, variables and parameters as compared to the original network, and yet the behaviour of a preselected set of significant metabolites in the reduced network resembles that of the original network. Moreover the reduced network largely retains the structure and kinetics of the original model. RESULTS: We apply our method to a yeast glycolysis model and a rat liver fatty acid beta-oxidation model. When the number of state variables in the yeast model is reduced from 12 to 7, the difference between metabolite concentrations in the reduced and the full model, averaged over time and species, is only 8%. Likewise, when the number of state variables in the rat-liver beta-oxidation model is reduced from 42 to 29, the difference between the reduced model and the full model is 7.5%. CONCLUSIONS: The method has improved our understanding of the dynamics of the two networks. We found that, contrary to the general disposition, the first few metabolites which were deleted from the network during our stepwise reduction approach, are not those with the shortest convergence times. It shows that our reduction approach performs differently from other approaches that are based on time-scale separation. The method can be used to facilitate fitting of the parameters or to embed a detailed model of interest in a more coarse-grained yet realistic environment. link: http://identifiers.org/pubmed/24885656

Rateitschak2012 - Interferon-gamma (IFNγ) induced STAT1 signalling (PC_IFNg100): BIOMD0000000585v0.0.1

Rateitschak2012 - Interferon-gamma (IFNγ) induced STAT1 signalling (PC_IFNg100)This model is described in the article:…

Details

The present work exemplifies how parameter identifiability analysis can be used to gain insights into differences in experimental systems and how uncertainty in parameter estimates can be handled. The case study, presented here, investigates interferon-gamma (IFNγ) induced STAT1 signalling in two cell types that play a key role in pancreatic cancer development: pancreatic stellate and cancer cells. IFNγ inhibits the growth for both types of cells and may be prototypic of agents that simultaneously hit cancer and stroma cells. We combined time-course experiments with mathematical modelling to focus on the common situation in which variations between profiles of experimental time series, from different cell types, are observed. To understand how biochemical reactions are causing the observed variations, we performed a parameter identifiability analysis. We successfully identified reactions that differ in pancreatic stellate cells and cancer cells, by comparing confidence intervals of parameter value estimates and the variability of model trajectories. Our analysis shows that useful information can also be obtained from nonidentifiable parameters. For the prediction of potential therapeutic targets we studied the consequences of uncertainty in the values of identifiable and nonidentifiable parameters. Interestingly, the sensitivity of model variables is robust against parameter variations and against differences between IFNγ induced STAT1 signalling in pancreatic stellate and cancer cells. This provides the basis for a prediction of therapeutic targets that are valid for both cell types. link: http://identifiers.org/pubmed/23284277

Parameters:

NameDescription
tauj = 451.937Reaction: j1 = 4*(mRNA-j1)/tauj, Rate Law: 4*(mRNA-j1)/tauj
k14 = 0.748449; k6 = 0.0666851; k4 = 0.0997621Reaction: Stat1Pd = k4*II*Stat1U/(1+k14*j4)-k6*Stat1Pd, Rate Law: k4*II*Stat1U/(1+k14*j4)-k6*Stat1Pd
scale_Stat1Pnex = 91677.7Reaction: Stat1Pnex = Stat1Pdn*scale_Stat1Pnex, Rate Law: missing
k5 = 298.763; k6 = 0.0666851Reaction: Stat1Pdn = k6*Stat1Pd-k5*Stat1Pdn, Rate Law: k6*Stat1Pd-k5*Stat1Pdn
k3 = 0.0959796; k12 = 12.2679; k14 = 0.748449; k11 = 8.90244; k4 = 0.0997621Reaction: Stat1U = ((k3*d4+k12*Stat1Un)-k11*Stat1U)-k4*II*Stat1U/(1+k14*j4), Rate Law: ((k3*d4+k12*Stat1Un)-k11*Stat1U)-k4*II*Stat1U/(1+k14*j4)
scale_Stat1Pcex = 19.0574Reaction: Stat1Pcex = Stat1Pd*scale_Stat1Pcex, Rate Law: missing
k12 = 12.2679; k5 = 298.763; k11 = 8.90244Reaction: Stat1Un = (k11*Stat1U-k12*Stat1Un)+k5*Stat1Pdn, Rate Law: (k11*Stat1U-k12*Stat1Un)+k5*Stat1Pdn
scale_Stat1nex = 1.21851Reaction: Stat1nex = (Stat1Un+Stat1Pdn)*scale_Stat1nex, Rate Law: missing
taud = 277.363Reaction: d1 = 4*(II-d1)/taud, Rate Law: 4*(II-d1)/taud
k1 = 9.4915E-4Reaction: Ifng = -k1*Ifng*Ir, Rate Law: -k1*Ifng*Ir
k13 = 0.00949819; k9 = 4179.56; k10 = 0.0583427Reaction: mRNA = (k13+k9*i4)-k10*mRNA, Rate Law: (k13+k9*i4)-k10*mRNA
scale_Stat1cex = 0.747697Reaction: Stat1cex = (Stat1U+Stat1Pd)*scale_Stat1cex, Rate Law: missing
scale_Stat1Pex = 34.4009Reaction: Stat1Pex = (Stat1Pd+Stat1Pdn)/2*scale_Stat1Pex, Rate Law: missing
tau = 79.3354Reaction: i1 = 4*(Stat1Pdn-i1)/tau, Rate Law: 4*(Stat1Pdn-i1)/tau

States:

NameDescription
Socs1ex[SOCS1; SOCS1]
Stat1PnexSTAT1Dn (observed)
Stat1PexSTAT1D (observed)
IfngIfng
j2j2
Stat1USTAT1Uc
i3i3
Stat1nexSTAT1n (observed)
i2i2
j4j4
d2d2
j1j1
i4i4
Stat1PdSTAT1D
d1d1
Stat1ex[STAT1; STAT1]
Stat1UnSTAT1Un
Stat1PdnSTAT1Dn
IrIr
Stat1PcexSTAT1Dc (observed)
j3j3
d3d3
i1i1
d4d4
mRNASOCS1
IIII
RSNCexRSNC (observed)
Stat1cexSTAT1c (observed)

Rattanakul2003_BoneFormation_Estrogenadministration: MODEL1012140001v0.0.1

This a model from the article: Modeling of bone formation and resorption mediated by parathyroid hormone: response to…

Details

Bone, a major reservoir of body calcium, is under the hormonal control of the parathyroid hormone (PTH). Several aspects of its growth, turnover, and mechanism, occur in the absence of gonadal hormones. Sex steroids such as estrogen, nonetheless, play an important role in bone physiology, and are extremely essential to maintain bone balance in adults. In order to provide a basis for understanding the underlying mechanisms of bone remodeling as it is mediated by PTH, we propose here a mathematical model of the process. The nonlinear system model is then utilized to study the temporal effect of PTH as well as the action of estrogen replacement therapy on bone turnover. Analysis of the model is done on the assumption, supported by reported clinical evidence, that the process is characterized by highly diversified dynamics, which warrants the use of singular perturbation arguments. The model is shown to exhibit limit cycle behavior, which can develop into chaotic dynamics for certain ranges of the system's parametric values. Effects of estrogen and PTH administrations are then investigated by extending on the core model. Analysis of the model seems to indicate that the paradoxical observation that intermittent PTH administration causes net bone deposition while continuous administration causes net bone loss, and certain other reported phenomena may be attributed to the highly diversified dynamics which characterizes this nonlinear remodeling process. link: http://identifiers.org/pubmed/12753937

Rattanakul2003_BoneFormation_PTHadministration: MODEL1012140000v0.0.1

This a model from the article: Modeling of bone formation and resorption mediated by parathyroid hormone: response to…

Details

Bone, a major reservoir of body calcium, is under the hormonal control of the parathyroid hormone (PTH). Several aspects of its growth, turnover, and mechanism, occur in the absence of gonadal hormones. Sex steroids such as estrogen, nonetheless, play an important role in bone physiology, and are extremely essential to maintain bone balance in adults. In order to provide a basis for understanding the underlying mechanisms of bone remodeling as it is mediated by PTH, we propose here a mathematical model of the process. The nonlinear system model is then utilized to study the temporal effect of PTH as well as the action of estrogen replacement therapy on bone turnover. Analysis of the model is done on the assumption, supported by reported clinical evidence, that the process is characterized by highly diversified dynamics, which warrants the use of singular perturbation arguments. The model is shown to exhibit limit cycle behavior, which can develop into chaotic dynamics for certain ranges of the system's parametric values. Effects of estrogen and PTH administrations are then investigated by extending on the core model. Analysis of the model seems to indicate that the paradoxical observation that intermittent PTH administration causes net bone deposition while continuous administration causes net bone loss, and certain other reported phenomena may be attributed to the highly diversified dynamics which characterizes this nonlinear remodeling process. link: http://identifiers.org/pubmed/12753937

Rattanakul2003_BoneFormationModel: BIOMD0000000274v0.0.1

This a model from the article: Modeling of bone formation and resorption mediated by parathyroid hormone: response t…

Details

Bone, a major reservoir of body calcium, is under the hormonal control of the parathyroid hormone (PTH). Several aspects of its growth, turnover, and mechanism, occur in the absence of gonadal hormones. Sex steroids such as estrogen, nonetheless, play an important role in bone physiology, and are extremely essential to maintain bone balance in adults. In order to provide a basis for understanding the underlying mechanisms of bone remodeling as it is mediated by PTH, we propose here a mathematical model of the process. The nonlinear system model is then utilized to study the temporal effect of PTH as well as the action of estrogen replacement therapy on bone turnover. Analysis of the model is done on the assumption, supported by reported clinical evidence, that the process is characterized by highly diversified dynamics, which warrants the use of singular perturbation arguments. The model is shown to exhibit limit cycle behavior, which can develop into chaotic dynamics for certain ranges of the system's parametric values. Effects of estrogen and PTH administrations are then investigated by extending on the core model. Analysis of the model seems to indicate that the paradoxical observation that intermittent PTH administration causes net bone deposition while continuous administration causes net bone loss, and certain other reported phenomena may be attributed to the highly diversified dynamics which characterizes this nonlinear remodeling process. link: http://identifiers.org/pubmed/12753937

Parameters:

NameDescription
epsilon = 0.1; k2 = 0.5; a2 = 0.009; b2 = 0.3; a3 = 0.675Reaction: y = epsilon*((a2+a3*x)*y*z/(k2+x^2)-b2*y), Rate Law: epsilon*((a2+a3*x)*y*z/(k2+x^2)-b2*y)
epsilon = 0.1; k3 = 0.025; b3 = 0.01; delta = 0.9; a4 = 0.01; a5 = 0.005Reaction: z = epsilon*delta*(a4*x-(b3*z+a5*x*z/(k3+x))), Rate Law: epsilon*delta*(a4*x-(b3*z+a5*x*z/(k3+x)))
k1 = 0.1; b1 = 0.1; a1 = 0.05Reaction: x = a1/(k1+y)-b1*x, Rate Law: a1/(k1+y)-b1*x

States:

NameDescription
x[Parathyroid hormone]
z[osteoblast]
y[osteoclast]

Ratushny2012_ASSURE_I: BIOMD0000000420v0.0.1

This model is from the article: Asymmetric positive feedback loops reliably control biological responses Alexander V…

Details

Positive feedback is a common mechanism enabling biological systems to respond to stimuli in a switch-like manner. Such systems are often characterized by the requisite formation of a heterodimer where only one of the pair is subject to feedback. This ASymmetric Self-UpREgulation (ASSURE) motif is central to many biological systems, including cholesterol homeostasis (LXRα/RXRα), adipocyte differentiation (PPARγ/RXRα), development and differentiation (RAR/RXR), myogenesis (MyoD/E12) and cellular antiviral defense (IRF3/IRF7). To understand why this motif is so prevalent, we examined its properties in an evolutionarily conserved transcriptional regulatory network in yeast (Oaf1p/Pip2p). We demonstrate that the asymmetry in positive feedback confers a competitive advantage and allows the system to robustly increase its responsiveness while precisely tuning the response to a consistent level in the presence of varying stimuli. This study reveals evolutionary advantages for the ASSURE motif, and mechanisms for control, that are relevant to pharmacologic intervention and synthetic biology applications. link: http://identifiers.org/pubmed/22531117

Parameters:

NameDescription
__RATE__=0.1Reaction: P2 =>, Rate Law: __RATE__*P2
ks = 10.0; h = 2.0; k0 = 0.1; dsp1p2kd = NaN; ka = 40.0Reaction: => P2, Rate Law: ks*(k0+(dsp1p2kd/ka)^h)/(1+(dsp1p2kd/ka)^h)

States:

NameDescription
P2[obsolete protein]
TargetTarget

Ratushny2012_ASSURE_II: BIOMD0000000421v0.0.1

This model is from the article: Asymmetric positive feedback loops reliably control biological responses Alexander V…

Details

Positive feedback is a common mechanism enabling biological systems to respond to stimuli in a switch-like manner. Such systems are often characterized by the requisite formation of a heterodimer where only one of the pair is subject to feedback. This ASymmetric Self-UpREgulation (ASSURE) motif is central to many biological systems, including cholesterol homeostasis (LXRα/RXRα), adipocyte differentiation (PPARγ/RXRα), development and differentiation (RAR/RXR), myogenesis (MyoD/E12) and cellular antiviral defense (IRF3/IRF7). To understand why this motif is so prevalent, we examined its properties in an evolutionarily conserved transcriptional regulatory network in yeast (Oaf1p/Pip2p). We demonstrate that the asymmetry in positive feedback confers a competitive advantage and allows the system to robustly increase its responsiveness while precisely tuning the response to a consistent level in the presence of varying stimuli. This study reveals evolutionary advantages for the ASSURE motif, and mechanisms for control, that are relevant to pharmacologic intervention and synthetic biology applications. link: http://identifiers.org/pubmed/22531117

Parameters:

NameDescription
__RATE__=0.1Reaction: P1 =>, Rate Law: __RATE__*P1
ks = 10.0; h = 2.0; k0 = 0.1; dsp1p2kd = NaN; ka = 40.0Reaction: => Target, Rate Law: ks*(k0+(dsp1p2kd/ka)^h)/(1+(dsp1p2kd/ka)^h)

States:

NameDescription
P1[obsolete protein]
TargetTarget

Ratushny2012_NF: BIOMD0000000417v0.0.1

This model is from the article: Asymmetric positive feedback loops reliably control biological responses Alexander V…

Details

Positive feedback is a common mechanism enabling biological systems to respond to stimuli in a switch-like manner. Such systems are often characterized by the requisite formation of a heterodimer where only one of the pair is subject to feedback. This ASymmetric Self-UpREgulation (ASSURE) motif is central to many biological systems, including cholesterol homeostasis (LXRα/RXRα), adipocyte differentiation (PPARγ/RXRα), development and differentiation (RAR/RXR), myogenesis (MyoD/E12) and cellular antiviral defense (IRF3/IRF7). To understand why this motif is so prevalent, we examined its properties in an evolutionarily conserved transcriptional regulatory network in yeast (Oaf1p/Pip2p). We demonstrate that the asymmetry in positive feedback confers a competitive advantage and allows the system to robustly increase its responsiveness while precisely tuning the response to a consistent level in the presence of varying stimuli. This study reveals evolutionary advantages for the ASSURE motif, and mechanisms for control, that are relevant to pharmacologic intervention and synthetic biology applications. link: http://identifiers.org/pubmed/22531117

Parameters:

NameDescription
__RATE__=0.1Reaction: Target =>, Rate Law: __RATE__*Target
dspspkd = NaN; ks = 10.0; h = 2.0; k0 = 0.1; ka = 40.0Reaction: => Target, Rate Law: ks*(k0+(dspspkd/ka)^h)/(1+(dspspkd/ka)^h)

States:

NameDescription
TargetTarget

Ratushny2012_SPF: BIOMD0000000418v0.0.1

This model is from the article: Asymmetric positive feedback loops reliably control biological responses Alexander V…

Details

Positive feedback is a common mechanism enabling biological systems to respond to stimuli in a switch-like manner. Such systems are often characterized by the requisite formation of a heterodimer where only one of the pair is subject to feedback. This ASymmetric Self-UpREgulation (ASSURE) motif is central to many biological systems, including cholesterol homeostasis (LXRα/RXRα), adipocyte differentiation (PPARγ/RXRα), development and differentiation (RAR/RXR), myogenesis (MyoD/E12) and cellular antiviral defense (IRF3/IRF7). To understand why this motif is so prevalent, we examined its properties in an evolutionarily conserved transcriptional regulatory network in yeast (Oaf1p/Pip2p). We demonstrate that the asymmetry in positive feedback confers a competitive advantage and allows the system to robustly increase its responsiveness while precisely tuning the response to a consistent level in the presence of varying stimuli. This study reveals evolutionary advantages for the ASSURE motif, and mechanisms for control, that are relevant to pharmacologic intervention and synthetic biology applications. link: http://identifiers.org/pubmed/22531117

Parameters:

NameDescription
__RATE__=0.1Reaction: P =>, Rate Law: __RATE__*P
dspspkd = NaN; ks = 10.0; h = 2.0; k0 = 0.1; ka = 40.0Reaction: => P, Rate Law: ks*(k0+(dspspkd/ka)^h)/(1+(dspspkd/ka)^h)

States:

NameDescription
P[obsolete protein]

Ratushny2012_SPF_I: BIOMD0000000419v0.0.1

This model is from the article: Asymmetric positive feedback loops reliably control biological responses Alexander V…

Details

Positive feedback is a common mechanism enabling biological systems to respond to stimuli in a switch-like manner. Such systems are often characterized by the requisite formation of a heterodimer where only one of the pair is subject to feedback. This ASymmetric Self-UpREgulation (ASSURE) motif is central to many biological systems, including cholesterol homeostasis (LXRα/RXRα), adipocyte differentiation (PPARγ/RXRα), development and differentiation (RAR/RXR), myogenesis (MyoD/E12) and cellular antiviral defense (IRF3/IRF7). To understand why this motif is so prevalent, we examined its properties in an evolutionarily conserved transcriptional regulatory network in yeast (Oaf1p/Pip2p). We demonstrate that the asymmetry in positive feedback confers a competitive advantage and allows the system to robustly increase its responsiveness while precisely tuning the response to a consistent level in the presence of varying stimuli. This study reveals evolutionary advantages for the ASSURE motif, and mechanisms for control, that are relevant to pharmacologic intervention and synthetic biology applications. link: http://identifiers.org/pubmed/22531117

Parameters:

NameDescription
__RATE__=0.1Reaction: P1 =>, Rate Law: __RATE__*P1
ks = 10.0; h = 2.0; k0 = 0.1; dsp1p2kd = NaN; ka = 40.0Reaction: => P1, Rate Law: ks*(k0+(dsp1p2kd/ka)^h)/(1+(dsp1p2kd/ka)^h)

States:

NameDescription
P2[obsolete protein]
P1[obsolete protein]
TargetTarget

Ray2013 - Meiotic initiation in S. cerevisiae: BIOMD0000000626v0.0.1

Ray2013 - Meiotic initiation in S. cerevisiaeA mathematical representation of early meiotic events, particularly feedbac…

Details

BACKGROUND: Meiosis is the sexual reproduction process common to eukaryotes. The diploid yeast Saccharomyces cerevisiae undergoes meiosis in sporulation medium to form four haploid spores. Initiation of the process is tightly controlled by intricate networks of positive and negative feedback loops. Intriguingly, expression of early meiotic proteins occurs within a narrow time window. Further, sporulation efficiency is strikingly different for yeast strains with distinct mutations or genetic backgrounds. To investigate signal transduction pathways that regulate transient protein expression and sporulation efficiency, we develop a mathematical model using ordinary differential equations. The model describes early meiotic events, particularly feedback mechanisms at the system level and phosphorylation of signaling molecules for regulating protein activities. RESULTS: The mathematical model is capable of simulating the orderly and transient dynamics of meiotic proteins including Ime1, the master regulator of meiotic initiation, and Ime2, a kinase encoded by an early gene. The model is validated by quantitative sporulation phenotypes of single-gene knockouts. Thus, we can use the model to make novel predictions on the cooperation between proteins in the signaling pathway. Virtual perturbations on feedback loops suggest that both positive and negative feedback loops are required to terminate expression of early meiotic proteins. Bifurcation analyses on feedback loops indicate that multiple feedback loops are coordinated to modulate sporulation efficiency. In particular, positive auto-regulation of Ime2 produces a bistable system with a normal meiotic state and a more efficient meiotic state. CONCLUSIONS: By systematically scanning through feedback loops in the mathematical model, we demonstrate that, in yeast, the decisions to terminate protein expression and to sporulate at different efficiencies stem from feedback signals toward the master regulator Ime1 and the early meiotic protein Ime2. We argue that the architecture of meiotic initiation pathway generates a robust mechanism that assures a rapid and complete transition into meiosis. This type of systems-level regulation is a commonly used mechanism controlling developmental programs in yeast and other organisms. Our mathematical model uncovers key regulations that can be manipulated to enhance sporulation efficiency, an important first step in the development of new strategies for producing gametes with high quality and quantity. link: http://identifiers.org/pubmed/23631506

Parameters:

NameDescription
dprimeime_1=1.0; c_1=0.01; cime_1=0.01; dime_1=1.0; pime_1=2.0; sime_1=10.0Reaction: => Ime1; pSok2, Rim11, Ime2, Rate Law: V*(cime_1/(cime_1+pSok2)*sime_1-(pime_1*Ime1*Rim11+dime_1*Ime1+dprimeime_1*Ime2*Ime1/(c_1+Ime1)))
usok_2=1.0; psok_2=0.7; csok_2=0.05Reaction: => pSok2; Ime1, pSok2, Rate Law: V*(csok_2/(csok_2+Ime1)*(1-pSok2)*psok_2-usok_2*pSok2)
dpime_1=1.0; pime_1=2.0Reaction: => pIme1; Ime1, Rim11, pIme1, Rate Law: V*(pime_1*Ime1*Rim11-dpime_1*pIme1)
c_2=1.4; sime_2=10.0; sprimeime_2=3.0; dime_2=8.0; c_3=2.0Reaction: => Ime2; pUme6, pIme1, Rate Law: V*((sime_2*pUme6*pIme1+sprimeime_2*Ime2^5/(c_2^5+Ime2^5))-dime_2*Ime2/(c_3+Ime2))
prim_11=0.01; urim_11=0.1Reaction: => Rim11; Rim11, Rate Law: V*(urim_11*(1-Rim11)-prim_11*Rim11)
pume_6=0.3; uume_6=0.01Reaction: => pUme6; Rim11, Rate Law: V*((1-pUme6)*pume_6*Rim11-uume_6*pUme6)

States:

NameDescription
pSok2[Protein SOK2]
pUme6[Transcriptional regulatory protein UME6]
pIme1[Meiosis-inducing protein 1]
Ime1[Meiosis-inducing protein 1]
Rim11[Serine/threonine-protein kinase RIM11/MSD1]
Ime2[Meiosis induction protein kinase IME2/SME1]

Ray2013 - S.cerevisiae meiosis-specific metabolic network: MODEL1303140001v0.0.1

Ray2013 - S.cerevisiae meiosis-specific metabolic networkMeiosis is a strongly concerved cell division program that gene…

Details

The diploid yeast Saccharomyces cerevisiae undergoes mitosis in glucose-rich medium but enters meiosis in acetate sporulation medium. The transition from mitosis to meiosis involves a remarkable adaptation of the metabolic machinery to the changing environment to meet new energy and biosynthesis requirements. Biochemical studies indicate that five metabolic pathways are active at different stages of sporulation: glutamate formation, tricarboxylic acid cycle, glyoxylate cycle, gluconeogenesis, and glycogenolysis. A dynamic synthesis of macromolecules, including nucleotides, amino acids, and lipids, is also observed. However, the metabolic requirements of sporulating cells are poorly understood. In this study, we apply flux balance analyses to uncover optimal principles driving the operation of metabolic networks over the entire period of sporulation. A meiosis-specific metabolic network is constructed, and flux distribution is simulated using ten objective functions combined with time-course expression-based reaction constraints. By systematically evaluating the correlation between computational and experimental fluxes on pathways and macromolecule syntheses, the metabolic requirements of cells are determined: sporulation requires maximization of ATP production and macromolecule syntheses in the early phase followed by maximization of carbohydrate breakdown and minimization of ATP production in the middle and late stages. Our computational models are validated by in silico deletion of enzymes known to be essential for sporulation. Finally, the models are used to predict novel metabolic genes required for sporulation. This study indicates that yeast cells have distinct metabolic requirements at different phases of meiosis, which may reflect regulation that realizes the optimal outcome of sporulation. Our meiosis-specific network models provide a framework for an in-depth understanding of the roles of enzymes and reactions, and may open new avenues for engineering metabolic pathways to improve sporulation efficiency. link: http://identifiers.org/pubmed/23675502

Razumova2000_MyofilamentContractileBehaviour: MODEL7909395757v0.0.1

This a model from the article: Different myofilament nearest-neighbor interactions have distinctive effects on contrac…

Details

Cooperativity in contractile behavior of myofilament systems almost assuredly arises because of interactions between neighboring sites. These interactions may be of different kinds. Tropomyosin thin-filament regulatory units may have neighbors in steric blocking positions (off) or steric permissive positions (on). The position of these neighbors influence the tendency for the regulatory unit to assume the on or off state. Likewise, the tendency of a myosin cross-bridge to achieve a force-bearing state may be influenced by whether neighboring cross-bridges are in force-bearing states. Also, a cross-bridge in the force-bearing state may influence the tendency of a regulatory unit to enter the on state. We used a mathematical model to examine the influence of each of these three kinds of neighbor interactions on the steady-state force-pCa relation and on the dynamic force redevelopment process. Each neighbor interaction was unique in its effects on maximal Ca(2+)-activated force, position, and symmetry of the force-pCa curve and on the Hill coefficient. Also, each neighbor interaction had a distinctive effect on the time course of force development as assessed by its rate coefficient, k(dev). These diverse effects suggest that variations in all three kinds of nearest-neighbor interactions may be responsible for a wide variety of currently unexplained observations of myofilament contractile behavior. link: http://identifiers.org/pubmed/10827989

reactionsystem_01v0.0.1

This is a testing SBML model

Details

This is a testing SBML model

Reddyhoff2015 - Acetaminophen metabolism and toxicity: BIOMD0000000609v0.0.1

Reddyhoff2015 - Acetaminophen metabolism and toxicityThis model examines acetaminophen metabolism and related hepatotoxi…

Details

Acetaminophen is a widespread and commonly used painkiller all over the world. However, it can cause liver damage when taken in large doses or at repeated chronic doses. Current models of acetaminophen metabolism are complex, and limited to numerical investigation though provide results that represent clinical investigation well. We derive a mathematical model based on mass action laws aimed at capturing the main dynamics of acetaminophen metabolism, in particular the contrast between normal and overdose cases, whilst remaining simple enough for detailed mathematical analysis that can identify key parameters and quantify their role in liver toxicity. We use singular perturbation analysis to separate the different timescales describing the sequence of events in acetaminophen metabolism, systematically identifying which parameters dominate during each of the successive stages. Using this approach we determined, in terms of the model parameters, the critical dose between safe and overdose cases, timescales for exhaustion and regeneration of important cofactors for acetaminophen metabolism and total toxin accumulation as a fraction of initial dose. link: http://identifiers.org/pubmed/26348886

Parameters:

NameDescription
dS = 2.0Reaction: Sulphate__PAPS => ; Sulphate__PAPS, Rate Law: compartment*dS*Sulphate__PAPS
bG = 1.374E-14Reaction: => GSH, Rate Law: compartment*bG
kS = 2.26E14Reaction: Paracetamol_APAP + Sulphate__PAPS => ; Paracetamol_APAP, Sulphate__PAPS, Rate Law: compartment*kS*Paracetamol_APAP*Sulphate__PAPS
k450 = 0.315Reaction: Paracetamol_APAP => NAPQI; Paracetamol_APAP, Rate Law: compartment*k450*Paracetamol_APAP
kN = 0.0315Reaction: NAPQI => Paracetamol_APAP; NAPQI, Rate Law: compartment*kN*NAPQI
bS = 2.65E-14Reaction: => Sulphate__PAPS, Rate Law: compartment*bS
dG = 2.0Reaction: GSH => ; GSH, Rate Law: compartment*dG*GSH
kGSH = 1.6E18Reaction: NAPQI + GSH => ; NAPQI, GSH, Rate Law: compartment*kGSH*NAPQI*GSH
kPSH = 110.0Reaction: NAPQI => Protein_adducts; NAPQI, Rate Law: compartment*kPSH*NAPQI
kG = 2.99Reaction: Paracetamol_APAP => ; Paracetamol_APAP, Rate Law: compartment*kG*Paracetamol_APAP

States:

NameDescription
Paracetamol APAP[urn:miriam:chembl.compound:+CHEMBL112]
Sulphate PAPS[3'-phospho-5'-adenylyl sulfate]
NAPQI[NAD(P)H dehydrogenase [quinone] 1]
Protein adducts[protein; CHEMBL112]
GSH[glutathione]

Reed2003 - Genome-scale metabolic network of Escherichia coli (iJR904): MODEL1507180060v0.0.1

Reed2003 - Genome-scale metabolic network of Escherichia coli (iJR904)This model is described in the article: [An expan…

Details

Diverse datasets, including genomic, transcriptomic, proteomic and metabolomic data, are becoming readily available for specific organisms. There is currently a need to integrate these datasets within an in silico modeling framework. Constraint-based models of Escherichia coli K-12 MG1655 have been developed and used to study the bacterium's metabolism and phenotypic behavior. The most comprehensive E. coli model to date (E. coli iJE660a GSM) accounts for 660 genes and includes 627 unique biochemical reactions.An expanded genome-scale metabolic model of E. coli (iJR904 GSM/GPR) has been reconstructed which includes 904 genes and 931 unique biochemical reactions. The reactions in the expanded model are both elementally and charge balanced. Network gap analysis led to putative assignments for 55 open reading frames (ORFs). Gene to protein to reaction associations (GPR) are now directly included in the model. Comparisons between predictions made by iJR904 and iJE660a models show that they are generally similar but differ under certain circumstances. Analysis of genome-scale proton balancing shows how the flux of protons into and out of the medium is important for maximizing cellular growth.E. coli iJR904 has improved capabilities over iJE660a. iJR904 is a more complete and chemically accurate description of E. coli metabolism than iJE660a. Perhaps most importantly, iJR904 can be used for analyzing and integrating the diverse datasets. iJR904 will help to outline the genotype-phenotype relationship for E. coli K-12, as it can account for genomic, transcriptomic, proteomic and fluxomic data simultaneously. link: http://identifiers.org/pubmed/12952533

Reed2004 - Methionine Cycle: BIOMD0000000698v0.0.1

Reed2004 - Methionine CycleThis model is described in the article: [A mathematical model of the methionine cycle.](http…

Details

Building on the work of Martinov et al. (2000), a mathematical model is developed for the methionine cycle. A large amount of information is available about the enzymes that catalyse individual reaction steps in the cycle, from methionine to S-adenosylmethionine to S-adenosylhomocysteine to homocysteine, and the removal of mass from the cycle by the conversion of homocysteine to cystathionine. Nevertheless, the behavior of the cycle is very complicated since many substrates alter the activities of the enzymes in the reactions that produce them, and some can also alter the activities of other enzymes in the cycle. The model consists of four differential equations, based on known reaction kinetics, that can be solved to give the time course of the concentrations of the four main substrates in the cycle under various circumstances. We show that the behavior of the model in response to genetic abnormalities and dietary deficiencies is similar to the changes seen in a wide variety of experimental studies. We conduct computational "experiments" that give understanding of the regulatory behavior of the methionine cycle under normal conditions and the behavior in the presence of genetic variation and dietary deficiencies. link: http://identifiers.org/pubmed/14637052

Parameters:

NameDescription
beta_1 = 1.7; beta_2 = 30.0Reaction: Homocysteine => Cystathionine; AdoMet, AdoHcy, Rate Law: Compartment*(beta_1*(AdoMet+AdoHcy)-beta_2)*Homocysteine
K_m2_METH_A = 10.0; K_m1_METH = 4.3; V_max_METH = 4521.0Reaction: AdoMet => AdoHcy, Rate Law: Compartment*V_max_METH/(1+K_m1_METH/AdoMet+K_m2_METH_A+K_m2_METH_A*K_m1_METH/AdoMet)
V_max_MATI = 561.0; K_m_MATI = 41.0; K_i_MATI = 50.0Reaction: Methionine => AdoMet, Rate Law: Compartment*V_max_MATI/(1+K_m_MATI/Methionine*(1+AdoMet/K_i_MATI))
V_max_BHMT = 2500.0; K_m_BHMT = 12.0Reaction: Homocysteine => Methionine; AdoMet, AdoHcy, Rate Law: Compartment*(0.7-0.025*((AdoMet+AdoHcy)-150))*V_max_BHMT*Homocysteine/(K_m_BHMT+Homocysteine)
K_d_MS = 1.0; K_m_Hcy_MS = 0.1; V_max_MS = 500.0; K_m_5mTHF_MS = 25.0Reaction: Homocysteine + _5mTHF => Methionine, Rate Law: Compartment*V_max_MS*_5mTHF*Homocysteine/(K_d_MS*K_m_Hcy_MS+K_m_Hcy_MS*_5mTHF+K_m_5mTHF_MS*Homocysteine+_5mTHF*Homocysteine)
k1=1.0Reaction: Metin => Methionine, Rate Law: Compartment*k1*Metin
alpha_1 = 100.0; alpha_2 = 10.0Reaction: AdoHcy => Homocysteine, Rate Law: Compartment*alpha_1*(AdoHcy-alpha_2*Homocysteine)
K_m_GNMT = 4500.0; K_i_GNMT = 20.0; V_max_GNMT = 10600.0Reaction: AdoMet => AdoHcy, Rate Law: Compartment*V_max_GNMT/(1+(K_m_GNMT/AdoMet)^2.3)*1/(1+AdoHcy/K_i_GNMT)
V_max_MATIII = 22870.0; K_m2_MATIII = 21.1; K_m1_MATIII = 16689.3750623336Reaction: Methionine => AdoMet, Rate Law: Compartment*V_max_MATIII/(1+K_m1_MATIII*K_m2_MATIII/(Methionine^2+Methionine*K_m2_MATIII))

States:

NameDescription
Homocysteine[L-Homocysteine; homocysteine]
5mTHF[tetrahydrofolate]
AdoMet[S-Adenosyl-L-methionine; (S)-S-adenosyl-L-methionine]
Cystathionine[Cystathionine; cystathionine]
Methionine[L-Methionine; methionine]
AdoHcy[S-Adenosylhomocysteine; S-Adenosyl-L-homocysteine]
MetinMetin

Reed2008_Glutathione_Metabolism: BIOMD0000000268v0.0.1

This is the model described in the article: A mathematical model of glutathione metabolism. Michael C Reed, Rachel L T…

Details

Glutathione (GSH) plays an important role in anti-oxidant defense and detoxification reactions. It is primarily synthesized in the liver by the transsulfuration pathway and exported to provide precursors for in situ GSH synthesis by other tissues. Deficits in glutathione have been implicated in aging and a host of diseases including Alzheimer's disease, Parkinson's disease, cardiovascular disease, cancer, Down syndrome and autism.We explore the properties of glutathione metabolism in the liver by experimenting with a mathematical model of one-carbon metabolism, the transsulfuration pathway, and glutathione synthesis, transport, and breakdown. The model is based on known properties of the enzymes and the regulation of those enzymes by oxidative stress. We explore the half-life of glutathione, the regulation of glutathione synthesis, and its sensitivity to fluctuations in amino acid input. We use the model to simulate the metabolic profiles previously observed in Down syndrome and autism and compare the model results to clinical data.We show that the glutathione pools in hepatic cells and in the blood are quite insensitive to fluctuations in amino acid input and offer an explanation based on model predictions. In contrast, we show that hepatic glutathione pools are highly sensitive to the level of oxidative stress. The model shows that overexpression of genes on chromosome 21 and an increase in oxidative stress can explain the metabolic profile of Down syndrome. The model also correctly simulates the metabolic profile of autism when oxidative stress is substantially increased and the adenosine concentration is raised. Finally, we discuss how individual variation arises and its consequences for one-carbon and glutathione metabolism. link: http://identifiers.org/pubmed/18442411

Parameters:

NameDescription
K_aic_ART = 100.0 uM; Vm_ART = 55000.0 uM/h; K_10f_ART = 5.9 uMReaction: c_10f + aic => c_thf, Rate Law: cytosol*Vm_ART*c_10f*aic/((K_10f_ART+c_10f)*(K_aic_ART+aic))
V_gsgHb = 40.0 uM/h; K_gsgHb = 1250.0 uMReaction: c_gsg => b_gsg, Rate Law: cytosol*V_gsgHb*c_gsg/(K_gsgHb+c_gsg)
V_oGlu_b = NaN uM/hReaction: => b_glu, Rate Law: blood*V_oGlu_b
Vm_GDC = 15000.0 uM/h; K_thf_GDC = 50.0 uM; K_gly_GDC = 3400.0 uMReaction: m_thf + m_gly => m_2cf + CO, Rate Law: mito*Vm_GDC*m_thf*m_gly/((K_thf_GDC+m_thf)*(K_gly_GDC+m_gly))
ssH2O2 = 0.01 uM; Vm_MS = 500.0 uM/h; Ki_MS = 0.01 uM; K_hcy_MS = 1.0 uM; K_5mf_MS = 25.0 uMReaction: c_5mf + hcy => c_thf + met; H2O2, Rate Law: cytosol*Vm_MS*c_5mf*hcy/((K_5mf_MS+c_5mf)*(K_hcy_MS+hcy))*(ssH2O2+Ki_MS)/(H2O2+Ki_MS)
Vm_DHFR = 2000.0 uM/h; K_dhf_DHFR = 0.5 uM; K_NADPH_DHFR = 4.0 uMReaction: c_dhf + NADPH => c_thf, Rate Law: cytosol*Vm_DHFR*c_dhf*NADPH/((K_dhf_DHFR+c_dhf)*(K_NADPH_DHFR+NADPH))
Ki_DNMT = 1.4 uM; Km_DNMT = 1.4 uM; Vm_DNMT = 180.0 uM/hReaction: sam => sah, Rate Law: cytosol*Vm_DNMT*sam/(Km_DNMT*(1+sah/Ki_DNMT)+sam)
K_10f_MTCH = 100.0 uM; Vf_mMTCH = 790000.0 uM/h; K_1cf_MTCH = 250.0 uM; Vr_MTCH = 20000.0 uM/hReaction: m_1cf => m_10f, Rate Law: mito*(Vf_mMTCH*m_1cf/(K_1cf_MTCH+m_1cf)-Vr_MTCH*m_10f/(K_10f_MTCH+m_10f))
K_gsgLb = 7100.0 uM; V_gsgLb = 4025.0 uM/hReaction: c_gsg => b_gsg, Rate Law: cytosol*V_gsgLb*c_gsg/(K_gsgLb+c_gsg)
K_bcysc = 2100.0 uM; V_bcysc = 14950.0 uM/hReaction: b_cys => c_cys, Rate Law: cytosol*V_bcysc*b_cys/(K_bcysc+b_cys)
K_gly_SHMT = 10000.0 uM; K_2cf_SHMT = 3200.0 uM; Vr_cSHMT = 1.5E7 uM/h; Vf_cSHMT = 5200.0 uM/h; K_ser_SHMT = 600.0 uM; K_thf_SHMT = 50.0 uMReaction: c_ser + c_thf => c_gly + c_2cf, Rate Law: cytosol*(Vf_cSHMT*c_thf*c_ser/((K_thf_SHMT+c_thf)*(K_ser_SHMT+c_ser))-Vr_cSHMT*c_gly*c_2cf/((K_gly_SHMT+c_gly)*(K_2cf_SHMT+c_2cf)))
k2_cNE = 22.0 1/h; k1_cNE = 0.03 1/(uM*h)Reaction: c_thf + HCHO => c_2cf, Rate Law: cytosol*(k1_cNE*c_thf*HCHO-k2_cNE*c_2cf)
Vm_CTGL = 1500.0 uM/h; K_cyt_CTGL = 500.0 uMReaction: cyt => c_cys, Rate Law: cytosol*Vm_CTGL*cyt/(K_cyt_CTGL+cyt)
k1_mNE = 0.03 1/(uM*h); k2_mNE = 20.0 1/hReaction: m_thf + HCHO => m_2cf, Rate Law: mito*(k1_mNE*m_thf*HCHO-k2_mNE*m_2cf)
Vr_MTD = 594000.0 uM/h; K_1cf_MTD = 10.0 uM; K_2cf_MTD = 2.0 uM; Vf_mMTD = 180000.0 uM/hReaction: m_2cf => m_1cf, Rate Law: mito*(Vf_mMTD*m_2cf/(K_2cf_MTD+m_2cf)-Vr_MTD*m_1cf/(K_1cf_MTD+m_1cf))
Ka_CBS = 0.035 uM; ssH2O2 = 0.01 uM; K_hcy_CBS = 1000.0 uM; K_ser_CBS = 2000.0 uM; Vm_CBS = 420000.0 uM/hReaction: hcy + c_ser => cyt; H2O2, sah, sam, Rate Law: cytosol*Vm_CBS*hcy*c_ser/((K_hcy_CBS+hcy)*(K_ser_CBS+c_ser))*((30/102.59)^2+1)/((30/(sam+sah))^2+1)*(H2O2+Ka_CBS)/(ssH2O2+Ka_CBS)
h_gshLb = 3.0 dimensionless; K_gshLb = 3000.0 uM; V_gshLb = 1100.0 uM/hReaction: c_gsh => b_gsh, Rate Law: cytosol*V_gshLb*c_gsh^h_gshLb/(K_gshLb^h_gshLb+c_gsh^h_gshLb)
K_gly_SHMT = 10000.0 uM; K_2cf_SHMT = 3200.0 uM; Vf_mSHMT = 11440.0 uM/h; Vr_mSHMT = 3.0E7 uM/h; K_ser_SHMT = 600.0 uM; K_thf_SHMT = 50.0 uMReaction: m_thf + m_ser => m_gly + m_2cf, Rate Law: mito*(Vf_mSHMT*m_thf*m_ser/((K_thf_SHMT+m_thf)*(K_ser_SHMT+m_ser))-Vr_mSHMT*m_gly*m_2cf/((K_gly_SHMT+m_gly)*(K_2cf_SHMT+m_2cf)))
Vm_GR = 892.5 uM/h; K_NADPH_GR = 10.4 uM; K_gsg_GR = 107.0 uMReaction: c_gsg + NADPH => c_gsh, Rate Law: cytosol*Vm_GR*c_gsg*NADPH/((K_gsg_GR+c_gsg)*(K_NADPH_GR+NADPH))
K_2cf_TS = 14.0 uM; K_DUMP_TS = 6.3 uM; Vm_TS = 5000.0 uM/hReaction: DUMP + c_2cf => c_dhf, Rate Law: cytosol*Vm_TS*DUMP*c_2cf/((K_DUMP_TS+DUMP)*(K_2cf_TS+c_2cf))
K_sah_SAHH = 6.5 uM; K_hcy_SAHH = 150.0 uM; Vr_SAHH = 4530.0 uM/h; Vf_SAHH = 320.0 uM/hReaction: sah => hcy, Rate Law: cytosol*(Vf_SAHH*sah/(K_sah_SAHH+sah)-Vr_SAHH*hcy/(K_hcy_SAHH+hcy))
Vm_mFTD = 1050.0 uM/h; K_10f_FTD = 20.0 uMReaction: m_10f => m_thf, Rate Law: mito*Vm_mFTD*m_10f/(K_10f_FTD+m_10f)
Vm_cFTS = 3900.0 uM/h; K_coo_cFTS = 43.0 uM; K_thf_cFTS = 3.0 uMReaction: c_thf + c_coo => c_10f, Rate Law: cytosol*Vm_cFTS*c_thf*c_coo/((K_thf_cFTS+c_thf)*(K_coo_cFTS+c_coo))
K_10f_PGT = 4.9 uM; K_GAR_PGT = 520.0 uM; Vm_PGT = 24300.0 uM/hReaction: c_10f + GAR => aic + c_thf, Rate Law: cytosol*Vm_PGT*c_10f*GAR/((K_10f_PGT+c_10f)*(K_GAR_PGT+GAR))
K_thf_mFTS = 3.0 uM; K_10f_mFTS = 22.0 uM; Vf_mFTS = 2000.0 uM/h; Vr_mFTS = 6300.0 uM/h; K_coo_mFTS = 43.0 uMReaction: m_thf + m_coo => m_10f, Rate Law: mito*(Vf_mFTS*m_thf*m_coo/((K_thf_mFTS+m_thf)*(K_coo_mFTS+m_coo))-Vr_mFTS*m_10f/(K_10f_mFTS+m_10f))
ssH2O2 = 0.01 uM; Vm_BHMT = 2160.0 uM/h; K_hcy_BHMT = 12.0 uM; Ki_BHMT = 0.01 uM; K_bet_BHMT = 100.0 uMReaction: hcy + BET => met + dmg; H2O2, sah, sam, Rate Law: cytosol*exp((-0.0021)*(sam+sah))*exp(0.0021*102.6)*Vm_BHMT*hcy*BET/((K_hcy_BHMT+hcy)*(K_bet_BHMT+BET))*(ssH2O2+Ki_BHMT)/(H2O2+Ki_BHMT)
K_mser = 5700.0 uM; V_cser = 10000.0 uM/h; K_cser = 5700.0 uM; V_mser = 10000.0 uM/hReaction: m_ser => c_ser, Rate Law: (V_mser*m_ser/(K_mser+m_ser)*mito/3-V_cser*c_ser/(K_cser+c_ser))*cytosol
Vf_cMTCH = 500000.0 uM/h; K_10f_MTCH = 100.0 uM; K_1cf_MTCH = 250.0 uM; Vr_MTCH = 20000.0 uM/hReaction: c_1cf => c_10f, Rate Law: cytosol*(Vf_cMTCH*c_1cf/(K_1cf_MTCH+c_1cf)-Vr_MTCH*c_10f/(K_10f_MTCH+c_10f))
Ki_GNMT = 18.0 uM; K_gly_GNMT = 130.0 uM; K_sam_GNMT = 63.0 uM; Vm_GNMT = 260.0 uM/hReaction: sam + c_gly => sah + src; c_5mf, Rate Law: cytosol*Vm_GNMT*sam*c_gly/((K_sam_GNMT+sam)*(K_gly_GNMT+c_gly))*1/(1+sah/Ki_GNMT)*4.8/(0.35+c_5mf)
K_cgly = 5700.0 uM; K_mgly = 5700.0 uM; V_mgly = 10000.0 uM/h; V_cgly = 10000.0 uM/hReaction: m_gly => c_gly, Rate Law: V_mgly*m_gly/(K_mgly+m_gly)*mito*1/3-V_cgly*c_gly/(K_cgly+c_gly)*cytosol
Vm_GS = 5400.0 uM/h; Ke_GS = 5600.0 1/uM; K_glc_GS = 22.0 uM; Kp_GS = 30.0 1/uM; K_gly_GS = 300.0 uMReaction: glc + c_gly => c_gsh, Rate Law: cytosol*Vm_GS*(c_gly*glc-c_gsh/Ke_GS)/(K_gly_GS*K_glc_GS+glc*K_gly_GS+c_gly*(K_glc_GS+glc)+c_gsh/Kp_GS)
K_gshHb = 150.0 uM; V_gshHb = 150.0 uM/hReaction: c_gsh => b_gsh, Rate Law: cytosol*V_gshHb*c_gsh/(K_gshHb+c_gsh)
Vm_cFTD = 500.0 uM/h; K_10f_FTD = 20.0 uMReaction: c_10f => c_thf, Rate Law: cytosol*Vm_cFTD*c_10f/(K_10f_FTD+c_10f)
Vm_GPX = 4500.0 uM/h; K_H2O2_GPX = 0.09 uM; K_gsh_GPX = 1330.0 uMReaction: c_gsh + H2O2 => c_gsg, Rate Law: cytosol*Vm_GPX*(c_gsh/(K_gsh_GPX+c_gsh))^2*H2O2/(K_H2O2_GPX+H2O2)
k_out_gly = 1.0 1/h; K_bglyc = 150.0 uM; V_bglyc = 4600.0 uM/hReaction: b_gly => c_gly, Rate Law: cytosol*(V_bglyc*b_gly/(K_bglyc+b_gly)-k_out_gly*c_gly)
K_bglutc = 300.0 uM; V_bglutc = 28000.0 uM/h; k_out_glu = 1.0 1/hReaction: b_glu => c_glu, Rate Law: cytosol*(V_bglutc*b_glu/(K_bglutc+b_glu)-k_out_glu*c_glu)
k_out_ser = 1.0 1/h; K_bserc = 150.0 uM; V_bserc = 2700.0 uM/hReaction: b_ser => c_ser, Rate Law: cytosol*(V_bserc*b_ser/(K_bserc+b_ser)-k_out_ser*c_ser)
K_2cf_MTHFR = 50.0 uM; K_NADPH_MTHFR = 16.0 uM; Vm_MTHFR = 5300.0 uM/hReaction: c_2cf + NADPH => c_5mf; sah, sam, Rate Law: cytosol*Vm_MTHFR*c_2cf*NADPH/((K_2cf_MTHFR+c_2cf)*(K_NADPH_MTHFR+NADPH))*72/((10+sam)-sah)
Vm_MAT1 = 260.0 uM/h; Ki_MAT1 = 2140.0 uM; Km_MAT1 = 41.0 uMReaction: met => sam; c_gsg, Rate Law: cytosol*Vm_MAT1*met/(Km_MAT1+met)*(0.23+0.8*exp((-0.0026)*sam))*(Ki_MAT1+66.71)/(Ki_MAT1+c_gsg)
K_src_SDH = 320.0 uM; Vm_SDH = 15000.0 uM/h; K_thf_SDH = 50.0 uMReaction: m_thf + src => m_2cf + m_gly, Rate Law: mito*Vm_SDH*m_thf*src/((K_thf_SDH+m_thf)*(K_src_SDH+src))
V_oGly_b = NaN uM/hReaction: => b_gly, Rate Law: blood*V_oGly_b
Ke_GCS = 5597.0 1/uM; Vm_GCS = 3600.0 uM/h; K_cys_GCS = 100.0 uM; Kp_GCS = 300.0 1/uM; Ka_GCS = 0.01 uM; ssH2O2 = 0.01 uM; K_glu_GCS = 1900.0 uM; Ki_GCS = 8200.0 uMReaction: c_cys + c_glu => glc; H2O2, c_gsh, Rate Law: cytosol*Vm_GCS*(c_cys*c_glu-glc/Ke_GCS)/(K_cys_GCS*K_glu_GCS+c_glu*K_cys_GCS+c_cys*(K_glu_GCS*(1+c_gsh/Ki_GCS)+c_glu)+glc/Kp_GCS+c_gsh/Ki_GCS)*(H2O2+Ka_GCS)/(ssH2O2+Ka_GCS)
V_oCys_b = NaN uM/hReaction: => b_cys, Rate Law: blood*V_oCys_b
K_1cf_MTD = 10.0 uM; Vf_cMTD = 80000.0 uM/h; K_2cf_MTD = 2.0 uM; Vr_cMTD = 600000.0 uM/hReaction: c_2cf => c_1cf + NADPH, Rate Law: cytosol*(Vf_cMTD*c_2cf/(K_2cf_MTD+c_2cf)-Vr_cMTD*c_1cf/(K_1cf_MTD+c_1cf))
Vm_DMGD = 15000.0 uM/h; K_dmg_DMGD = 50.0 uM; K_thf_DMGD = 50.0 uMReaction: m_thf + dmg => m_2cf + src, Rate Law: mito*Vm_DMGD*m_thf*dmg/((K_thf_DMGD+m_thf)*(K_dmg_DMGD+dmg))

States:

NameDescription
sam[S-adenosyl-L-methionine; S-Adenosyl-L-methionine]
met[methionine; Methionine]
dmg[N,N-dimethylglycine; N,N-Dimethylglycine]
c 1cf[644350]
m coo[formate; Formate]
glc[L-gamma-glutamyl-L-cysteine; gamma-L-Glutamyl-L-cysteine]
m thf[(6S)-5,6,7,8-tetrahydrofolic acid; Tetrahydrofolate]
c thf[(6S)-5,6,7,8-tetrahydrofolic acid; Tetrahydrofolate]
b gly[glycine; Glycine]
b gsg[glutathione disulfide; Glutathione disulfide]
NADPH[NADPH; NADPH]
c cys[cysteine; Cysteine]
b cys[cysteine; Cysteine]
cyt[834]
m 2cf[(6R)-5,10-methylenetetrahydrofolic acid; 5,10-Methylenetetrahydrofolate]
src[sarcosine; Sarcosine]
c glu[glutamic acid; Glutamate]
b gsh[glutathione; Glutathione]
hcy[homocysteine; Homocysteine]
m 10f[10-formyltetrahydrofolic acid; 10-Formyltetrahydrofolate]
CO[carbon dioxide; CO2]
m ser[serine; Serine]
c ser[serine; Serine]
c dhf[dihydrofolic acid; Dihydrofolate]
b glu[glutamic acid; Glutamate]
c gsh[glutathione; Glutathione]
sah[S-adenosyl-L-homocysteine; S-Adenosyl-L-homocysteine]
c 5mf[5-methyltetrahydrofolic acid; 5-Methyltetrahydrofolate]
c 2cf[(6R)-5,10-methylenetetrahydrofolic acid; 5,10-Methylenetetrahydrofolate]
aic[AICA ribonucleotide; 1-(5'-Phosphoribosyl)-5-amino-4-imidazolecarboxamide]
DUMP[dUMP; dUMP]
m gly[glycine; Glycine]
c coo[formate; Formate]
H2O2[hydrogen peroxide; Hydrogen peroxide]
c 10f[10-formyltetrahydrofolic acid; 10-Formyltetrahydrofolate]
c gsg[glutathione disulfide; Glutathione disulfide]
m 1cf[644350]

Regan2020 - Adhesion_CIP_Migration_CellCycle_Apoptosis_v2: MODEL2006170001v0.0.1

This 121-node Boolean regulatory network model that synthesizes mechanosensitive signaling that links anchorage and matr…

Details

Epithelial cells respond to their physical neighborhood with mechano-sensitive behaviors required for development and tissue maintenance. These include anchorage dependence, matrix stiffness-dependent proliferation, contact inhibition of proliferation and migration, and collective migration that balances cell crawling with the maintenance of cell junctions. While required for development and tissue repair, these coordinated responses to the microenvironment also contribute to cancer metastasis. Predictive models of the signaling networks that coordinate these behaviors are critical in controlling cell behavior to halt disease. Here we propose a Boolean regulatory network model that synthesizes mechanosensitive signaling that links anchorage to a matrix of varying stiffness and cell density sensing to contact inhibition, proliferation, migration, and apoptosis. Our model can reproduce anchorage dependence and anoikis, detachment-induced cytokinesis errors, the effect of matrix stiffness on proliferation, and contact inhibition of proliferation and migration by two mechanisms that converge on the YAP transcription factor. In addition, we offer testable predictions related to cell cycle-dependent anoikis sensitivity, the molecular requirements for abolishing contact inhibition, and substrate stiffness dependent expression of the catalytic subunit of PI3K. Moreover, our model predicts heterogeneity in migratory vs. non-migratory phenotypes in sub-confluent monolayers, and co-inhibition but semi-independent induction of proliferation vs. migration as a function of cell density and mitogenic stimulation. Our model serves as a stepping-stone towards modeling mechanosensitive routes to the epithelial to mesenchymal transition, capturing the effects of the mesenchymal state on anoikis resistance, and understanding the balance between migration versus proliferation at each stage of the epithelial to mesenchymal transition. link: http://identifiers.org/doi/10.1016/j.csbj.2020.07.016

Rehm2006_Caspase: BIOMD0000000256v0.0.1

This is the standard model described in the article: Systems analysis of effector caspase activation and its control…

Details

Activation of effector caspases is a final step during apoptosis. Single-cell imaging studies have demonstrated that this process may occur as a rapid, all-or-none response, triggering a complete substrate cleavage within 15 min. Based on biochemical data from HeLa cells, we have developed a computational model of apoptosome-dependent caspase activation that was sufficient to remodel the rapid kinetics of effector caspase activation observed in vivo. Sensitivity analyses predicted a critical role for caspase-3-dependent feedback signalling and the X-linked-inhibitor-of-apoptosis-protein (XIAP), but a less prominent role for the XIAP antagonist Smac. Single-cell experiments employing a caspase fluorescence resonance energy transfer substrate verified these model predictions qualitatively and quantitatively. XIAP was predicted to control this all-or-none response, with concentrations as high as 0.15 microM enabling, but concentrations >0.30 microM significantly blocking substrate cleavage. Overexpression of XIAP within these threshold concentrations produced cells showing slow effector caspase activation and submaximal substrate cleavage. Our study supports the hypothesis that high levels of XIAP control caspase activation and substrate cleavage, and may promote apoptosis resistance and sublethal caspase activation in vivo. link: http://identifiers.org/pubmed/16932741

Parameters:

NameDescription
k4 = 12.0 1/(uM*min)Reaction: C9 + C3 => C9P + C3, Rate Law: cell*k4*C9*C3
apo_lim = NaN microM; th_Apop = 2.3 minReaction: PC9 + APAF1 => C9, Rate Law: cell*apo_lim*ln(2)/th_Apop
k49 = 0.0058 1/minReaction: BIR12_C3 =>, Rate Law: cell*k49*BIR12_C3
k48 = 0.0347 1/minReaction: BIR3R_SMAC =>, Rate Law: cell*k48*BIR3R_SMAC
k20 = 12.0 1/(uM*min)Reaction: C3 + BIR3R_C9 => C3 + BIR3R_p2frag + C9P, Rate Law: cell*k20*C3*BIR3R_C9
k2r = 0.0116 1/min; k2 = 7.308E-4 uM/minReaction: => XIAP, Rate Law: cell*(k2-k2r*XIAP)
k22r = 0.144 1/min; k22 = 156.0 1/(uM*min)Reaction: C9 + XIAP_C3 => XIAP_C9_C3, Rate Law: cell*(k22*C9*XIAP_C3-k22r*XIAP_C9_C3)
k24 = 0.0Reaction: BIR3R_p2frag => BIR3R, Rate Law: cell*k24*BIR3R_p2frag
k44 = 0.0347 1/minReaction: XIAP_2SMAC =>, Rate Law: cell*k44*XIAP_2SMAC
k12 = 12.0 1/(uM*min)Reaction: C3 + XIAP_C9 => BIR12 + BIR3R_C9 + C3, Rate Law: cell*k12*C3*XIAP_C9
k28r = 156.0 1/(uM*min); k28 = 420.0 1/(uM^2*min)Reaction: XIAP_C3 + SMAC => XIAP_2SMAC + C3, Rate Law: cell*(k28*XIAP_C3*SMAC*SMAC-k28r*XIAP_2SMAC*C3)
k43 = 0.0347 1/minReaction: XIAP_p2frag_2SMAC =>, Rate Law: cell*k43*XIAP_p2frag_2SMAC
k10r = 0.144 1/min; k10 = 156.0 1/(uM*min)Reaction: C3 + BIR12 => BIR12_C3, Rate Law: cell*(k10*C3*BIR12-k10r*BIR12_C3)
k21r = 0.144 1/min; k21 = 156.0 1/(uM*min)Reaction: C9 + XIAP => XIAP_C9, Rate Law: cell*(k21*C9*XIAP-k21r*XIAP_C9)
k1 = 4.68E-4 uM/min; k1r = 0.0039 1/minReaction: => PC3, Rate Law: cell*(k1-k1r*PC3)
k19 = 12.0 1/(uM*min)Reaction: C3 + XIAP_C9 => C3 + C9P + XIAP_p2frag, Rate Law: cell*k19*C3*XIAP_C9
k9r = 0.144 1/min; k9 = 156.0 1/(uM*min)Reaction: C3 + XIAP_p2frag => XIAP_p2frag_C3, Rate Law: cell*(k9*C3*XIAP_p2frag-k9r*XIAP_p2frag_C3)
k39 = 0.0347 1/minReaction: XIAP_C9_C3 =>, Rate Law: cell*k39*XIAP_C9_C3
k5 = 48.0 1/(uM*min)Reaction: C9P + PC3 => C9P + C3, Rate Law: cell*k5*C9P*PC3
k25 = 0.0Reaction: XIAP_p2frag => XIAP, Rate Law: cell*k25*XIAP_p2frag
k41 = 0.0058 1/minReaction: XIAP_p2frag =>, Rate Law: cell*k41*XIAP_p2frag
k45 = 0.0058 1/minReaction: BIR12 =>, Rate Law: cell*k45*BIR12
k18 = 12.0 1/(uM*min)Reaction: C3 + XIAP_C9_C3 => C3 + C9P + XIAP_p2frag_C3, Rate Law: cell*k18*C3*XIAP_C9_C3
k40 = 0.0347 1/minReaction: XIAP_C9 =>, Rate Law: cell*k40*XIAP_C9
th_CytC = 1.5 minReaction: CytC_mit => CytC_cell, Rate Law: cell*CytC_mit*ln(2)/th_CytC
k26 = 420.0 1/(uM^2*min); k26r = 0.133 1/minReaction: XIAP + SMAC => XIAP_2SMAC, Rate Law: cell*(k26*XIAP*SMAC*SMAC-k26r*XIAP_2SMAC)
k31r = 14.2 1/min; k31 = 0.33 1/(uM*min)Reaction: BIR3R + SMAC => BIR3R_SMAC, Rate Law: cell*(k31*BIR3R*SMAC-k31r*BIR3R_SMAC)
k36 = 0.0058 1/minReaction: C9 =>, Rate Law: cell*k36*C9
k3 = 6.0 1/(uM*min)Reaction: C9 + PC3 => C3 + C9, Rate Law: cell*k3*C9*PC3
k33r = 156.0 1/(uM*min); k33 = 0.33 1/(uM*min)Reaction: BIR3R_C9 + SMAC => BIR3R_SMAC + C9, Rate Law: cell*(k33*BIR3R_C9*SMAC-k33r*BIR3R_SMAC*C9)
k7 = 156.0 1/(uM*min); k7r = 0.144 1/minReaction: C3 + XIAP => XIAP_C3, Rate Law: cell*(k7*C3*XIAP-k7r*XIAP_C3)
th_SMAC = 7.0 minReaction: SMAC_mito => SMAC, Rate Law: cell*SMAC_mito*ln(2)/th_SMAC
k46 = 0.0347 1/minReaction: BIR3R =>, Rate Law: cell*k46*BIR3R
k47 = 0.0058 1/minReaction: BIR12_SMAC =>, Rate Law: cell*k47*BIR12_SMAC
k11 = 12.0 1/(uM*min)Reaction: C3 + XIAP => BIR12 + BIR3R + C3, Rate Law: cell*k11*C3*XIAP
k17 = 12.0 1/(uM*min)Reaction: C3 + XIAP_2SMAC => C3 + BIR12_SMAC + BIR3R_SMAC, Rate Law: cell*k17*C3*XIAP_2SMAC
k13 = 12.0 1/(uM*min)Reaction: C3 + XIAP_C3 => C3 + BIR12_C3 + BIR3R, Rate Law: cell*k13*C3*XIAP_C3
k23 = 156.0 1/(uM*min); k23r = 0.144 1/minReaction: C9 + BIR3R => BIR3R_C9, Rate Law: cell*(k23*C9*BIR3R-k23r*BIR3R_C9)
k8 = 156.0 1/(uM*min); k8r = 0.144 1/minReaction: C3 + XIAP_C9 => XIAP_C9_C3, Rate Law: cell*(k8*C3*XIAP_C9-k8r*XIAP_C9_C3)
k42 = 0.0347 1/minReaction: XIAP_p2frag_C3 =>, Rate Law: cell*k42*XIAP_p2frag_C3
k16 = 12.0 1/(uM*min)Reaction: C3 + XIAP_C9_C3 => C3 + BIR12_C3 + BIR3R_C9, Rate Law: cell*k16*C3*XIAP_C9_C3
k32 = 4.45 1/(uM*min); k32r = 156.0 1/(uM*min)Reaction: BIR12_C3 + SMAC => BIR12_SMAC + C3, Rate Law: cell*(k32*BIR12_C3*SMAC-k32r*BIR12_SMAC*C3)
k53 = 12.0 1/(uM*min)Reaction: C3 + Substrate => C3 + ClvgPrds, Rate Law: cell*k53*C3*Substrate
k14 = 12.0 1/(uM*min)Reaction: C3 + XIAP_p2frag => C3 + BIR12 + BIR3R_p2frag, Rate Law: cell*k14*C3*XIAP_p2frag
k27 = 420.0 1/(uM^2*min); k27r = 156.0 1/(uM*min)Reaction: XIAP_C9 + SMAC => XIAP_2SMAC + C9, Rate Law: cell*(k27*XIAP_C9*SMAC*SMAC-k27r*XIAP_2SMAC*C9)
k6 = 2.4 1/(uM*min)Reaction: PC3 + C3 => C3, Rate Law: cell*k6*PC3*C3
k50 = 0.0058 1/minReaction: BIR3R_C9 =>, Rate Law: cell*k50*BIR3R_C9
k15 = 12.0 1/(uM*min)Reaction: C3 + XIAP_p2frag_C3 => C3 + BIR12_C3 + BIR3R_p2frag, Rate Law: cell*k15*C3*XIAP_p2frag_C3
k29r = 156.0 1/(uM^2*min); k29 = 420.0 1/(uM^2*min)Reaction: XIAP_C9_C3 + SMAC => XIAP_2SMAC + C3 + C9, Rate Law: cell*(k29*XIAP_C9_C3*SMAC*SMAC-k29r*XIAP_2SMAC*C3*C9)
k34r = 156.0 1/min; k34 = 420.0 1/(uM^2*min)Reaction: XIAP_p2frag + SMAC => XIAP_p2frag_2SMAC, Rate Law: cell*(k34*XIAP_p2frag*SMAC*SMAC-k34r*XIAP_p2frag_2SMAC)
k30r = 31.9 1/min; k30 = 4.45 1/(uM*min)Reaction: BIR12 + SMAC => BIR12_SMAC, Rate Law: cell*(k30*BIR12*SMAC-k30r*BIR12_SMAC)
k52 = 0.0058 1/minReaction: SMAC =>, Rate Law: cell*k52*SMAC
k51 = 0.0347 1/minReaction: BIR3R_p2frag =>, Rate Law: cell*k51*BIR3R_p2frag

States:

NameDescription
C3[Caspase-3]
BIR12 C3[E3 ubiquitin-protein ligase XIAP; Caspase-3]
XIAP[E3 ubiquitin-protein ligase XIAP]
BIR3R p2frag[E3 ubiquitin-protein ligase XIAP; Caspase-9]
XIAP p2frag 2SMAC[Caspase-9; E3 ubiquitin-protein ligase XIAP; Diablo homolog, mitochondrial]
XIAP p2frag[Caspase-9; E3 ubiquitin-protein ligase XIAP]
Substrate[Caspase-3]
ClvgPrdsClevage Products
C9[Caspase-9]
XIAP C9 C3[Caspase-3; E3 ubiquitin-protein ligase XIAP; Caspase-9]
CytC cell[Cytochrome c1, heme protein, mitochondrial]
BIR3R[E3 ubiquitin-protein ligase XIAP]
CytC mit[Cytochrome c1, heme protein, mitochondrial]
XIAP C3[Caspase-3; E3 ubiquitin-protein ligase XIAP]
XIAP C9[E3 ubiquitin-protein ligase XIAP; Caspase-9]
SMAC[Diablo homolog, mitochondrial]
XIAP 2SMAC[E3 ubiquitin-protein ligase XIAP]
PC3[Caspase-3]
APAF1[Apoptotic protease-activating factor 1]
BIR3R C9[E3 ubiquitin-protein ligase XIAP; Caspase-9]
BIR12[E3 ubiquitin-protein ligase XIAP]
PC9[Caspase-9]
BIR3R SMAC[E3 ubiquitin-protein ligase XIAP; Diablo homolog, mitochondrial]
BIR12 SMAC[E3 ubiquitin-protein ligase XIAP; Diablo homolog, mitochondrial]
SMAC mito[Diablo homolog, mitochondrial]
XIAP p2frag C3[Caspase-9; Caspase-3; E3 ubiquitin-protein ligase XIAP]

Reidl2006_CalciumOscillationInCilia: MODEL7908934508v0.0.1

This a model from the article: Model of calcium oscillations due to negative feedback in olfactory cilia. Reidl J, B…

Details

We present a mathematical model for calcium oscillations in the cilia of olfactory sensory neurons. The underlying mechanism is based on direct negative regulation of cyclic nucleotide-gated channels by calcium/calmodulin and does not require any autocatalysis such as calcium-induced calcium release. The model is in quantitative agreement with available experimental data, both with respect to oscillations and to fast adaptation. We give predictions for the ranges of parameters in which oscillations should be observable. Relevance of the model to calcium oscillations in other systems is discussed. link: http://identifiers.org/pubmed/16326896

Reiterer2013 - pseudophosphatase STYX role in ERK signalling: BIOMD0000000557v0.0.1

Reiterer2013 - pseudophosphatase STYX role in ERK signallingThis model is described in the article: [Pseudophosphatase…

Details

Serine/threonine/tyrosine-interacting protein (STYX) is a catalytically inactive member of the dual-specificity phosphatases (DUSPs) family. Whereas the role of DUSPs in cellular signaling is well explored, the function of STYX is still unknown. Here, we identify STYX as a spatial regulator of ERK signaling. We used predictive-model simulation to test several hypotheses for possible modes of STYX action. We show that STYX localizes to the nucleus, competes with nuclear DUSP4 for binding to ERK, and acts as a nuclear anchor that regulates ERK nuclear export. Depletion of STYX increases ERK activity in both cytosol and nucleus. Importantly, depletion of STYX causes an ERK-dependent fragmentation of the Golgi apparatus and inhibits Golgi polarization and directional cell migration. Finally, we show that overexpression of STYX reduces ERK1/2 activation, thereby blocking PC12 cell differentiation. Overall, our results identify STYX as an important regulator of ERK1/2 signaling critical for cell migration and PC12 cell differentiation. link: http://identifiers.org/pubmed/23847209

Parameters:

NameDescription
kd3_pERKc = 0.432Reaction: pERK_DUSPc => ERKc + DUSPc; pERK_DUSPc, Rate Law: kd3_pERKc*pERK_DUSPc*cytosol
kd3_ppERKc = 0.388Reaction: ppERK_DUSPc => pERKc + DUSPc; ppERK_DUSPc, Rate Law: kd3_ppERKc*ppERK_DUSPc*cytosol
kd1_pERKn = 1.0; kd2_pERKn = 160.0Reaction: pERKn + DUSPn => pERK_DUSPn; pERKn, DUSPn, pERK_DUSPn, Rate Law: (kd1_pERKn*pERKn*DUSPn/0.22*nucleus-kd2_pERKn*pERK_DUSPn)*nucleus
duspn_ind = 20.0; Kduspn = 1000.0; Tduspn = 10.0; duspn_basal = 1.0Reaction: => duspn; ppERKn, ppERKn, Rate Law: duspn_basal*(1+duspn_ind*ppERKn^2/(ppERKn^2*nucleus+Kduspn^2))*0.693/Tduspn
k_pERKin = 0.144; k_pERKout = 1.08Reaction: pERKc => pERKn; pERKc, pERKn, Rate Law: k_pERKin*pERKc*cytosol-k_pERKout*pERKn*nucleus
kd2_ppERKc = 60.0; kd1_ppERKc = 1.0Reaction: ppERKc + DUSPc => ppERK_DUSPc; ppERKc, DUSPc, ppERK_DUSPc, Rate Law: (kd1_ppERKc*ppERKc*DUSPc/0.94*cytosol-kd2_ppERKc*ppERK_DUSPc)*cytosol
kd2_ppERKn = 60.0; kd1_ppERKn = 1.0Reaction: ppERKn + DUSPn => ppERK_DUSPn; ppERKn, DUSPn, ppERK_DUSPn, Rate Law: (kd1_ppERKn*ppERKn*DUSPn/0.22*nucleus-kd2_ppERKn*ppERK_DUSPn)*nucleus
k3_ERKc = 13.2Reaction: ERK_ppMEKc => pERKc; ERK_ppMEKc, Rate Law: k3_ERKc*ERK_ppMEKc*cytosol
k1_ppES = 1.0; k2_ppES = 60.0Reaction: ppERKn + STYXn => ppERK_STYXn; ppERKn, STYXn, ppERK_STYXn, Rate Law: (k1_ppES*ppERKn*STYXn/0.22*nucleus-k2_ppES*ppERK_STYXn)*nucleus
k1_pES = 1.0; k2_pES = 60.0Reaction: pERKn + STYXn => pERK_STYXn; pERKn, STYXn, pERK_STYXn, Rate Law: (k1_pES*pERKn*STYXn/0.22*nucleus-k2_pES*pERK_STYXn)*nucleus
kd3_ppERKn = 38.88Reaction: ppERK_DUSPn => pERKn + DUSPn; ppERK_DUSPn, Rate Law: kd3_ppERKn*ppERK_DUSPn*nucleus
TDUSPn = 45.0; v2 = 10.0Reaction: => DUSPn; duspn, duspn, Rate Law: v2*duspn/0.22*0.693/TDUSPn*nucleus
k_ppERKout = 0.78; k_ppERKin = 0.66Reaction: ppERKc => ppERKn; ppERKc, ppERKn, Rate Law: k_ppERKin*ppERKc*cytosol-k_ppERKout*ppERKn*nucleus
TDUSPn = 45.0Reaction: DUSPn => ; DUSPn, Rate Law: DUSPn*0.693/TDUSPn*nucleus
Tduspn = 10.0Reaction: duspn => ; duspn, Rate Law: duspn*0.693/Tduspn*nucleus
kd3_pERKn = 43.2Reaction: pERK_DUSPn => ERKn + DUSPn; pERK_DUSPn, Rate Law: kd3_pERKn*pERK_DUSPn*nucleus
k_ERKin = 0.144; k_ERKout = 1.08Reaction: ERKc => ERKn; ERKc, ERKn, Rate Law: k_ERKin*ERKc*cytosol-k_ERKout*ERKn*nucleus
k2_ERKc = 350.0; k1_ERKc = 1.0Reaction: pERKc => pERK_ppMEKc; ppMEKc_tot, ERK_ppMEKc, pERKc, ppMEKc_tot, pERK_ppMEKc, ERK_ppMEKc, Rate Law: k1_ERKc*pERKc*((ppMEKc_tot*cytosol-pERK_ppMEKc*cytosol)-ERK_ppMEKc)*cytosol-k2_ERKc*pERK_ppMEKc*cytosol
k1_ES = 1.0; k2_ES = 60.0Reaction: ERKn + STYXn => ERK_STYXn; ERKn, STYXn, ERK_STYXn, Rate Law: (k1_ES*ERKn*STYXn/0.22*nucleus-k2_ES*ERK_STYXn)*nucleus
k_ppMEKc_tot = 100.0Reaction: ppMEKc_tot = u_ppMEKc_tot*k_ppMEKc_tot, Rate Law: missing
kd2_pERKc = 160.0; kd1_pERKc = 1.0Reaction: pERKc + DUSPc => pERK_DUSPc; pERKc, DUSPc, pERK_DUSPc, Rate Law: (kd1_pERKc*pERKc*DUSPc/0.94*cytosol-kd2_pERKc*pERK_DUSPc)*cytosol

States:

NameDescription
STYXn[Serine/threonine/tyrosine-interacting protein]
pERKn[Mitogen-activated protein kinase 3]
ERK ppMEKc[Mitogen-activated protein kinase 3; Dual specificity mitogen-activated protein kinase kinase 1]
DUSPn[Dual specificity protein phosphatase 4]
ERKc obs[Mitogen-activated protein kinase 3]
ppERKn[Mitogen-activated protein kinase 3]
ERKc[Mitogen-activated protein kinase 3]
pERKc obs[Mitogen-activated protein kinase 3]
ppERK DUSPn[Mitogen-activated protein kinase 3; Dual specificity protein phosphatase 4]
pERK DUSPn[Mitogen-activated protein kinase 3; Dual specificity protein phosphatase 4]
DUSPc[Dual specificity protein phosphatase 4]
ppERKc obs[Mitogen-activated protein kinase 3]
ppMEKc tot[Dual specificity mitogen-activated protein kinase kinase 1]
ppERK STYXn[Mitogen-activated protein kinase 3; Serine/threonine/tyrosine-interacting protein]
duspn[Dual specificity protein phosphatase 4]
ppERKc[Mitogen-activated protein kinase 3]
pERK ppMEKc[Mitogen-activated protein kinase 3; Dual specificity mitogen-activated protein kinase kinase 1]
pERK DUSPc[Mitogen-activated protein kinase 3; Dual specificity protein phosphatase 4]
ppERK DUSPc[Mitogen-activated protein kinase 3; Dual specificity protein phosphatase 4]
ERK STYXn[Mitogen-activated protein kinase 3; Serine/threonine/tyrosine-interacting protein]
pERK STYXn[Mitogen-activated protein kinase 3; Serine/threonine/tyrosine-interacting protein]
u ppMEKc tot[Dual specificity mitogen-activated protein kinase kinase 1]
ERK ppMEKc obs[Mitogen-activated protein kinase 3; Dual specificity mitogen-activated protein kinase kinase 1]
ERKn[Mitogen-activated protein kinase 3]
pERKc[Mitogen-activated protein kinase 3]

Renz2021 - Collection of 13 SBML L3V1 constraint-based models (FBC Version 2) of Staphylococcus aureus: MODEL2007150001v0.0.1

Staphylococcus aureus is a high-priority pathogen causing severe infections with high morbidity and mortality worldwide.…

Details

Staphylococcus aureus is a high-priority pathogen causing severe infections with high morbidity and mortality worldwide. Many S. aureus strains are methicillin-resistant (MRSA) or even multi-drug resistant. It is one of the most successful and prominent modern pathogens. An effective fight against S. aureus infections requires novel targets for antimicrobial and antistaphylococcal therapies. Recent advances in whole-genome sequencing and high-throughput techniques facilitate the generation of genome-scale metabolic models (GEMs). Among the multiple applications of GEMs is drug-targeting in pathogens. Hence, comprehensive and predictive metabolic reconstructions of S. aureus could facilitate the identification of novel targets for antimicrobial therapies. This review aims at giving an overview of all available GEMs of multiple S. aureus strains. We downloaded all 114 available GEMs of S. aureus for further analysis. The scope of each model was evaluated, including the number of reactions, metabolites, and genes. Furthermore, all models were quality-controlled using MEMOTE, an open-source application with standardized metabolic tests. Growth capabilities and model similarities were examined. This review should lead as a guide for choosing the appropriate GEM for a given research question. With the information about the availability, the format, and the strengths and potentials of each model, one can either choose an existing model or combine several models to create models with even higher predictive values. This facilitates model-driven discoveries of novel antimicrobial targets to fight multi-drug resistant S. aureus strains. link: http://identifiers.org/doi/10.1038/s41540-021-00188-4

Renz2021 - Collection of 64 SBML L3V1 genome-scale metabolic models (FBC Version 2) of Staphylococcus aureus: MODEL2007110001v0.0.1

Staphylococcus aureus is a high-priority pathogen causing severe infections with high morbidity and mortality worldwide.…

Details

Staphylococcus aureus is a high-priority pathogen causing severe infections with high morbidity and mortality worldwide. Many S. aureus strains are methicillin-resistant (MRSA) or even multi-drug resistant. It is one of the most successful and prominent modern pathogens. An effective fight against S. aureus infections requires novel targets for antimicrobial and antistaphylococcal therapies. Recent advances in whole-genome sequencing and high-throughput techniques facilitate the generation of genome-scale metabolic models (GEMs). Among the multiple applications of GEMs is drug-targeting in pathogens. Hence, comprehensive and predictive metabolic reconstructions of S. aureus could facilitate the identification of novel targets for antimicrobial therapies. This review aims at giving an overview of all available GEMs of multiple S. aureus strains. We downloaded all 114 available GEMs of S. aureus for further analysis. The scope of each model was evaluated, including the number of reactions, metabolites, and genes. Furthermore, all models were quality-controlled using MEMOTE, an open-source application with standardized metabolic tests. Growth capabilities and model similarities were examined. This review should lead as a guide for choosing the appropriate GEM for a given research question. With the information about the availability, the format, and the strengths and potentials of each model, one can either choose an existing model or combine several models to create models with even higher predictive values. This facilitates model-driven discoveries of novel antimicrobial targets to fight multi-drug resistant S. aureus strains. link: http://identifiers.org/doi/10.1038/s41540-021-00188-4

Reppas2015 - tumor control via alternating immunostimulating and immunosuppressive phases: BIOMD0000000749v0.0.1

The paper describes a model of tumor control via alternating immunostimulating and immunosuppressive phases. Created b…

Details

Despite recent advances in the field of Oncoimmunology, the success potential of immunomodulatory therapies against cancer remains to be elucidated. One of the reasons is the lack of understanding on the complex interplay between tumor growth dynamics and the associated immune system responses. Toward this goal, we consider a mathematical model of vascularized tumor growth and the corresponding effector cell recruitment dynamics. Bifurcation analysis allows for the exploration of model's dynamic behavior and the determination of these parameter regimes that result in immune-mediated tumor control. In this work, we focus on a particular tumor evasion regime that involves tumor and effector cell concentration oscillations of slowly increasing and decreasing amplitude, respectively. Considering a temporal multiscale analysis, we derive an analytically tractable mapping of model solutions onto a weakly negatively damped harmonic oscillator. Based on our analysis, we propose a theory-driven intervention strategy involving immunostimulating and immunosuppressive phases to induce long-term tumor control. link: http://identifiers.org/pubmed/26305801

Parameters:

NameDescription
B = 0.5 1; lm = 1.34 1/dReaction: => R, Rate Law: tumor_microenvironment*lm*B*R/3
d0 = 0.37 1/dReaction: E =>, Rate Law: tumor_microenvironment*d0*E
k = 2.72 1; r = 0.57 1/dReaction: => E; R, Rate Law: tumor_microenvironment*r*R*R*R*E/(k+R*R*R)
ld = 0.3 1; B = 0.5 1; lm = 1.34 1/dReaction: => R, Rate Law: tumor_microenvironment*lm*(1-B)*ld/tanh(R/ld)
d1 = 0.01 1/dReaction: E => ; R, f, Rate Law: tumor_microenvironment*d1*E*R*R*R*f
sigma = 13000.0 1/dReaction: => E, Rate Law: tumor_microenvironment*sigma
c = 0.03 1/dReaction: R => ; E_0, f, Rate Law: tumor_microenvironment*c*E_0*R*f
la = 0.14 1/dReaction: R =>, Rate Law: tumor_microenvironment*la*R/3
B = 0.5 1Reaction: f = R^(B-1)/(R^(B-1)+1), Rate Law: missing

States:

NameDescription
f[Mathematical Operator]
E 0[Effector Immune Cell]
E[Effector Immune Cell]
R[malignant cell]

Resendis-Antonio2007 - Genome-scale metabolic network of Rhizobium etli (iOR363): MODEL1507180006v0.0.1

Resendis-Antonio2007 - Genome-scale metabolic network of Rhizobium etli (iOR363)This model is described in the article:…

Details

Rhizobiaceas are bacteria that fix nitrogen during symbiosis with plants. This symbiotic relationship is crucial for the nitrogen cycle, and understanding symbiotic mechanisms is a scientific challenge with direct applications in agronomy and plant development. Rhizobium etli is a bacteria which provides legumes with ammonia (among other chemical compounds), thereby stimulating plant growth. A genome-scale approach, integrating the biochemical information available for R. etli, constitutes an important step toward understanding the symbiotic relationship and its possible improvement. In this work we present a genome-scale metabolic reconstruction (iOR363) for R. etli CFN42, which includes 387 metabolic and transport reactions across 26 metabolic pathways. This model was used to analyze the physiological capabilities of R. etli during stages of nitrogen fixation. To study the physiological capacities in silico, an objective function was formulated to simulate symbiotic nitrogen fixation. Flux balance analysis (FBA) was performed, and the predicted active metabolic pathways agreed qualitatively with experimental observations. In addition, predictions for the effects of gene deletions during nitrogen fixation in Rhizobia in silico also agreed with reported experimental data. Overall, we present some evidence supporting that FBA of the reconstructed metabolic network for R. etli provides results that are in agreement with physiological observations. Thus, as for other organisms, the reconstructed genome-scale metabolic network provides an important framework which allows us to compare model predictions with experimental measurements and eventually generate hypotheses on ways to improve nitrogen fixation. link: http://identifiers.org/pubmed/17922569

Restif2006 - Whooping cough: BIOMD0000000249v0.0.1

Restif2006 - Whooping coughThis model is described in the article: [Integrating life history and cross-immunity into th…

Details

Models for the diversity and evolution of pathogens have branched into two main directions: the adaptive dynamics of quantitative life-history traits (notably virulence) and the maintenance and invasion of multiple, antigenically diverse strains that interact with the host's immune memory. In a first attempt to reconcile these two approaches, we developed a simple modelling framework where two strains of pathogens, defined by a pair of life-history traits (infectious period and infectivity), interfere through a given level of cross-immunity. We used whooping cough as a potential example, but the framework proposed here could be applied to other acute infectious diseases. Specifically, we analysed the effects of these parameters on the invasion dynamics of one strain into a population, where the second strain is endemic. Whereas the deterministic version of the model converges towards stable coexistence of the two strains in most cases, stochastic simulations showed that transient epidemic dynamics can cause the extinction of either strain. Thus ecological dynamics, modulated by the immune parameters, eventually determine the adaptive value of different pathogen genotypes. We advocate an integrative view of pathogen dynamics at the crossroads of immunology, epidemiology and evolution, as a way towards efficient control of infectious diseases. link: http://identifiers.org/pubmed/16615206

Parameters:

NameDescription
beta_1 = NaN per_yearReaction: S => I_1; I_1, I_1p, N, Rate Law: beta_1*(I_1+I_1p)/N*S
gamma_2 = NaN per_yearReaction: I_2 => R_2, Rate Law: gamma_2*I_2
beta_2 = NaN per_yearReaction: S => I_2; I_2, I_2p, N, Rate Law: beta_2*(I_2+I_2p)/N*S
mu = NaN per_yearReaction: S =>, Rate Law: mu*S
gamma_1 = NaN per_yearReaction: I_1 => R_1, Rate Law: gamma_1*I_1
psi = 0.2 dimensionless; beta_2 = NaN per_yearReaction: R_1 => I_2p; I_2, I_2p, N, Rate Law: (1-psi)*beta_2*(I_2+I_2p)/N*R_1
psi = 0.2 dimensionless; beta_1 = NaN per_yearReaction: R_2 => I_1p; I_1, I_1p, N, Rate Law: (1-psi)*beta_1*(I_1+I_1p)/N*R_2
sigma = NaN per_yearReaction: R_1 => S, Rate Law: sigma*R_1

States:

NameDescription
I 2p[Homo sapiens; Bordetella pertussis]
S[Homo sapiens]
I 1p[Homo sapiens; Bordetella pertussis]
R 1[Homo sapiens]
I 2[Homo sapiens; Bordetella pertussis]
R p[Homo sapiens]
R 2[Homo sapiens]
I 1[Homo sapiens; Bordetella pertussis]

Restif2007 - Vaccination invasion: BIOMD0000000294v0.0.1

Restif2007 - Vaccination invasionThis model is described in the article: [Vaccination and the dynamics of immune evasio…

Details

Vaccines exert strong selective pressures on pathogens, favouring the spread of antigenic variants. We propose a simple mathematical model to investigate the dynamics of a novel pathogenic strain that emerges in a population where a previous strain is maintained at low endemic level by a vaccine. We compare three methods to assess the ability of the novel strain to invade and persist: algebraic rate of invasion; deterministic dynamics; and stochastic dynamics. These three techniques provide complementary predictions on the fate of the system. In particular, we emphasize the importance of stochastic simulations, which account for the possibility of extinctions of either strain. More specifically, our model suggests that the probability of persistence of an invasive strain (i) can be minimized for intermediate levels of vaccine cross-protection (i.e. immune protection against the novel strain) and (ii) is lower if cross-immunity acts through a reduced infectious period rather than through reduced susceptibility. link: http://identifiers.org/pubmed/17210532

Parameters:

NameDescription
sigmaV = NaN per_yearReaction: V => S, Rate Law: sigmaV*V
beta = NaN per_year; tau = 0.7 dimensionlessReaction: V => Iv2; J2, N, I2, Rate Law: beta*(1-tau)*V*(I2+J2+Iv2)/N
beta = NaN per_year; theta = 0.5 dimensionlessReaction: R1 => J2; N, I2, Iv2, Rate Law: beta*(1-theta)*R1*(I2+J2+Iv2)/N
mu = NaN per_yearReaction: Iv2 =>, Rate Law: mu*Iv2
beta = NaN per_yearReaction: S => I2; J2, N, Iv2, Rate Law: beta*S*(I2+J2+Iv2)/N
gamma = NaN per_year; eta = 0.5 dimensionlessReaction: Iv2 => R, Rate Law: gamma/(1-eta)*Iv2
sigma = NaN per_yearReaction: R => S, Rate Law: sigma*R
p = 1.0 dimensionless; mu = NaN per_yearReaction: => V; N, Rate Law: mu*p*N
gamma = NaN per_year; nu = 0.5 dimensionlessReaction: J2 => R, Rate Law: gamma/(1-nu)*J2
gamma = NaN per_yearReaction: I1 => R1, Rate Law: gamma*I1

States:

NameDescription
J1[Homo sapiens; Bordetella pertussis]
S[Homo sapiens]
I1[Homo sapiens; Bordetella pertussis]
I2[Homo sapiens; Bordetella pertussis]
R1[Homo sapiens]
Iv2[Homo sapiens; Bordetella pertussis]
V[Homo sapiens]
R2[Homo sapiens]
J2[Homo sapiens; Bordetella pertussis]
R[Homo sapiens]

Revilla2003 - Controlling HIV infection using recombinant viruses: BIOMD0000000707v0.0.1

This a model from the article: Fighting a virus with a virus: a dynamic model for HIV-1 therapy. Revilla T, Garcia-R…

Details

A mathematical model examined a potential therapy for controlling viral infections using genetically modified viruses. The control of the infection is an indirect effect of the selective elimination by an engineered virus of infected cells that are the source of the pathogens. Therefore, this engineered virus could greatly compensate for a dysfunctional immune system compromised by AIDS. In vitro studies using engineered viruses have been shown to decrease the HIV-1 load about 1000-fold. However, the efficacy of this potential treatment for reducing the viral load in AIDS patients is unknown. The present model studied the interactions among the HIV-1 virus, its main host cell (activated CD4+ T cells), and a therapeutic engineered virus in an in vivo context; and it examined the conditions for controlling the pathogen. This model predicted a significant drop in the HIV-1 load, but the treatment does not eradicate HIV. A basic estimation using a currently engineered virus indicated an HIV-1 load reduction of 92% and a recovery of host cells to 17% of their normal level. Greater success (98% HIV reduction, 44% host cells recovery) is expected as more competent engineered viruses are designed. These results suggest that therapy using viruses could be an alternative to extend the survival of AIDS patients. link: http://identifiers.org/pubmed/12941536

Parameters:

NameDescription
q = 2.0Reaction: Recombinant_Virus =>, Rate Law: Plasma*q*Recombinant_Virus
lamda = 2.0Reaction: => Normal_Th_cells, Rate Law: Plasma*lamda
d = 0.01Reaction: Normal_Th_cells =>, Rate Law: Plasma*d*Normal_Th_cells
u = 2.0Reaction: Pathogen_Virus =>, Rate Law: Plasma*u*Pathogen_Virus
c = 2000.0Reaction: => Recombinant_Virus; Double_Infected_Th_Cells, Rate Law: Plasma*c*Double_Infected_Th_Cells
a = 0.33Reaction: Single_Infected_Th_Cells =>, Rate Law: Plasma*a*Single_Infected_Th_Cells
b = 2.0Reaction: Double_Infected_Th_Cells =>, Rate Law: Plasma*b*Double_Infected_Th_Cells
alpha = 0.004Reaction: Single_Infected_Th_Cells => Double_Infected_Th_Cells; Recombinant_Virus, Rate Law: Plasma*alpha*Recombinant_Virus*Single_Infected_Th_Cells
beta = 0.004Reaction: Normal_Th_cells => Single_Infected_Th_Cells; Pathogen_Virus, Rate Law: Plasma*beta*Normal_Th_cells*Pathogen_Virus
k = 50.0Reaction: => Pathogen_Virus; Single_Infected_Th_Cells, Rate Law: Plasma*k*Single_Infected_Th_Cells

States:

NameDescription
Pathogen Virus[Human immunodeficiency virus 1]
Normal Th cells[helper T-lymphocyte]
Single Infected Th Cells[helper T-lymphocyte; infected cell]
Double Infected Th Cells[infected cell; helper T-lymphocyte]
Recombinant Virus[Genetically Modified Organism; Viruses]

Rex2013 - Genome scale metabolic model of D.shibae (iDsh827): MODEL1308180000v0.0.1

Rex2013 - Genome scale metabolic model of D.shibae (iDsh827)The aerobic anoxygenic phototroph Dinoroseobacter shibae DFL…

Details

The Roseobacter clade is a ubiquitous group of marine α-proteobacteria. To gain insight into the versatile metabolism of this clade, we took a constraint-based approach and created a genome-scale metabolic model (iDsh827) of Dinoroseobacter shibae DFL12T. Our model is the first accounting for the energy demand of motility, the light-driven ATP generation and experimentally determined specific biomass composition. To cover a large variety of environmental conditions, as well as plasmid and single gene knock-out mutants, we simulated 391,560 different physiological states using flux balance analysis. We analyzed our results with regard to energy metabolism, validated them experimentally, and revealed a pronounced metabolic response to the availability of light. Furthermore, we introduced the energy demand of motility as an important parameter in genome-scale metabolic models. The results of our simulations also gave insight into the changing usage of the two degradation routes for dimethylsulfoniopropionate, an abundant compound in the ocean. A side product of dimethylsulfoniopropionate degradation is dimethyl sulfide, which seeds cloud formation and thus enhances the reflection of sunlight. By our exhaustive simulations, we were able to identify single-gene knock-out mutants, which show an increased production of dimethyl sulfide. In addition to the single-gene knock-out simulations we studied the effect of plasmid loss on the metabolism. Moreover, we explored the possible use of a functioning phosphofructokinase for D. shibae. link: http://identifiers.org/pubmed/24098096

Reyes-Palomares2012 - a combined model hepatic polyamine and sulfur aminoacid metabolism - version2: BIOMD0000000450v0.0.1

Reyes-Palomares2012 - a combined model hepatic polyamine and sulfur aminoacid metabolism - version2Mammalian polyamine m…

Details

Many molecular details remain to be uncovered concerning the regulation of polyamine metabolism. A previous model of mammalian polyamine metabolism showed that S-adenosyl methionine availability could play a key role in polyamine homeostasis. To get a deeper insight in this prediction, we have built a combined model by integration of the previously published polyamine model and one-carbon and glutathione metabolism model, published by different research groups. The combined model is robust and it is able to achieve physiological steady-state values, as well as to reproduce the predictions of the individual models. Furthermore, a transition between two versions of our model with new regulatory factors added properly simulates the switch in methionine adenosyl transferase isozymes occurring when the liver enters in proliferative conditions. The combined model is useful to support the previous prediction on the role of S-adenosyl methionine availability in polyamine homeostasis. Furthermore, it could be easily adapted to get deeper insights on the connections of polyamines with energy metabolism. link: http://identifiers.org/pubmed/21814788

Parameters:

NameDescription
parameter_19 = 220.0; Ki=50.0; Km=4.0Reaction: met => sam; sam, met, sam, met, sam, Rate Law: cytosol*parameter_19/(1+(Km*(1+sam/Ki)/met)^0.76)
Vm_cFTD = 500.0; K_10f_FTD = 20.0Reaction: c_10f => c_thf; c_10f, c_10f, Rate Law: cytosol*Vm_cFTD*c_10f/(K_10f_FTD+c_10f)
Vm_mFTD = 1050.0; K_10f_FTD = 20.0Reaction: m_10f => m_thf; m_10f, m_10f, Rate Law: mito*Vm_mFTD*m_10f/(K_10f_FTD+m_10f)
K_DUMP_TS = 6.3; Vm_TS = 5000.0; K_2cf_TS = 14.0Reaction: DUMP + c_2cf => c_dhf; DUMP, c_2cf, DUMP, c_2cf, Rate Law: cytosol*Vm_TS*DUMP*c_2cf/((K_DUMP_TS+DUMP)*(K_2cf_TS+c_2cf))
Vm_PGT = 24300.0; K_GAR_PGT = 520.0; K_10f_PGT = 4.9Reaction: c_10f + GAR => aic + c_thf; GAR, c_10f, GAR, c_10f, Rate Law: cytosol*Vm_PGT*c_10f*GAR/((K_10f_PGT+c_10f)*(K_GAR_PGT+GAR))
K_gly_GDC = 3400.0; Vm_GDC = 15000.0; K_thf_GDC = 50.0Reaction: m_thf + m_gly => m_2cf + CO; m_gly, m_thf, m_gly, m_thf, Rate Law: mito*Vm_GDC*m_thf*m_gly/((K_thf_GDC+m_thf)*(K_gly_GDC+m_gly))
K_aic_ART = 100.0; Vm_ART = 55000.0; K_10f_ART = 5.9Reaction: c_10f + aic => c_thf; aic, c_10f, aic, c_10f, Rate Law: cytosol*Vm_ART*c_10f*aic/((K_10f_ART+c_10f)*(K_aic_ART+aic))
Kisspms=25.0; Kdspms=60.0; Kaspms=0.1; Kiaspms=0.06; Vmspms=193.8Reaction: species_1 + species_4 => species_3; species_1, species_3, species_4, species_1, species_3, species_4, Rate Law: cytosol*Vmspms*species_1*species_4/(Kiaspms*Kdspms*(1+species_3/Kisspms)+Kdspms*species_1+Kaspms*(1+species_3/Kisspms)*species_4+species_1*species_4)
k_out_glu = 1.0; K_bglutc = 300.0; V_bglutc = 28000.0Reaction: b_glu => c_glu; b_glu, c_glu, b_glu, c_glu, Rate Law: cytosol*(V_bglutc*b_glu/(K_bglutc+b_glu)-k_out_glu*c_glu)
k1_mNE = 0.03; k2_mNE = 20.0Reaction: m_thf + HCHO => m_2cf; HCHO, m_2cf, m_thf, HCHO, m_2cf, m_thf, Rate Law: mito*(k1_mNE*m_thf*HCHO-k2_mNE*m_2cf)
K_thf_DMGD = 50.0; Vm_DMGD = 15000.0; K_dmg_DMGD = 50.0Reaction: m_thf + dmg => m_2cf + src; dmg, m_thf, dmg, m_thf, Rate Law: mito*Vm_DMGD*m_thf*dmg/((K_thf_DMGD+m_thf)*(K_dmg_DMGD+dmg))
Ka_CBS = 0.035; ssH2O2 = 0.01; Vm_CBS = 420000.0; K_hcy_CBS = 1000.0; K_ser_CBS = 2000.0Reaction: hcy + c_ser => cyt; H2O2, sah, sam, H2O2, c_ser, hcy, sah, sam, H2O2, c_ser, hcy, sah, sam, Rate Law: cytosol*Vm_CBS*hcy*c_ser/((K_hcy_CBS+hcy)*(K_ser_CBS+c_ser))*((30/102.59)^2+1)/((30/(sam+sah))^2+1)*(H2O2+Ka_CBS)/(ssH2O2+Ka_CBS)
K_cyt_CTGL = 500.0; Vm_CTGL = 1500.0Reaction: cyt => c_cys; cyt, cyt, Rate Law: cytosol*Vm_CTGL*cyt/(K_cyt_CTGL+cyt)
K_sam_GNMT = 63.0; Vm_GNMT = 260.0; K_gly_GNMT = 130.0; Ki_GNMT = 18.0Reaction: sam + c_gly => sah + src; c_5mf, c_5mf, c_gly, sah, sam, c_5mf, c_gly, sah, sam, Rate Law: cytosol*Vm_GNMT*sam*c_gly/((K_sam_GNMT+sam)*(K_gly_GNMT+c_gly))*1/(1+sah/Ki_GNMT)*4.8/(0.35+c_5mf)
parameter_15 = 0.72Reaction: species_8 => species_9; species_8, species_8, Rate Law: cytosol*parameter_15*species_8
k2_cNE = 22.0; k1_cNE = 0.03Reaction: c_thf + HCHO => c_2cf; HCHO, c_2cf, c_thf, HCHO, c_2cf, c_thf, Rate Law: cytosol*(k1_cNE*c_thf*HCHO-k2_cNE*c_2cf)
K_src_SDH = 320.0; Vm_SDH = 15000.0; K_thf_SDH = 50.0Reaction: m_thf + src => m_2cf + m_gly; m_thf, src, m_thf, src, Rate Law: mito*Vm_SDH*m_thf*src/((K_thf_SDH+m_thf)*(K_src_SDH+src))
K_ser_SHMT = 600.0; Vf_mSHMT = 11440.0; K_2cf_SHMT = 3200.0; K_gly_SHMT = 10000.0; K_thf_SHMT = 50.0; Vr_mSHMT = 3.0E7Reaction: m_thf + m_ser => m_gly + m_2cf; m_2cf, m_gly, m_ser, m_thf, m_2cf, m_gly, m_ser, m_thf, Rate Law: mito*(Vf_mSHMT*m_thf*m_ser/((K_thf_SHMT+m_thf)*(K_ser_SHMT+m_ser))-Vr_mSHMT*m_gly*m_2cf/((K_gly_SHMT+m_gly)*(K_2cf_SHMT+m_2cf)))
K_dhf_DHFR = 0.5; Vm_DHFR = 2000.0; K_NADPH_DHFR = 4.0Reaction: c_dhf + NADPH => c_thf; NADPH, c_dhf, NADPH, c_dhf, Rate Law: cytosol*Vm_DHFR*c_dhf*NADPH/((K_dhf_DHFR+c_dhf)*(K_NADPH_DHFR+NADPH))
Kiasamdc=2.5; Kapsamdc=0.5; parameter_3 = 21.1340139923629; Kmsamdc=50.0; Kissamdc=500.0Reaction: sam => species_1; species_3, species_2, sam, species_1, species_2, species_3, sam, species_1, species_2, species_3, Rate Law: cytosol*parameter_3/(1+species_3/Kissamdc)*sam/(Kmsamdc*(1+Kapsamdc/species_2+species_1/Kiasamdc)+sam)
V_gsgHb = 40.0; K_gsgHb = 1250.0Reaction: c_gsg => b_gsg; c_gsg, c_gsg, Rate Law: cytosol*V_gsgHb*c_gsg/(K_gsgHb+c_gsg)
Vf_mFTS = 2000.0; Vr_mFTS = 6300.0; K_coo_mFTS = 43.0; K_thf_mFTS = 3.0; K_10f_mFTS = 22.0Reaction: m_thf + m_coo => m_10f; m_10f, m_coo, m_thf, m_10f, m_coo, m_thf, Rate Law: mito*(Vf_mFTS*m_thf*m_coo/((K_thf_mFTS+m_thf)*(K_coo_mFTS+m_coo))-Vr_mFTS*m_10f/(K_10f_mFTS+m_10f))
K_10f_MTCH = 100.0; Vr_MTCH = 20000.0; Vf_cMTCH = 500000.0; K_1cf_MTCH = 250.0Reaction: c_1cf => c_10f; c_10f, c_1cf, c_10f, c_1cf, Rate Law: cytosol*(Vf_cMTCH*c_1cf/(K_1cf_MTCH+c_1cf)-Vr_MTCH*c_10f/(K_10f_MTCH+c_10f))
V_bcysc = 14950.0; K_bcysc = 2100.0Reaction: b_cys => c_cys; b_cys, b_cys, Rate Law: cytosol*V_bcysc*b_cys/(K_bcysc+b_cys)
V_gsgLb = 4025.0; K_gsgLb = 7100.0Reaction: c_gsg => b_gsg; c_gsg, c_gsg, Rate Law: cytosol*V_gsgLb*c_gsg/(K_gsgLb+c_gsg)
Vmspds=657.0; Kiaspds=0.8; KaSpds=0.3; Kpspds=40.0; Kidspds=100.0Reaction: species_1 + species_2 => species_4; species_1, species_2, species_4, species_1, species_2, species_4, Rate Law: cytosol*Vmspds*species_1*species_2/(Kiaspds*Kpspds*(1+species_4/Kidspds)+Kpspds*species_1+KaSpds*(1+species_4/Kidspds)*species_2+species_1*species_2)
Kmaccoassat=1.5; Kmdssat=130.0; C=4.44; Kmsssat=35.0; parameter_2 = 42.2853792055417; Kmcoassat=40.0Reaction: species_3 + species_8 => species_5 + species_9; species_4, species_3, species_4, species_8, species_9, species_3, species_4, species_8, species_9, Rate Law: cytosol*1/C*parameter_2*species_3*species_8/(Kmsssat*(1+species_4/Kmdssat)*Kmaccoassat*(1+species_9/Kmcoassat)+Kmaccoassat*(1+species_9/Kmcoassat)*species_3+Kmsssat*(1+species_4/Kmdssat)*species_8+species_3*species_8)
parameter_14 = 0.24Reaction: species_9 => species_8; species_9, species_9, Rate Law: cytosol*parameter_14*species_9
Kmaccoassat=1.5; Kmdssat=130.0; Kmsssat=35.0; parameter_2 = 42.2853792055417; Kmcoassat=40.0Reaction: species_4 + species_8 => species_6 + species_9; species_3, species_3, species_4, species_8, species_9, species_3, species_4, species_8, species_9, Rate Law: cytosol*parameter_2*species_4*species_8/(Kmdssat*(1+species_3/Kmsssat)*Kmaccoassat*(1+species_9/Kmcoassat)+Kmaccoassat*(1+species_9/Kmcoassat)*species_4+Kmdssat*(1+species_3/Kmsssat)*species_8+species_4*species_8)
K_ser_SHMT = 600.0; K_2cf_SHMT = 3200.0; Vr_cSHMT = 1.5E7; Vf_cSHMT = 5200.0; K_gly_SHMT = 10000.0; K_thf_SHMT = 50.0Reaction: c_ser + c_thf => c_gly + c_2cf; c_2cf, c_gly, c_ser, c_thf, c_2cf, c_gly, c_ser, c_thf, Rate Law: cytosol*(Vf_cSHMT*c_thf*c_ser/((K_thf_SHMT+c_thf)*(K_ser_SHMT+c_ser))-Vr_cSHMT*c_gly*c_2cf/((K_gly_SHMT+c_gly)*(K_2cf_SHMT+c_2cf)))
Kmodc=60.0; Kipodc=1300.0; parameter_1 = 72.2557178994351Reaction: species_7 => species_2; species_2, species_7, species_2, species_7, Rate Law: cytosol*parameter_1*species_7/(Kmodc*(1+species_2/Kipodc)+species_7)
K_1cf_MTD = 10.0; Vf_cMTD = 80000.0; K_2cf_MTD = 2.0; Vr_cMTD = 600000.0Reaction: c_2cf => c_1cf + NADPH; c_1cf, c_2cf, c_1cf, c_2cf, Rate Law: cytosol*(Vf_cMTD*c_2cf/(K_2cf_MTD+c_2cf)-Vr_cMTD*c_1cf/(K_1cf_MTD+c_1cf))
K_gsh_GPX = 1330.0; K_H2O2_GPX = 0.09; Vm_GPX = 4500.0Reaction: c_gsh + H2O2 => c_gsg; H2O2, c_gsh, H2O2, c_gsh, Rate Law: cytosol*Vm_GPX*(c_gsh/(K_gsh_GPX+c_gsh))^2*H2O2/(K_H2O2_GPX+H2O2)
K_gshHb = 150.0; V_gshHb = 150.0Reaction: c_gsh => b_gsh; c_gsh, c_gsh, Rate Law: cytosol*V_gshHb*c_gsh/(K_gshHb+c_gsh)
k_out_coo = 100.0; k_in_coo = 100.0Reaction: m_coo => c_coo; c_coo, m_coo, c_coo, m_coo, Rate Law: k_in_coo*m_coo*mito/3-k_out_coo*c_coo*cytosol
K_gshLb = 3000.0; V_gshLb = 1100.0; h_gshLb = 3.0Reaction: c_gsh => b_gsh; c_gsh, c_gsh, Rate Law: cytosol*V_gshLb*c_gsh^h_gshLb/(K_gshLb^h_gshLb+c_gsh^h_gshLb)
V_cgly = 10000.0; V_mgly = 10000.0; K_mgly = 5700.0; K_cgly = 5700.0Reaction: m_gly => c_gly; c_gly, m_gly, c_gly, m_gly, Rate Law: V_mgly*m_gly/(K_mgly+m_gly)*mito*1/3-V_cgly*c_gly/(K_cgly+c_gly)*cytosol
Ka_GCS = 0.01; K_cys_GCS = 100.0; ssH2O2 = 0.01; Ke_GCS = 5597.0; Ki_GCS = 8200.0; K_glu_GCS = 1900.0; Kp_GCS = 300.0; Vm_GCS = 3600.0Reaction: c_cys + c_glu => glc; H2O2, c_gsh, H2O2, c_cys, c_glu, c_gsh, glc, H2O2, c_cys, c_glu, c_gsh, glc, Rate Law: cytosol*Vm_GCS*(c_cys*c_glu-glc/Ke_GCS)/(K_cys_GCS*K_glu_GCS+c_glu*K_cys_GCS+c_cys*(K_glu_GCS*(1+c_gsh/Ki_GCS)+c_glu)+glc/Kp_GCS+c_gsh/Ki_GCS)*(H2O2+Ka_GCS)/(ssH2O2+Ka_GCS)
Vm_cFTS = 3900.0; K_thf_cFTS = 3.0; K_coo_cFTS = 43.0Reaction: c_thf + c_coo => c_10f; c_coo, c_thf, c_coo, c_thf, Rate Law: cytosol*Vm_cFTS*c_thf*c_coo/((K_thf_cFTS+c_thf)*(K_coo_cFTS+c_coo))
K_glc_GS = 22.0; Vm_GS = 5400.0; Ke_GS = 5600.0; K_gly_GS = 300.0; Kp_GS = 30.0Reaction: glc + c_gly => c_gsh; c_gly, c_gsh, glc, c_gly, c_gsh, glc, Rate Law: cytosol*Vm_GS*(c_gly*glc-c_gsh/Ke_GS)/(K_gly_GS*K_glc_GS+glc*K_gly_GS+c_gly*(K_glc_GS+glc)+c_gsh/Kp_GS)
K_bglyc = 150.0; V_bglyc = 4600.0; k_out_gly = 1.0Reaction: b_gly => c_gly; b_gly, c_gly, b_gly, c_gly, Rate Law: cytosol*(V_bglyc*b_gly/(K_bglyc+b_gly)-k_out_gly*c_gly)
Vm_MTHFR = 5300.0; K_NADPH_MTHFR = 16.0; K_2cf_MTHFR = 50.0Reaction: c_2cf + NADPH => c_5mf; sah, sam, NADPH, c_2cf, sah, sam, NADPH, c_2cf, sah, sam, Rate Law: cytosol*Vm_MTHFR*c_2cf*NADPH/((K_2cf_MTHFR+c_2cf)*(K_NADPH_MTHFR+NADPH))*72/((10+sam)-sah)
k1=0.6Reaction: species_2 => ; species_2, species_2, Rate Law: cytosol*k1*species_2
V_oGly_b = 630.0Reaction: => b_gly, Rate Law: blood*V_oGly_b
K_gsg_GR = 107.0; Vm_GR = 892.5; K_NADPH_GR = 10.4Reaction: c_gsg + NADPH => c_gsh; NADPH, c_gsg, NADPH, c_gsg, Rate Law: cytosol*Vm_GR*c_gsg*NADPH/((K_gsg_GR+c_gsg)*(K_NADPH_GR+NADPH))
Kmdpao=50.0; Kmaspao=0.6; Kmspao=15.0; Vmpao=621.0; Kmadpao=14.0Reaction: species_6 => species_2; species_5, species_4, species_3, species_3, species_4, species_5, species_6, species_3, species_4, species_5, species_6, Rate Law: cytosol*Vmpao*species_6/(Kmadpao*(1+species_6/Kmadpao+species_5/Kmaspao+species_4/Kmdpao+species_3/Kmspao))

States:

NameDescription
species 9[coenzyme A]
met[methionine; Methionine]
c 1cf[644350]
glc[L-gamma-glutamyl-L-cysteine; gamma-L-Glutamyl-L-cysteine]
species 1[S-adenosylmethioninamine]
c gly[glycine; Glycine]
c thf[(6S)-5,6,7,8-tetrahydrofolic acid; Tetrahydrofolate]
species 4[spermidine]
m thf[(6S)-5,6,7,8-tetrahydrofolic acid; Tetrahydrofolate]
m coo[formate; Formate]
NADPH[NADPH; NADPH]
b gsg[glutathione disulfide; Glutathione disulfide]
b gly[glycine; Glycine]
c cys[cysteine; Cysteine]
cyt[834]
m 2cf[(6R)-5,10-methylenetetrahydrofolic acid; 5,10-Methylenetetrahydrofolate]
species 8[acetyl-CoA]
src[sarcosine; Sarcosine]
c glu[glutamic acid; Glutamate]
species 5[N(1)-acetylspermine]
b gsh[glutathione; Glutathione]
species 2[putrescine]
CO[carbon dioxide; CO2]
species 6[N(1)-acetylspermidine]
GAR[N(1)-(5-phospho-D-ribosyl)glycinamide; C003838]
c dhf[dihydrofolic acid; Dihydrofolate]
c gsh[glutathione; Glutathione]
c 2cf[(6R)-5,10-methylenetetrahydrofolic acid; 5,10-Methylenetetrahydrofolate]
aic[AICA ribonucleotide; 1-(5'-Phosphoribosyl)-5-amino-4-imidazolecarboxamide]
species 3[spermine]
c 10f[10-formyltetrahydrofolic acid; 10-Formyltetrahydrofolate]
m gly[glycine; Glycine]
c coo[formate; Formate]
species 7[ornithine]
c gsg[glutathione disulfide; Glutathione disulfide]

Reyes-Palomares2012 - a combined model hepatic polyamine and sulfur aminoacid metabolism - version1: BIOMD0000000674v0.0.1

Reyes-Palomares2012 - a combined model hepatic polyamine and sulfur aminoacid metabolism - version1Mammalian polyamine m…

Details

Many molecular details remain to be uncovered concerning the regulation of polyamine metabolism. A previous model of mammalian polyamine metabolism showed that S-adenosyl methionine availability could play a key role in polyamine homeostasis. To get a deeper insight in this prediction, we have built a combined model by integration of the previously published polyamine model and one-carbon and glutathione metabolism model, published by different research groups. The combined model is robust and it is able to achieve physiological steady-state values, as well as to reproduce the predictions of the individual models. Furthermore, a transition between two versions of our model with new regulatory factors added properly simulates the switch in methionine adenosyl transferase isozymes occurring when the liver enters in proliferative conditions. The combined model is useful to support the previous prediction on the role of S-adenosyl methionine availability in polyamine homeostasis. Furthermore, it could be easily adapted to get deeper insights on the connections of polyamines with energy metabolism. link: http://identifiers.org/pubmed/21814788

Parameters:

NameDescription
K_DUMP_TS = 6.3; Vm_TS = 5000.0; K_2cf_TS = 14.0Reaction: DUMP + c_2cf => c_dhf; DUMP, c_2cf, Rate Law: cytosol*Vm_TS*DUMP*c_2cf/((K_DUMP_TS+DUMP)*(K_2cf_TS+c_2cf))
Ki_MAT1 = 2140.0; Vm_MAT1 = 260.0; Km_MAT1 = 41.0Reaction: met => sam; c_gsg, met, sam, Rate Law: cytosol*Vm_MAT1*met/(Km_MAT1+met)*(0.23+0.8*exp((-0.0026)*sam))*(Ki_MAT1+66.71)/(Ki_MAT1+c_gsg)
Vm_PGT = 24300.0; K_GAR_PGT = 520.0; K_10f_PGT = 4.9Reaction: c_10f + GAR => aic + c_thf; GAR, c_10f, Rate Law: cytosol*Vm_PGT*c_10f*GAR/((K_10f_PGT+c_10f)*(K_GAR_PGT+GAR))
Km_MAT3 = 300.0; Ki_MAT3 = 4030.0; Ka_MAT3 = 360.0; Vm_MAT3 = 220.0Reaction: met => sam; c_gsg, met, sam, Rate Law: cytosol*Vm_MAT3*met^1.21/(Km_MAT3+met^1.21)*(1+7.2*sam^2/(Ka_MAT3^2+sam^2))*(Ki_MAT3+66.71)/(Ki_MAT3+c_gsg)
K_gly_GDC = 3400.0; Vm_GDC = 15000.0; K_thf_GDC = 50.0Reaction: m_thf + m_gly => m_2cf + CO; m_gly, m_thf, Rate Law: mito*Vm_GDC*m_thf*m_gly/((K_thf_GDC+m_thf)*(K_gly_GDC+m_gly))
K_aic_ART = 100.0; Vm_ART = 55000.0; K_10f_ART = 5.9Reaction: c_10f + aic => c_thf; aic, c_10f, Rate Law: cytosol*Vm_ART*c_10f*aic/((K_10f_ART+c_10f)*(K_aic_ART+aic))
ssH2O2 = 0.01; K_hcy_BHMT = 12.0; K_bet_BHMT = 100.0; Ki_BHMT = 0.01; Vm_BHMT = 2160.0Reaction: hcy + BET => met + dmg; H2O2, sah, sam, BET, hcy, Rate Law: cytosol*exp((-0.0021)*(sam+sah))*exp(0.0021*102.6)*Vm_BHMT*hcy*BET/((K_hcy_BHMT+hcy)*(K_bet_BHMT+BET))*(ssH2O2+Ki_BHMT)/(H2O2+Ki_BHMT)
Kisspms=25.0; Kdspms=60.0; Kaspms=0.1; Kiaspms=0.06; Vmspms=193.8Reaction: species_1 + species_4 => species_3; species_1, species_3, species_4, Rate Law: cytosol*Vmspms*species_1*species_4/(Kiaspms*Kdspms*(1+species_3/Kisspms)+Kdspms*species_1+Kaspms*(1+species_3/Kisspms)*species_4+species_1*species_4)
K_bserc = 150.0; k_out_ser = 1.0; V_bserc = 2700.0Reaction: b_ser => c_ser; b_ser, c_ser, Rate Law: cytosol*(V_bserc*b_ser/(K_bserc+b_ser)-k_out_ser*c_ser)
k1_mNE = 0.03; k2_mNE = 20.0Reaction: m_thf + HCHO => m_2cf; HCHO, m_2cf, m_thf, Rate Law: mito*(k1_mNE*m_thf*HCHO-k2_mNE*m_2cf)
K_thf_DMGD = 50.0; Vm_DMGD = 15000.0; K_dmg_DMGD = 50.0Reaction: m_thf + dmg => m_2cf + src; dmg, m_thf, Rate Law: mito*Vm_DMGD*m_thf*dmg/((K_thf_DMGD+m_thf)*(K_dmg_DMGD+dmg))
Ka_CBS = 0.035; ssH2O2 = 0.01; Vm_CBS = 420000.0; K_hcy_CBS = 1000.0; K_ser_CBS = 2000.0Reaction: hcy + c_ser => cyt; H2O2, sah, sam, c_ser, hcy, Rate Law: cytosol*Vm_CBS*hcy*c_ser/((K_hcy_CBS+hcy)*(K_ser_CBS+c_ser))*((30/102.59)^2+1)/((30/(sam+sah))^2+1)*(H2O2+Ka_CBS)/(ssH2O2+Ka_CBS)
K_cyt_CTGL = 500.0; Vm_CTGL = 1500.0Reaction: cyt => c_cys; cyt, Rate Law: cytosol*Vm_CTGL*cyt/(K_cyt_CTGL+cyt)
K_sam_GNMT = 63.0; Vm_GNMT = 260.0; K_gly_GNMT = 130.0; Ki_GNMT = 18.0Reaction: sam + c_gly => sah + src; c_5mf, c_gly, sah, sam, Rate Law: cytosol*Vm_GNMT*sam*c_gly/((K_sam_GNMT+sam)*(K_gly_GNMT+c_gly))*1/(1+sah/Ki_GNMT)*4.8/(0.35+c_5mf)
Ki_DNMT = 1.4; Vm_DNMT = 180.0; Km_DNMT = 1.4Reaction: sam => sah; sah, sam, Rate Law: cytosol*Vm_DNMT*sam/(Km_DNMT*(1+sah/Ki_DNMT)+sam)
k2_cNE = 22.0; k1_cNE = 0.03Reaction: c_thf + HCHO => c_2cf; HCHO, c_2cf, c_thf, Rate Law: cytosol*(k1_cNE*c_thf*HCHO-k2_cNE*c_2cf)
V_mser = 10000.0; V_cser = 10000.0; K_mser = 5700.0; K_cser = 5700.0Reaction: m_ser => c_ser; c_ser, m_ser, Rate Law: (V_mser*m_ser/(K_mser+m_ser)*mito/3-V_cser*c_ser/(K_cser+c_ser))*cytosol
K_src_SDH = 320.0; Vm_SDH = 15000.0; K_thf_SDH = 50.0Reaction: m_thf + src => m_2cf + m_gly; m_thf, src, Rate Law: mito*Vm_SDH*m_thf*src/((K_thf_SDH+m_thf)*(K_src_SDH+src))
K_ser_SHMT = 600.0; Vf_mSHMT = 11440.0; K_2cf_SHMT = 3200.0; K_gly_SHMT = 10000.0; K_thf_SHMT = 50.0; Vr_mSHMT = 3.0E7Reaction: m_thf + m_ser => m_gly + m_2cf; m_2cf, m_gly, m_ser, m_thf, Rate Law: mito*(Vf_mSHMT*m_thf*m_ser/((K_thf_SHMT+m_thf)*(K_ser_SHMT+m_ser))-Vr_mSHMT*m_gly*m_2cf/((K_gly_SHMT+m_gly)*(K_2cf_SHMT+m_2cf)))
K_dhf_DHFR = 0.5; Vm_DHFR = 2000.0; K_NADPH_DHFR = 4.0Reaction: c_dhf + NADPH => c_thf; NADPH, c_dhf, Rate Law: cytosol*Vm_DHFR*c_dhf*NADPH/((K_dhf_DHFR+c_dhf)*(K_NADPH_DHFR+NADPH))
Kiasamdc=2.5; Kapsamdc=0.5; parameter_3 = 21.1340139923629; Kmsamdc=50.0; Kissamdc=500.0Reaction: sam => species_1; species_3, species_2, sam, species_1, Rate Law: cytosol*parameter_3/(1+species_3/Kissamdc)*sam/(Kmsamdc*(1+Kapsamdc/species_2+species_1/Kiasamdc)+sam)
V_gsgHb = 40.0; K_gsgHb = 1250.0Reaction: c_gsg => b_gsg; c_gsg, Rate Law: cytosol*V_gsgHb*c_gsg/(K_gsgHb+c_gsg)
Vf_mFTS = 2000.0; Vr_mFTS = 6300.0; K_coo_mFTS = 43.0; K_thf_mFTS = 3.0; K_10f_mFTS = 22.0Reaction: m_thf + m_coo => m_10f; m_10f, m_coo, m_thf, Rate Law: mito*(Vf_mFTS*m_thf*m_coo/((K_thf_mFTS+m_thf)*(K_coo_mFTS+m_coo))-Vr_mFTS*m_10f/(K_10f_mFTS+m_10f))
K_10f_MTCH = 100.0; Vr_MTCH = 20000.0; Vf_cMTCH = 500000.0; K_1cf_MTCH = 250.0Reaction: c_1cf => c_10f; c_10f, c_1cf, Rate Law: cytosol*(Vf_cMTCH*c_1cf/(K_1cf_MTCH+c_1cf)-Vr_MTCH*c_10f/(K_10f_MTCH+c_10f))
Vmspds=657.0; Kiaspds=0.8; KaSpds=0.3; Kpspds=40.0; Kidspds=100.0Reaction: species_1 + species_2 => species_4; species_1, species_2, species_4, Rate Law: cytosol*Vmspds*species_1*species_2/(Kiaspds*Kpspds*(1+species_4/Kidspds)+Kpspds*species_1+KaSpds*(1+species_4/Kidspds)*species_2+species_1*species_2)
Kmaccoassat=1.5; Kmdssat=130.0; C=4.44; Kmsssat=35.0; parameter_2 = 42.2853792055417; Kmcoassat=40.0Reaction: species_3 + species_8 => species_5 + species_9; species_4, species_3, species_8, species_9, Rate Law: cytosol*1/C*parameter_2*species_3*species_8/(Kmsssat*(1+species_4/Kmdssat)*Kmaccoassat*(1+species_9/Kmcoassat)+Kmaccoassat*(1+species_9/Kmcoassat)*species_3+Kmsssat*(1+species_4/Kmdssat)*species_8+species_3*species_8)
V_gsgLb = 4025.0; K_gsgLb = 7100.0Reaction: c_gsg => b_gsg; c_gsg, Rate Law: cytosol*V_gsgLb*c_gsg/(K_gsgLb+c_gsg)
Kmaccoassat=1.5; Kmdssat=130.0; Kmsssat=35.0; parameter_2 = 42.2853792055417; Kmcoassat=40.0Reaction: species_4 + species_8 => species_6 + species_9; species_3, species_4, species_8, species_9, Rate Law: cytosol*parameter_2*species_4*species_8/(Kmdssat*(1+species_3/Kmsssat)*Kmaccoassat*(1+species_9/Kmcoassat)+Kmaccoassat*(1+species_9/Kmcoassat)*species_4+Kmdssat*(1+species_3/Kmsssat)*species_8+species_4*species_8)
ssH2O2 = 0.01; Ki_MS = 0.01; K_5mf_MS = 25.0; Vm_MS = 500.0; K_hcy_MS = 1.0Reaction: c_5mf + hcy => c_thf + met; H2O2, c_5mf, hcy, Rate Law: cytosol*Vm_MS*c_5mf*hcy/((K_5mf_MS+c_5mf)*(K_hcy_MS+hcy))*(ssH2O2+Ki_MS)/(H2O2+Ki_MS)
K_hcy_SAHH = 150.0; K_sah_SAHH = 6.5; Vf_SAHH = 320.0; Vr_SAHH = 4530.0Reaction: sah => hcy; hcy, sah, Rate Law: cytosol*(Vf_SAHH*sah/(K_sah_SAHH+sah)-Vr_SAHH*hcy/(K_hcy_SAHH+hcy))
K_ser_SHMT = 600.0; K_2cf_SHMT = 3200.0; Vr_cSHMT = 1.5E7; Vf_cSHMT = 5200.0; K_gly_SHMT = 10000.0; K_thf_SHMT = 50.0Reaction: c_ser + c_thf => c_gly + c_2cf; c_2cf, c_gly, c_ser, c_thf, Rate Law: cytosol*(Vf_cSHMT*c_thf*c_ser/((K_thf_SHMT+c_thf)*(K_ser_SHMT+c_ser))-Vr_cSHMT*c_gly*c_2cf/((K_gly_SHMT+c_gly)*(K_2cf_SHMT+c_2cf)))
Kmodc=60.0; Kipodc=1300.0; parameter_1 = 72.2557178994351Reaction: species_7 => species_2; species_2, species_7, Rate Law: cytosol*parameter_1*species_7/(Kmodc*(1+species_2/Kipodc)+species_7)
K_1cf_MTD = 10.0; Vf_cMTD = 80000.0; K_2cf_MTD = 2.0; Vr_cMTD = 600000.0Reaction: c_2cf => c_1cf + NADPH; c_1cf, c_2cf, Rate Law: cytosol*(Vf_cMTD*c_2cf/(K_2cf_MTD+c_2cf)-Vr_cMTD*c_1cf/(K_1cf_MTD+c_1cf))
V_bmetc = 913.4; K_bmetc = 150.0; k_out_met = 1.0Reaction: b_met => met; b_met, met, Rate Law: cytosol*(V_bmetc*b_met/(K_bmetc+b_met)-k_out_met*met)
K_gsh_GPX = 1330.0; K_H2O2_GPX = 0.09; Vm_GPX = 4500.0Reaction: c_gsh + H2O2 => c_gsg; H2O2, c_gsh, Rate Law: cytosol*Vm_GPX*(c_gsh/(K_gsh_GPX+c_gsh))^2*H2O2/(K_H2O2_GPX+H2O2)
k_out_coo = 100.0; k_in_coo = 100.0Reaction: m_coo => c_coo; c_coo, m_coo, Rate Law: k_in_coo*m_coo*mito/3-k_out_coo*c_coo*cytosol
b_ser_basal = 150.0; aa_input = 0.25Reaction: b_ser = aa_input*b_ser_basal, Rate Law: missing
K_gshLb = 3000.0; V_gshLb = 1100.0; h_gshLb = 3.0Reaction: c_gsh => b_gsh; c_gsh, Rate Law: cytosol*V_gshLb*c_gsh^h_gshLb/(K_gshLb^h_gshLb+c_gsh^h_gshLb)
K_gshHb = 150.0; V_gshHb = 150.0Reaction: c_gsh => b_gsh; c_gsh, Rate Law: cytosol*V_gshHb*c_gsh/(K_gshHb+c_gsh)
V_cgly = 10000.0; V_mgly = 10000.0; K_mgly = 5700.0; K_cgly = 5700.0Reaction: m_gly => c_gly; c_gly, m_gly, Rate Law: V_mgly*m_gly/(K_mgly+m_gly)*mito*1/3-V_cgly*c_gly/(K_cgly+c_gly)*cytosol
Ka_GCS = 0.01; K_cys_GCS = 100.0; ssH2O2 = 0.01; Ke_GCS = 5597.0; Ki_GCS = 8200.0; K_glu_GCS = 1900.0; Kp_GCS = 300.0; Vm_GCS = 3600.0Reaction: c_cys + c_glu => glc; H2O2, c_gsh, c_cys, c_glu, glc, Rate Law: cytosol*Vm_GCS*(c_cys*c_glu-glc/Ke_GCS)/(K_cys_GCS*K_glu_GCS+c_glu*K_cys_GCS+c_cys*(K_glu_GCS*(1+c_gsh/Ki_GCS)+c_glu)+glc/Kp_GCS+c_gsh/Ki_GCS)*(H2O2+Ka_GCS)/(ssH2O2+Ka_GCS)
Vm_cFTS = 3900.0; K_thf_cFTS = 3.0; K_coo_cFTS = 43.0Reaction: c_thf + c_coo => c_10f; c_coo, c_thf, Rate Law: cytosol*Vm_cFTS*c_thf*c_coo/((K_thf_cFTS+c_thf)*(K_coo_cFTS+c_coo))
K_glc_GS = 22.0; Vm_GS = 5400.0; Ke_GS = 5600.0; K_gly_GS = 300.0; Kp_GS = 30.0Reaction: glc + c_gly => c_gsh; c_gly, c_gsh, glc, Rate Law: cytosol*Vm_GS*(c_gly*glc-c_gsh/Ke_GS)/(K_gly_GS*K_glc_GS+glc*K_gly_GS+c_gly*(K_glc_GS+glc)+c_gsh/Kp_GS)
K_bglyc = 150.0; V_bglyc = 4600.0; k_out_gly = 1.0Reaction: b_gly => c_gly; b_gly, c_gly, Rate Law: cytosol*(V_bglyc*b_gly/(K_bglyc+b_gly)-k_out_gly*c_gly)
Vm_MTHFR = 5300.0; K_NADPH_MTHFR = 16.0; K_2cf_MTHFR = 50.0Reaction: c_2cf + NADPH => c_5mf; sah, sam, NADPH, c_2cf, Rate Law: cytosol*Vm_MTHFR*c_2cf*NADPH/((K_2cf_MTHFR+c_2cf)*(K_NADPH_MTHFR+NADPH))*72/((10+sam)-sah)
k1=0.6Reaction: species_2 => ; species_2, Rate Law: cytosol*k1*species_2
b_met_basal = 30.0; aa_input = 0.25Reaction: b_met = aa_input*b_met_basal, Rate Law: missing
K_gsg_GR = 107.0; Vm_GR = 892.5; K_NADPH_GR = 10.4Reaction: c_gsg + NADPH => c_gsh; NADPH, c_gsg, Rate Law: cytosol*Vm_GR*c_gsg*NADPH/((K_gsg_GR+c_gsg)*(K_NADPH_GR+NADPH))
Kmdpao=50.0; Kmaspao=0.6; Kmspao=15.0; Vmpao=621.0; Kmadpao=14.0Reaction: species_6 => species_2; species_5, species_4, species_3, species_6, Rate Law: cytosol*Vmpao*species_6/(Kmadpao*(1+species_6/Kmadpao+species_5/Kmaspao+species_4/Kmdpao+species_3/Kmspao))

States:

NameDescription
sam[S-Adenosyl-L-methionine; S-adenosyl-L-methionine]
met[methionine; Methionine]
c 1cf[644350]
glc[gamma-L-Glutamyl-L-cysteine; L-gamma-glutamyl-L-cysteine]
species 1[S-adenosylmethioninamine]
c gly[Glycine; glycine]
c thf[(6S)-5,6,7,8-tetrahydrofolic acid; Tetrahydrofolate]
species 4[spermidine]
m coo[formate; Formate]
m thf[(6S)-5,6,7,8-tetrahydrofolic acid; Tetrahydrofolate]
b met[Methionine; methionine]
NADPH[NADPH; NADPH]
b gsg[glutathione disulfide; Glutathione disulfide]
b gly[glycine; Glycine]
b cys[Cysteine; cysteine]
c cys[cysteine; Cysteine]
cyt[834]
m 2cf[5,10-Methylenetetrahydrofolate; (6R)-5,10-methylenetetrahydrofolic acid]
src[Sarcosine; sarcosine]
species 5[N(1)-acetylspermine]
b gsh[Glutathione; glutathione]
species 2[putrescine]
hcy[homocysteine; Homocysteine]
species 6[N(1)-acetylspermidine]
m ser[serine; Serine]
c ser[serine; Serine]
b glu[glutamic acid; Glutamate]
c gsh[Glutathione; glutathione]
c 5mf[5-methyltetrahydrofolic acid; 5-Methyltetrahydrofolate]
sah[S-adenosyl-L-homocysteine; S-Adenosyl-L-homocysteine]
c 2cf[(6R)-5,10-methylenetetrahydrofolic acid; 5,10-Methylenetetrahydrofolate]
species 3[spermine]
c 10f[10-formyltetrahydrofolic acid; 10-Formyltetrahydrofolate]
m gly[Glycine; glycine]
c coo[Formate; formate]
c gsg[glutathione disulfide; Glutathione disulfide]
b ser[serine; Serine]
HCHO[formaldehyde; Formaldehyde]

Rhodes2019 - Immune-Mediated theory of Metastasis: BIOMD0000000926v0.0.1

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

Details

Accumulating experimental and clinical evidence suggest that the immune response to cancer is not exclusively anti-tumor. Indeed, the pro-tumor roles of the immune system - as suppliers of growth and pro-angiogenic factors or defenses against cytotoxic immune attacks, for example - have been long appreciated, but relatively few theoretical works have considered their effects. Inspired by the recently proposed "immune-mediated" theory of metastasis, we develop a mathematical model for tumor-immune interactions at two anatomically distant sites, which includes both anti- and pro-tumor immune effects, and the experimentally observed tumor-induced phenotypic plasticity of immune cells (tumor "education" of the immune cells). Upon confrontation of our model to experimental data, we use it to evaluate the implications of the immune-mediated theory of metastasis. We find that tumor education of immune cells may explain the relatively poor performance of immunotherapies, and that many metastatic phenomena, including metastatic blow-up, dormancy, and metastasis to sites of injury, can be explained by the immune-mediated theory of metastasis. Our results suggest that further work is warranted to fully elucidate the pro-tumor effects of the immune system in metastatic cancer. link: http://identifiers.org/doi/10.1101/565531

Parameters:

NameDescription
alpha_2 = 1000000.0 1/dReaction: => CT_immune_Cell_x_2, Rate Law: compartment*alpha_2
lambda_2 = 3.25514658385093E-18Reaction: => CT_immune_Cell_x_2, Rate Law: compartment*lambda_2*CT_immune_Cell_x_2
f_2 = 2.5E-19Reaction: => TE_immune_Cell_y_2, Rate Law: compartment*f_2*TE_immune_Cell_y_2
g_1 = 0.379999999285661; gamma_1 = 1.00000000000479Reaction: => Tumor_Cell_u_1, Rate Law: compartment*gamma_1*g_1*Tumor_Cell_u_1
omega_1 = 0.59Reaction: CT_immune_Cell_x_1 =>, Rate Law: compartment*omega_1*CT_immune_Cell_x_1
rho_1 = 0.001Reaction: CT_immune_Cell_x_1 => ; Tumor_Cell_u_1, Rate Law: compartment*rho_1*Tumor_Cell_u_1*CT_immune_Cell_x_1
g_2 = 0.38; gamma_2 = 1.0Reaction: => Tumor_Cell_u_2, Rate Law: compartment*gamma_2*g_2*Tumor_Cell_u_2
s_1 = 0.01; est = 5.57430539315428E-11Reaction: => Tumor_Cell_u_2; Tumor_Cell_u_1, Rate Law: compartment*s_1*Tumor_Cell_u_1*est
theta_2 = 65.67; sigma_2 = 0.299993171807403Reaction: => Necrotic_Cell_v_2; Tumor_Cell_u_2, Rate Law: compartment*theta_2*sigma_2*Tumor_Cell_u_2
lambda_1 = 8.13659542487314E-6Reaction: => CT_immune_Cell_x_1, Rate Law: compartment*lambda_1*CT_immune_Cell_x_1
sigma_1 = 0.299993171804526Reaction: Tumor_Cell_u_1 =>, Rate Law: compartment*sigma_1*Tumor_Cell_u_1
f_1 = 2.49998437509766E-7Reaction: => TE_immune_Cell_y_1, Rate Law: compartment*f_1*TE_immune_Cell_y_1
alpha_1 = 1000000.0 1/dReaction: => CT_immune_Cell_x_1, Rate Law: compartment*alpha_1
ed_2 = 5.0E-17Reaction: CT_immune_Cell_x_2 => TE_immune_Cell_y_2, Rate Law: compartment*ed_2*CT_immune_Cell_x_2
ed_1 = 5.0E-5Reaction: CT_immune_Cell_x_1 => TE_immune_Cell_y_1, Rate Law: compartment*ed_1*CT_immune_Cell_x_1
myu_2 = 0.05Reaction: Necrotic_Cell_v_2 =>, Rate Law: compartment*myu_2*Necrotic_Cell_v_2
tau_2 = 0.05 1/dReaction: TE_immune_Cell_y_2 => ; TE_immune_Cell_y_1, Rate Law: compartment*tau_2*TE_immune_Cell_y_2
omega_2 = 0.59Reaction: CT_immune_Cell_x_2 =>, Rate Law: compartment*omega_2*CT_immune_Cell_x_2
p = 1.0E-4; tilde_s_1 = 0.05 1/dReaction: => TE_immune_Cell_y_2; TE_immune_Cell_y_1, Rate Law: compartment*p*tilde_s_1*TE_immune_Cell_y_1
tilde_s_1 = 0.05 1/dReaction: TE_immune_Cell_y_1 =>, Rate Law: compartment*tilde_s_1*TE_immune_Cell_y_1
sigma_2 = 0.299993171807403Reaction: Tumor_Cell_u_2 =>, Rate Law: compartment*sigma_2*Tumor_Cell_u_2
sigma_1 = 0.299993171804526; theta_1 = 65.67Reaction: => Necrotic_Cell_v_1; Tumor_Cell_u_1, Rate Law: compartment*theta_1*sigma_1*Tumor_Cell_u_1
myu_1 = 0.01Reaction: Necrotic_Cell_v_1 =>, Rate Law: compartment*myu_1*Necrotic_Cell_v_1
tau_1 = 0.05 1/dReaction: TE_immune_Cell_y_1 =>, Rate Law: compartment*tau_1*TE_immune_Cell_y_1
rho_2 = 0.01Reaction: CT_immune_Cell_x_2 => ; Tumor_Cell_u_2, Rate Law: compartment*rho_2*Tumor_Cell_u_2*CT_immune_Cell_x_2
s_1 = 0.01Reaction: Tumor_Cell_u_1 =>, Rate Law: compartment*s_1*Tumor_Cell_u_1

States:

NameDescription
TE immune Cell y 2[C4968]
CT immune Cell x 1[C12543; cancer]
CT immune Cell x 2[C4968; C12543]
Necrotic Cell v 2[C36123; C4968]
TE immune Cell y 1[cancer]
Tumor Cell u 2[BTO:0006256; C4968]
Tumor Cell u 1[cancer; BTO:0006256]
Necrotic Cell v 1[cancer; C36123]

Ribba2012 - Low-grade gliomas, tumour growth inhibition model: BIOMD0000000521v0.0.1

Ribba2012 - Low-grade gliomas, tumour growth inhibition modelUsing longitudinal mean tumour diameter (MTD) data, this mo…

Details

PURPOSE: To develop a tumor growth inhibition model for adult diffuse low-grade gliomas (LGG) able to describe tumor size evolution in patients treated with chemotherapy or radiotherapy. EXPERIMENTAL DESIGN: Using longitudinal mean tumor diameter (MTD) data from 21 patients treated with first-line procarbazine, 1-(2-chloroethyl)-3-cyclohexyl-l-nitrosourea, and vincristine (PCV) chemotherapy, we formulated a model consisting of a system of differential equations, incorporating tumor-specific and treatment-related parameters that reflect the response of proliferative and quiescent tumor tissue to treatment. The model was then applied to the analysis of longitudinal tumor size data in 24 patients treated with first-line temozolomide (TMZ) chemotherapy and in 25 patients treated with first-line radiotherapy. RESULTS: The model successfully described the MTD dynamics of LGG before, during, and after PCV chemotherapy. Using the same model structure, we were also able to successfully describe the MTD dynamics in LGG patients treated with TMZ chemotherapy or radiotherapy. Tumor-specific parameters were found to be consistent across the three treatment modalities. The model is robust to sensitivity analysis, and preliminary results suggest that it can predict treatment response on the basis of pretreatment tumor size data. CONCLUSIONS: Using MTD data, we propose a tumor growth inhibition model able to describe LGG tumor size evolution in patients treated with chemotherapy or radiotherapy. In the future, this model might be used to predict treatment efficacy in LGG patients and could constitute a rational tool to conceive more effective chemotherapy schedules. link: http://identifiers.org/pubmed/22761472

Parameters:

NameDescription
gamma = 0.729; k_PQ = 0.00295; KDE = 0.24Reaction: Q = k_PQ-gamma*C*KDE*Q, Rate Law: k_PQ-gamma*C*KDE*Q
gamma = 0.729; delta_QP = 0.0087; KDE = 0.24; k_Qp_P = 0.0031Reaction: Qp = (gamma*C*KDE*Q-k_Qp_P*Qp)-delta_QP*Qp, Rate Law: (gamma*C*KDE*Q-k_Qp_P*Qp)-delta_QP*Qp
gamma = 0.729; Pstar = 0.0; k_PQ = 0.00295; K = 100.0; lambda_P = 0.121; KDE = 0.24; k_Qp_P = 0.0031Reaction: P = ((lambda_P*P*(1-Pstar/K)+k_Qp_P*Qp)-k_PQ*P)-gamma*C*KDE*P, Rate Law: ((lambda_P*P*(1-Pstar/K)+k_Qp_P*Qp)-k_PQ*P)-gamma*C*KDE*P
KDE = 0.24Reaction: C = (-KDE)*C, Rate Law: (-KDE)*C

States:

NameDescription
Q[Portion of tissue; inactive]
P[Portion of tissue; proliferative]
C[procarbazine; vincristine; CHEMBL514; blood plasma]
Qp[damage; inactive; Portion of tissue]

Ribba2018 - Mathematical Model of Tumor Uptake for Immunocytokine-Based Cancer Immunotherapy: MODEL1909050002v0.0.1

This is a model developed to predict concentrations of cergutuzumab amunaleukin (CEA-IL2v) after various systemic dosing…

Details

Purpose: Optimal dosing is critical for immunocytokine-based cancer immunotherapy to maximize efficacy and minimize toxicity. Cergutuzumab amunaleukin (CEA-IL2v) is a novel CEA-targeted immunocytokine. We set out to develop a mathematical model to predict intratumoral CEA-IL2v concentrations following various systemic dosing intensities.Experimental Design: Sequential measurements of CEA-IL2v plasma concentrations in 74 patients with solid tumors were applied in a series of differential equations to devise a model that also incorporates the peripheral concentrations of IL2 receptor-positive cell populations (i.e., CD8+, CD4+, NK, and B cells), which affect tumor bioavailability of CEA-IL2v. Imaging data from a subset of 14 patients were subsequently utilized to additionally predict antibody uptake in tumor tissues.Results: We created a pharmacokinetic/pharmacodynamic mathematical model that incorporates the expansion of IL2R-positive target cells at multiple dose levels and different schedules of CEA-IL2v. Model-based prediction of drug levels correlated with the concentration of IL2R-positive cells in the peripheral blood of patients. The pharmacokinetic model was further refined and extended by adding a model of antibody uptake, which is based on drug dose and the biological properties of the tumor. In silico predictions of our model correlated with imaging data and demonstrated that a dose-dense schedule comprising escalating doses and shortened intervals of drug administration can improve intratumoral drug uptake and overcome consumption of CEA-IL2v by the expanding population of IL2R-positive cells.Conclusions: The model presented here allows simulation of individualized treatment plans for optimal dosing and scheduling of immunocytokines for anticancer immunotherapy. Clin Cancer Res; 24(14); 3325-33. ©2018 AACRSee related commentary by Ruiz-Cerdá et al., p. 3236. link: http://identifiers.org/pubmed/29463551

Richards2016 - Genome-scale metabolic reconstruction of Methanococcus maripaludis (iMR539): MODEL1607200000v0.0.1

Richards2016 - Genome-scale metabolic reconstruction of Methanococcus maripaludis (iMR539)This model is described in the…

Details

Hydrogenotrophic methanogenesis occurs in multiple environments, ranging from the intestinal tracts of animals to anaerobic sediments and hot springs. Energy conservation in hydrogenotrophic methanogens was long a mystery; only within the last decade was it reported that net energy conservation for growth depends on electron bifurcation. In this work, we focus on Methanococcus maripaludis, a well-studied hydrogenotrophic marine methanogen. To better understand hydrogenotrophic methanogenesis and compare it with methylotrophic methanogenesis that utilizes oxidative phosphorylation rather than electron bifurcation, we have built iMR539, a genome scale metabolic reconstruction that accounts for 539 of the 1,722 protein-coding genes of M. maripaludis strain S2. Our reconstructed metabolic network uses recent literature to not only represent the central electron bifurcation reaction but also incorporate vital biosynthesis and assimilation pathways, including unique cofactor and coenzyme syntheses. We show that our model accurately predicts experimental growth and gene knockout data, with 93% accuracy and a Matthews correlation coefficient of 0.78. Furthermore, we use our metabolic network reconstruction to probe the implications of electron bifurcation by showing its essentiality, as well as investigating the infeasibility of aceticlastic methanogenesis in the network. Additionally, we demonstrate a method of applying thermodynamic constraints to a metabolic model to quickly estimate overall free-energy changes between what comes in and out of the cell. Finally, we describe a novel reconstruction-specific computational toolbox we created to improve usability. Together, our results provide a computational network for exploring hydrogenotrophic methanogenesis and confirm the importance of electron bifurcation in this process.Understanding and applying hydrogenotrophic methanogenesis is a promising avenue for developing new bioenergy technologies around methane gas. Although a significant portion of biological methane is generated through this environmentally ubiquitous pathway, existing methanogen models portray the more traditional energy conservation mechanisms that are found in other methanogens. We have constructed a genome scale metabolic network of Methanococcus maripaludis that explicitly accounts for all major reactions involved in hydrogenotrophic methanogenesis. Our reconstruction demonstrates the importance of electron bifurcation in central metabolism, providing both a window into hydrogenotrophic methanogenesis and a hypothesis-generating platform to fuel metabolic engineering efforts. link: http://identifiers.org/pubmed/27736793

Rienksma2014 - Genome-scale constraint-based metabolic model of M.tuberculosis: MODEL1411110000v0.0.1

Model created from Excel spreadsheet: sMtb2.xls

Details

Systems-level metabolic network reconstructions and the derived constraint-based (CB) mathematical models are efficient tools to explore bacterial metabolism. Approximately one-fourth of the Mycobacterium tuberculosis (Mtb) genome contains genes that encode proteins directly involved in its metabolism. These represent potential drug targets that can be systematically probed with CB models through the prediction of genes essential (or the combination thereof) for the pathogen to grow. However, gene essentiality depends on the growth conditions and, so far, no in vitro model precisely mimics the host at the different stages of mycobacterial infection, limiting model predictions. These limitations can be circumvented by combining expression data from in vivo samples with a validated CB model, creating an accurate description of pathogen metabolism in the host. To this end, we present here a thoroughly curated and extended genome-scale CB metabolic model of Mtb quantitatively validated using 13C measurements. We describe some of the efforts made in integrating CB models and high-throughput data to generate condition specific models, and we will discuss challenges ahead. This knowledge and the framework herein presented will enable to identify potential new drug targets, and will foster the development of optimal therapeutic strategies. link: http://identifiers.org/doi/10.1016/j.smim.2014.09.013

Risso2009 - Genome-scale metabolic network of Rhodoferax ferrireducens (iCR744): MODEL1507180024v0.0.1

Risso2009 - Genome-scale metabolic network of Rhodoferax ferrireducens (iCR744)This model is described in the article:…

Details

BACKGROUND: Rhodoferax ferrireducens is a metabolically versatile, Fe(III)-reducing, subsurface microorganism that is likely to play an important role in the carbon and metal cycles in the subsurface. It also has the unique ability to convert sugars to electricity, oxidizing the sugars to carbon dioxide with quantitative electron transfer to graphite electrodes in microbial fuel cells. In order to expand our limited knowledge about R. ferrireducens, the complete genome sequence of this organism was further annotated and then the physiology of R. ferrireducens was investigated with a constraint-based, genome-scale in silico metabolic model and laboratory studies. RESULTS: The iterative modeling and experimental approach unveiled exciting, previously unknown physiological features, including an expanded range of substrates that support growth, such as cellobiose and citrate, and provided additional insights into important features such as the stoichiometry of the electron transport chain and the ability to grow via fumarate dismutation. Further analysis explained why R. ferrireducens is unable to grow via photosynthesis or fermentation of sugars like other members of this genus and uncovered novel genes for benzoate metabolism. The genome also revealed that R. ferrireducens is well-adapted for growth in the subsurface because it appears to be capable of dealing with a number of environmental insults, including heavy metals, aromatic compounds, nutrient limitation and oxidative stress. CONCLUSION: This study demonstrates that combining genome-scale modeling with the annotation of a new genome sequence can guide experimental studies and accelerate the understanding of the physiology of under-studied yet environmentally relevant microorganisms. link: http://identifiers.org/pubmed/19772637

Roberts2010 - Genome-scale metabolic network of Clostridium thermocellum (iSR432): MODEL1507180004v0.0.1

Roberts2010 - Genome-scale metabolic network of Clostridium thermocellum (iSR432)This model is described in the article:…

Details

BACKGROUND: Microorganisms possess diverse metabolic capabilities that can potentially be leveraged for efficient production of biofuels. Clostridium thermocellum (ATCC 27405) is a thermophilic anaerobe that is both cellulolytic and ethanologenic, meaning that it can directly use the plant sugar, cellulose, and biochemically convert it to ethanol. A major challenge in using microorganisms for chemical production is the need to modify the organism to increase production efficiency. The process of properly engineering an organism is typically arduous. RESULTS: Here we present a genome-scale model of C. thermocellum metabolism, iSR432, for the purpose of establishing a computational tool to study the metabolic network of C. thermocellum and facilitate efforts to engineer C. thermocellum for biofuel production. The model consists of 577 reactions involving 525 intracellular metabolites, 432 genes, and a proteomic-based representation of a cellulosome. The process of constructing this metabolic model led to suggested annotation refinements for 27 genes and identification of areas of metabolism requiring further study. The accuracy of the iSR432 model was tested using experimental growth and by-product secretion data for growth on cellobiose and fructose. Analysis using this model captures the relationship between the reduction-oxidation state of the cell and ethanol secretion and allowed for prediction of gene deletions and environmental conditions that would increase ethanol production. CONCLUSIONS: By incorporating genomic sequence data, network topology, and experimental measurements of enzyme activities and metabolite fluxes, we have generated a model that is reasonably accurate at predicting the cellular phenotype of C. thermocellum and establish a strong foundation for rational strain design. In addition, we are able to draw some important conclusions regarding the underlying metabolic mechanisms for observed behaviors of C. thermocellum and highlight remaining gaps in the existing genome annotations. link: http://identifiers.org/pubmed/20307315

Robertson-Tessi M 2012 A model of tumor Immune interaction: BIOMD0000000731v0.0.1

Its a mathematical model presenting the interaction between a growing tumor and immune system. Model involves tumor cell…

Details

A mathematical model of the interactions between a growing tumor and the immune system is presented. The equations and parameters of the model are based on experimental and clinical results from published studies. The model includes the primary cell populations involved in effector T-cell mediated tumor killing: regulatory T cells, helper T cells, and dendritic cells. A key feature is the inclusion of multiple mechanisms of immunosuppression through the main cytokines and growth factors mediating the interactions between the cell populations. Decreased access of effector cells to the tumor interior with increasing tumor size is accounted for. The model is applied to tumors with different growth rates and antigenicities to gauge the relative importance of various immunosuppressive mechanisms. The most important factors leading to tumor escape are TGF-β-induced immunosuppression, conversion of helper T cells into regulatory T cells, and the limitation of immune cell access to the full tumor at large tumor sizes. The results suggest that for a given tumor growth rate, there is an optimal antigenicity maximizing the response of the immune system. Further increases in antigenicity result in increased immunosuppression, and therefore a decrease in tumor killing rate. This result may have implications for immunotherapies which modulate the effective antigenicity. Simulation of dendritic cell therapy with the model suggests that for some tumors, there is an optimal dose of transfused dendritic cells. link: http://identifiers.org/pubmed/22051568

Parameters:

NameDescription
alpha5 = 5.1 1/ms; k4 = 0.33 1Reaction: Pool => sl_TRegs; Mr, M, l_DC, Rate Law: MISC*alpha5*Mr/(1+k4*M/l_DC)
C1 = 0.3 pg/l; alpha6 = 2.1 1/msReaction: Pool => func_TRegs; IL2, sl_TRegs, Rate Law: MISC*alpha6*sl_TRegs*IL2/(C1+IL2)
deltaR = 0.1 1/msReaction: func_TRegs => Sink, Rate Law: MISC*deltaR*func_TRegs
R1 = 2.0E7; Tx = 0.999999666666889; I1 = 0.4 pg/l; alpha = 6.31E-5 1/msReaction: Pool => ul_DC; IL10, func_TRegs, Rate Law: MISC*alpha*Tx/((1+IL10/I1)*(1+func_TRegs/R1))
t1 = 0.05 ksReaction: IL10 => Sink, Rate Law: MISC*IL10/t1
lambda = 0.5 1/msReaction: ul_DC => l_DC; Mh, Rate Law: MISC*lambda*ul_DC/(1+ul_DC/Mh)
alpha4 = 1.9 1/ms; C1 = 0.3 pg/l; S2 = 2.9 pg/lReaction: Pool => func_CD4_HTC; sl_CD4_HTC, IL2, TGFb, Rate Law: MISC*alpha4*sl_CD4_HTC*IL2/((1+TGFb/S2)*(C1+IL2))
alpha3 = 9.9 1/ms; k4 = 0.33 1Reaction: Pool => sl_CD4_HTC; Mh, M, ul_DC, l_DC, Rate Law: MISC*alpha3*Mh/(1+k4*M/(ul_DC+l_DC))
C1 = 0.3 pg/l; S2 = 2.9 pg/l; alpha2 = 16.0 1/msReaction: Pool => func_CD8_ETC; sl_CD8_ETC, IL2, TGFb, Rate Law: MISC*alpha2*sl_CD8_ETC*IL2/((1+TGFb/S2)*(C1+IL2))
p3 = 1.4E-8; p4 = 1.3E-10; Tx = 0.999999666666889Reaction: Pool => IL10; func_TRegs, Tumorcells, Rate Law: MISC*(p3*func_TRegs+p4*Tx)
pc = 1.7E-5; S4 = 0.9 pg/l; I2 = 0.75 pg/lReaction: Pool => IL2; sl_CD4_HTC, TGFb, IL10, Rate Law: MISC*pc*sl_CD4_HTC/((1+TGFb/S4)*(1+IL10/I2))
Tx = 0.999999666666889; p2 = 1.1E-7; p1 = 1.8E-8Reaction: Pool => TGFb; func_TRegs, Tumorcells, Rate Law: MISC*(p1*func_TRegs+p2*Tx)
deltaE = 1.0 1/msReaction: func_CD8_ETC => Sink, Rate Law: MISC*deltaE*func_CD8_ETC
k4 = 0.33 1; alpha1 = 23.0 1/msReaction: Pool => sl_CD8_ETC; Me, M, l_DC, Rate Law: MISC*alpha1*Me/(1+k4*M/l_DC)
ts = 0.07 ksReaction: TGFb => Sink, Rate Law: MISC*TGFb/ts
deltaD = 0.5 1/msReaction: l_DC => Sink, Rate Law: MISC*deltaD*l_DC
deltaU = 0.14 1/msReaction: ul_DC => Sink, Rate Law: MISC*deltaU*ul_DC
deltaH = 0.1 1/msReaction: func_CD4_HTC => Sink, Rate Law: MISC*deltaH*func_CD4_HTC
P = 3.0 1; gamma1 = 0.333; gamma = 333.0 1/ms; m = 0.5 1Reaction: Pool => Tumorcells, Rate Law: MISC*Tumorcells/((1/gamma1)^P+(Tumorcells^(1-m)/gamma)^P)^(1/P)
deltaA = 0.2 1/msReaction: sl_CD8_ETC => Sink, Rate Law: MISC*deltaA*sl_CD8_ETC
S3 = 1.7 pg/l; alpha7 = 0.022 1/msReaction: func_CD4_HTC => func_TRegs; TGFb, func_CD4_HTC, Rate Law: MISC*alpha7*func_CD4_HTC*TGFb/(S3+TGFb)
Tx = 0.999999666666889; k3 = 11.0 1; S1 = 3.5 pg/l; k2 = 1.2 1; r0 = 0.9 1/msReaction: Tumorcells => Sink; func_CD8_ETC, func_TRegs, TGFb, Rate Law: MISC*r0*Tx/(1+k2*Tx/func_CD8_ETC)*1/((1+k3*func_TRegs/func_CD8_ETC)*(1+TGFb/S1))
tc = 0.08 ksReaction: IL2 => Sink, Rate Law: MISC*IL2/tc

States:

NameDescription
Tumorcells[EFO:0000616]
l DC[dendritic cell]
func TRegs[regulatory T-lymphocyte]
TGFb[Transforming growth factor beta-1]
sl CD4 HTC[helper T-lymphocyte]
IL2[Interleukin-2]
sl CD8 ETC[cytotoxic T-lymphocyte]
sl TRegs[regulatory T-lymphocyte]
ul DC[dendritic cell]
IL10[Interleukin-10]
func CD8 ETC[cytotoxic T-lymphocyte]
func CD4 HTC[helper T-lymphocyte]
Pool[empty set]
Sink[empty set]

Roblitz2013 - Menstrual Cycle following GnRH analogue administration: BIOMD0000000494v0.0.1

Roblitz2013 - Menstrual Cycle following GnRH analogue administrationThe model describes the menstrual cycle feedback mec…

Details

The paper presents a differential equation model for the feedback mechanisms between gonadotropin-releasing hormone (GnRH), follicle-stimulating hormone (FSH), luteinizing hormone (LH), development of follicles and corpus luteum, and the production of estradiol (E2), progesterone (P4), inhibin A (IhA), and inhibin B (IhB) during the female menstrual cycle. Compared to earlier human cycle models, there are three important differences: The model presented here (a) does not involve any delay equations, (b) is based on a deterministic modeling of the GnRH pulse pattern, and (c) contains less differential equations and less parameters. These differences allow for a faster simulation and parameter identification. The focus is on modeling GnRH-receptor binding, in particular, by inclusion of a pharmacokinetic/pharmacodynamic (PK/PD) model for a GnRH agonist, Nafarelin, and a GnRH antagonist, Cetrorelix, into the menstrual cycle model. The final mathematical model describes the hormone profiles (LH, FSH, P4, E2) throughout the menstrual cycle of 12 healthy women. It correctly predicts hormonal changes following single and multiple dose administration of Nafarelin or Cetrorelix at different stages in the cycle. link: http://identifiers.org/pubmed/23206386

Parameters:

NameDescription
p244 = 138.303203866118Reaction: FSH_R => R_FSH_des; FSH_R, Rate Law: default*p244*FSH_R*default/default
facFSH = 1.0; p240 = 3.52890638983354Reaction: FSH_bld + R_FSH => FSH_R; FSH_bld, R_FSH, Rate Law: default*p240/facFSH*FSH_bld*default*R_FSH*default/default
p234 = 183.363164488992Reaction: LH_R => R_LH_des; LH_R, Rate Law: default*p234*LH_R*default/default
p154 = 5.23500984428137Reaction: E2 => sa75_degraded; E2, Rate Law: default*p154*E2*default/default
p302 = 322.176481116879Reaction: GnRH + R_GnRH_a => GnRH_R_a; GnRH, R_GnRH_a, Rate Law: default*p302*GnRH*default*R_GnRH_a*default/default
p80 = 20.0; p83 = 8.0E-4; p84 = 5.0; p41 = 0.610291748702345Reaction: Lut3 => Lut4; GnRH_R_a, Ago_R_a, Ago_R_a, GnRH_R_a, Lut3, Rate Law: default*p41*(1+p80*((GnRH_R_a*default+Ago_R_a*default)/p83)^p84/(1+((GnRH_R_a*default+Ago_R_a*default)/p83)^p84))*Lut3*default/default
p313 = 644.35Reaction: Ago_R_a => Ago_c + R_GnRH_a; Ago_R_a, Rate Law: default*p313*Ago_R_a*default/default
p319 = 32.22Reaction: Ago_R_a => Ago_R_i; Ago_R_a, Rate Law: default*p319*Ago_R_a*default/default
p275 = 2.65Reaction: Ago_c => s102; Ago_c, Rate Law: default*p275*Ago_c*default/default
p38 = 0.958117057454806Reaction: Sc2 => Lut1; Sc2, Rate Law: default*p38*Sc2*default/default
p513 = 644.35Reaction: Ant_R => R_GnRH_a + Ant_c; Ant_R, Rate Law: default*p513*Ant_R/default
p307 = 32.2176481116879Reaction: R_GnRH_i => R_GnRH_a; R_GnRH_i, Rate Law: default*p307*R_GnRH_i*default/default
p36 = 12.2060139609808Reaction: OvF => sa53_degraded; OvF, Rate Law: default*p36*OvF*default/default
p312 = 322.18Reaction: R_GnRH_a + Ago_c => Ago_R_a; Ago_c, R_GnRH_a, Rate Law: default*p312*R_GnRH_a*default*Ago_c*default/default
p49 = 3.66180418829425; p48 = 0.608121; p47 = 3.0Reaction: s62 => AF1; FSH_R, FSH_R, Rate Law: default*p49*(FSH_R*default/p48)^p47/(1+(FSH_R*default/p48)^p47)/default
p241 = 114.247359942724Reaction: FSH_bld => sa31_degraded; FSH_bld, Rate Law: default*p241*FSH_bld*default/default
p3 = 192.2041; p6 = 10.0; p7 = 1.0; p1 = 7309.91587614104; p2 = 7309.91587614104; facE2 = 1.0; facP4 = 1.0; p4 = 2.3708Reaction: s33 => LH_Pit; E2, P4, E2, P4, Rate Law: default*(p1+p2*(E2*default/(p3*facE2))^p6/(1+(E2*default/(p3*facE2))^p6))/(1+(P4*default/(p4*facP4))^p7)/default
p303 = 644.352962233757Reaction: GnRH_R_a => GnRH + R_GnRH_a; GnRH_R_a, Rate Law: default*p303*GnRH_R_a*default/default
p300 = 0.447467334884553Reaction: GnRH => sa3_degraded; GnRH, Rate Law: default*p300*GnRH*default/default
p306 = 3.22176481116878Reaction: R_GnRH_a => R_GnRH_i; R_GnRH_a, Rate Law: default*p306*R_GnRH_a*default/default
p305 = 32.2176481116879Reaction: GnRH_R_i => R_GnRH_i; GnRH_R_i, Rate Law: default*p305*GnRH_R_i*default/default
p175 = 134240.200465366; p174 = 447.467334884553; p173 = 89.4934669769107Reaction: s82 => InhB; AF2, Sc2, AF2, Sc2, Rate Law: default*(p173+p174*AF2*default+p175*Sc2*default)/default
p51 = 4.88231609092536; p52 = 2.7262; p46 = 3.68896Reaction: AF2 => AF3; LH_R, R_Foll, AF2, LH_R, R_Foll, Rate Law: default*p51*(LH_R*default/p52)^p46*R_Foll*default*AF2*default/default
p37 = 1.22060139609808Reaction: Sc1 => Sc2; Sc1, Rate Law: default*p37*Sc1*default/default
p161 = 0.972; p152 = 2.09450510112762; p165 = 8675.13871487382; p164 = 1713.71039914086; p160 = 3480.27; facE2 = 1.0; p158 = 51.558081260068; p159 = 9.28Reaction: s74 => E2; AF3, AF4, Lut1, Lut4, AF2, PrF, LH_bld, AF2, AF3, AF4, LH_bld, Lut1, Lut4, PrF, Rate Law: default*facE2*(p158+p152*AF2*default+p159*AF3*default*LH_bld*default+p160*AF4*default+p161*PrF*default*LH_bld*default+p164*Lut1*default+p165*Lut4*default)/default
p230 = 2.1430105602291; facLH = 1.0Reaction: LH_bld + R_LH => LH_R; LH_bld, R_LH, Rate Law: default*p230/facLH*LH_bld*default*R_LH*default/default
freq = 3.17876449742659; mass = 0.00120799195301476Reaction: s87 => GnRH; E2, P4, Rate Law: default*freq*mass/default
p52 = 2.7262; p44 = 2.0; p55 = 10.0; p33 = 12.2060139609808Reaction: s67 => AF4; LH_R, AF4, AF4, LH_R, Rate Law: default*p33*(LH_R*default/p52)^p44*AF4*default*(1-AF4*default/p55)/default
p475 = 5.0Reaction: Ant_c => s115; Ant_c, Rate Law: default*p475*Ant_c/default
p320 = 3.22Reaction: Ago_R_i => Ago_R_a; Ago_R_i, Rate Law: default*p320*Ago_R_i*default/default
p57 = 10.0; p56 = 0.02; p26 = 1.20834079112225Reaction: s71 => Sc1; OvF, OvF, Rate Law: default*p26*(OvF*default/p56)^p57/(1+(OvF*default/p56)^p57)/default
p474 = 45.56; p473 = 34.9Reaction: s114 => Ant_c; Ant_d, Ant_d, Rate Law: default*p474/p473*Ant_d/default
p55 = 10.0; p31 = 0.122060139609808Reaction: s66 => AF3; FSH_R, AF3, AF3, FSH_R, Rate Law: default*p31*FSH_R*default*AF3*default*(1-AF3*default/p55)/default
p474 = 45.56Reaction: Ant_d => s113; Ant_d, Rate Law: default*p474*Ant_d/default
p95 = 1.34267048505459; p93 = 5.0; p92 = 1.2348; facP4 = 1.0Reaction: R_Foll => sa61_degraded; P4, P4, R_Foll, Rate Law: default*p95*(P4*default/(p92*facP4))^p93/(1+(P4*default/(p92*facP4))^p93)*R_Foll*default/default
p311 = 8.94934669769107E-5Reaction: s85 => R_GnRH_i, Rate Law: default*p311/default
p314 = 0.009Reaction: Ago_R_i => s106; Ago_R_i, Rate Law: default*p314*Ago_R_i*default/default
p242 = 61.0291748702345Reaction: R_FSH_des => R_FSH; R_FSH_des, Rate Law: default*p242*R_FSH_des*default/default
p155 = 5.12958654018257Reaction: P4 => sa78_degraded; P4, Rate Law: default*p155*P4*default/default
p18 = 3.0E-4; p12 = 5.0; p28 = 0.27201539287632; p20 = 2.0; facFSH = 1.0; p17 = 0.0569894397708967Reaction: s95 => FSH_bld; GnRH_R_a, FSH_pit, Ago_R_a, Ago_R_a, FSH_pit, GnRH_R_a, Rate Law: default*facFSH/p12*(p17+p28*((GnRH_R_a*default+Ago_R_a*default)/p18)^p20/(1+((GnRH_R_a*default+Ago_R_a*default)/p18)^p20))*FSH_pit*default/default
p50 = 1.22060139609808Reaction: AF1 => AF2; FSH_R, AF1, FSH_R, Rate Law: default*p50*FSH_R*default*AF1*default/default
p309 = 32.2176481116879Reaction: GnRH_R_a => GnRH_R_i; GnRH_R_a, Rate Law: default*p309*GnRH_R_a*default/default
p310 = 3.222Reaction: GnRH_R_i => GnRH_R_a; GnRH_R_i, Rate Law: default*p310*GnRH_R_i*default/default
p157 = 172.453910864507Reaction: InhB => sa86_degraded; InhB, Rate Law: default*p157*InhB*default/default
p52 = 2.7262; p35 = 122.060139609808; p45 = 6.0Reaction: PrF => sa52_degraded; R_Foll, LH_R, LH_R, PrF, R_Foll, Rate Law: default*p35*(LH_R*default/p52)^p45*R_Foll*default*PrF*default/default
p308 = 0.0894934669769107Reaction: R_GnRH_i => sa1_degraded; R_GnRH_i, Rate Law: default*p308*R_GnRH_i*default/default
p18 = 3.0E-4; p28 = 0.27201539287632; p20 = 2.0; p17 = 0.0569894397708967Reaction: FSH_pit => s94; GnRH_R_a, Ago_R_a, Ago_R_a, FSH_pit, GnRH_R_a, Rate Law: default*(p17+p28*((GnRH_R_a*default+Ago_R_a*default)/p18)^p20/(1+((GnRH_R_a*default+Ago_R_a*default)/p18)^p20))*FSH_pit*default/default
p274 = 54.2Reaction: Ago_d => s107; Ago_d, Rate Law: default*p274*Ago_d*default/default
p24 = 5.0; p22 = 95.81; p21 = 22129.0495793807; freq = 3.17876449742659; p11 = 10.0; p13 = 3.0; p23 = 70.0; p25 = 2.0Reaction: s38 => FSH_pit; InhA_delay, InhB, InhA_delay, InhB, Rate Law: default*p21/(1+(InhA_delay*default/p22)^p24+(InhB*default/p23)^p25)*1/(1+(freq/p11)^p13)/default
p304 = 0.00894934669769107Reaction: GnRH_R_i => csa1_degraded; GnRH_R_i, Rate Law: default*p304*GnRH_R_i*default/default
p8 = 3.0E-4; p9 = 5.0; p16 = 0.00476024700196886; p5 = 0.190415249686773Reaction: LH_Pit => s92; GnRH_R_a, Ago_R_a, Ago_R_a, GnRH_R_a, LH_Pit, Rate Law: default*(p16+p5*((GnRH_R_a*default+Ago_R_a*default)/p8)^p9/(1+((GnRH_R_a*default+Ago_R_a*default)/p8)^p9))*LH_Pit*default/default
p166 = 0.9426346876678; p167 = 761.643100053696; facP4 = 1.0Reaction: s76 => P4; Lut4, Lut4, Rate Law: default*facP4*(p166+p167*Lut4*default)/default
p232 = 68.9493466976911Reaction: R_LH_des => R_LH; R_LH_des, Rate Law: default*p232*R_LH_des*default/default
p43 = 5.0; p52 = 2.7262; p32 = 122.060139609808Reaction: AF3 => AF4; R_Foll, LH_R, AF3, LH_R, R_Foll, Rate Law: default*p32*(LH_R*default/p52)^p43*R_Foll*default*AF3*default/default
p231 = 74.8505459101486Reaction: LH_bld => sa28_degraded; LH_bld, Rate Law: default*p231*LH_bld*default/default
p8 = 3.0E-4; p9 = 5.0; p12 = 5.0; p16 = 0.00476024700196886; p5 = 0.190415249686773; facLH = 1.0Reaction: s93 => LH_bld; GnRH_R_a, LH_Pit, Ago_R_a, Ago_R_a, GnRH_R_a, LH_Pit, Rate Law: default*facLH/p12*(p16+p5*((GnRH_R_a*default+Ago_R_a*default)/p8)^p9/(1+((GnRH_R_a*default+Ago_R_a*default)/p8)^p9))*LH_Pit*default/default
p514 = 0.009Reaction: Ant_R => s116; Ant_R, Rate Law: default*p514*Ant_R/default
p177 = 60.0; p171 = 216.85; p168 = 1.44522999821013; p169 = 2.28494719885448; p172 = 114.247359942724; p170 = 28.2110255951316; p178 = 180.0Reaction: s78 => InhA; PrF, Sc1, Lut1, Lut2, Lut3, Lut4, Lut1, Lut2, Lut3, Lut4, PrF, Sc1, Rate Law: default*(p168+p169*PrF*default+p177*Sc1*default+p178*Lut1*default+p170*Lut2*default+p171*Lut3*default+p172*Lut4*default)/default
p91 = 5.0; p94 = 0.2186056917845; p90 = 3.0Reaction: s64 => R_Foll; FSH_bld, FSH_bld, Rate Law: default*p94*(FSH_bld*default/p90)^p91/(1+(FSH_bld*default/p90)^p91)/default
p315 = 32.22Reaction: Ago_R_i => R_GnRH_i; Ago_R_i, Rate Law: default*p315*Ago_R_i*default/default
p274 = 54.2; p273 = 38.12Reaction: s108 => Ago_c; Ago_d, Ago_d, Rate Law: default*p274/p273*Ago_d*default/default
p53 = 3.0; p52 = 2.7262; p54 = 10.0; p45 = 6.0; p27 = 7.98433864327904Reaction: s69 => OvF; R_Foll, LH_R, PrF, LH_R, PrF, R_Foll, Rate Law: default*p27*R_Foll*default*(LH_R*default/p52)^p45*(PrF*default/p53)^p54/(1+(PrF*default/p53)^p54)/default
p52 = 2.7262; p34 = 332.754608913549Reaction: AF4 => PrF; LH_R, R_Foll, AF4, LH_R, R_Foll, Rate Law: default*p34*LH_R*default/p52*R_Foll*default*AF4*default/default
p512 = 322.18Reaction: R_GnRH_a + Ant_c => Ant_R; Ant_c, R_GnRH_a, Rate Law: default*p512*R_GnRH_a*default*Ant_c/default

States:

NameDescription
Lut3[corpus luteum]
GnRH[Progonadoliberin-1]
R FollR_Foll
Ago d[gonadotropin releasing hormone agonist]
Sc2Sc2
sa75 degradedsa75_degraded
GnRH R a[Gonadotropin-releasing hormone receptor]
Ago R a[gonadotropin releasing hormone agonist; Gonadotropin-releasing hormone receptor]
s38s38
P4[progesterone; blood]
FSH R[Follicle-stimulating hormone receptor; Glycoprotein hormones alpha chain]
OvF[follicular fluid formation in ovarian follicle antrum involved in fused antrum stage]
s69s69
Ago R i[gonadotropin releasing hormone agonist; Gonadotropin-releasing hormone receptor]
GnRH R i[Gonadotropin-releasing hormone receptor]
Sc1Sc1
sa86 degradedsa86_degraded
s106s106
R FSH[Follicle-stimulating hormone receptor]
sa61 degradedsa61_degraded
s33s33
s94s94
sa31 degradedsa31_degraded
s82s82
sa78 degradedsa78_degraded
AF2[mature ovarian follicle]
Ago c[gonadotropin releasing hormone agonist]
s85s85
s78s78
s92s92
R GnRH i[Gonadotropin-releasing hormone receptor]
csa1 degradedcsa1_degraded
s93s93
s115s115
s71s71
AF3[mature ovarian follicle]
AF4[mature ovarian follicle]
R LH des[Lutropin-choriogonadotropic hormone receptor; receptor internalization]
FSH pit[hypophysis; Glycoprotein hormones alpha chain]
s87s87
Ant c[GnRH antagonist]
Ant d[GnRH antagonist]
s116s116
s67s67
E2[17beta-estradiol; blood]
PrF[follicular fluid formation in ovarian follicle antrum involved in fused antrum stage]
s64s64
s62s62
s108s108
s114s114
AF1[mature ovarian follicle]
R GnRH a[Gonadotropin-releasing hormone receptor]
R LH[Lutropin-choriogonadotropic hormone receptor]
sa28 degradedsa28_degraded
FSH bld[Glycoprotein hormones alpha chain; blood]
s66s66

Roda2020 - SIR model of COVID-19 spread in Wuhan: BIOMD0000000957v0.0.1

=Since the COVID-19 outbreak in Wuhan City in December of 2019, numerous model predictions on the COVID-19 epidemics in…

Details

Since the COVID-19 outbreak in Wuhan City in December of 2019, numerous model predictions on the COVID-19 epidemics in Wuhan and other parts of China have been reported. These model predictions have shown a wide range of variations. In our study, we demonstrate that nonidentifiability in model calibrations using the confirmed-case data is the main reason for such wide variations. Using the Akaike Information Criterion (AIC) for model selection, we show that an SIR model performs much better than an SEIR model in representing the information contained in the confirmed-case data. This indicates that predictions using more complex models may not be more reliable compared to using a simpler model. We present our model predictions for the COVID-19 epidemic in Wuhan after the lockdown and quarantine of the city on January 23, 2020. We also report our results of modeling the impacts of the strict quarantine measures undertaken in the city after February 7 on the time course of the epidemic, and modeling the potential of a second outbreak after the return-to-work in the city. link: http://identifiers.org/pubmed/32289100

Rodenfels2019 - Heat Oscillations Driven by the Embryonic Cell Cycle Reveal the Energetic Costs of Signaling: BIOMD0000000952v0.0.1

All living systems function out of equilibrium and exchange energy in the form of heat with their environment. Thus, hea…

Details

All living systems function out of equilibrium and exchange energy in the form of heat with their environment. Thus, heat flow can inform on the energetic costs of cellular processes, which are largely unknown. Here, we have repurposed an isothermal calorimeter to measure heat flow between developing zebrafish embryos and the surrounding medium. Heat flow increased over time with cell number. Unexpectedly, a prominent oscillatory component of the heat flow, with periods matching the synchronous early reductive cleavage divisions, persisted even when DNA synthesis and mitosis were blocked by inhibitors. Instead, the heat flow oscillations were driven by the phosphorylation and dephosphorylation reactions catalyzed by the cell-cycle oscillator, the biochemical network controlling mitotic entry and exit. We propose that the high energetic cost of cell-cycle signaling reflects the significant thermodynamic burden of imposing accurate and robust timing on cell proliferation during development. link: http://identifiers.org/pubmed/30713074

Rodrigues2014-Vaccination models and optimal control strategies to dengue: MODEL2003190001v0.0.1

As the development of a dengue vaccine is ongoing, we simulate an hypothetical vaccine as an extra protection to the pop…

Details

As the development of a dengue vaccine is ongoing, we simulate an hypothetical vaccine as an extra protection to the population. In a first phase, the vaccination process is studied as a new compartment in the model, and different ways of distributing the vaccines investigated: pediatric and random mass vaccines, with distinct levels of efficacy and durability. In a second step, the vaccination is seen as a control variable in the epidemiological process. In both cases, epidemic and endemic scenarios are included in order to analyze distinct outbreak realities. link: http://identifiers.org/pubmed/24513243

Rodrigues2019 - A mathematical model for chemoimmunotherapy of chronic lymphocytic leukemia: BIOMD0000000879v0.0.1

THis is a simple ordinary differential equation model describing chemoimmunotherapy of chronic lymphocytic leukemia, inc…

Details

Immunotherapy is currently regarded as the most promising treatment to fight against cancer. This is particularly true in the treatment of chronic lymphocytic leukemia, an indolent neoplastic disease of B-lymphocytes which eventually causes the immune system's failure. In this and other areas of cancer research, mathematical modeling is pointed out as a prominent tool to analyze theoretical and practical issues. Its lack in studies of chemoimmunotherapy of chronic lymphocytic leukemia is what motivated us to come up with a simple ordinary differential equation model. It is based on ideas of de Pillis and Radunskaya and on standard pharmacokinetics-pharmacodynamics assumptions. In order to check the positivity of the state variables, we first establish an invariant region where these time-dependent variables remain positive. Afterwards, the action of the immune system, as well as the chemoimmunotherapeutic role in promoting cancer cure are investigated by means of numerical simulations and the classical linear stability analysis. The role of adoptive cellular immunotherapy is also addressed. Our overall conclusion is that chemoimmunotherapeutic protocols can be effective in treating chronic lymphocytic leukemia provided that chemotherapy is not a limiting factor to the immunotherapy efficacy. link: http://identifiers.org/doi/10.1016/j.amc.2018.12.008

Parameters:

NameDescription
d = 0.001Reaction: I =>, Rate Law: compartment*d*I
rho = 1.0E-12; gamma = 100.0Reaction: => I; N, Rate Law: compartment*rho*N*I/(gamma+N)
s_0 = 700000.0Reaction: => I, Rate Law: compartment*s_0
c_1 = 5.0E-11Reaction: N => ; I, Rate Law: compartment*c_1*N*I
lambda = 4.16Reaction: Q =>, Rate Law: compartment*lambda*Q
mu = 8.0; a = 2000.0Reaction: N => ; Q, Rate Law: compartment*mu*N*Q/(a+Q)
Immunotherapy_Input = 0.0Reaction: => I, Rate Law: compartment*Immunotherapy_Input
Chemotherapy_Input = 8640.0Reaction: => Q, Rate Law: compartment*Chemotherapy_Input
c_2 = 1.0E-13Reaction: I => ; N, Rate Law: compartment*c_2*N*I
delta = 10000.0; b = 5000000.0Reaction: I => ; Q, Rate Law: compartment*delta*I*Q/(b+Q)
k = 1.0E12; r = 0.01Reaction: => N, Rate Law: compartment*r*N*(1-N/k)

States:

NameDescription
Q[CHEBI:4027]
I[Lymphocyte]
N[C25553]

Rodriguez-Caso2006_Polyamine_Metabolism: BIOMD0000000190v0.0.1

SBML creators: Armando Reyes-Palomares * , Carlos Rodríguez-Caso +, Raul Montañez * , Marta Cascante $, Francisca Sánche…

Details

Polyamines are considered as essential compounds in living cells, since they are involved in cell proliferation, transcription, and translation processes. Furthermore, polyamine homeostasis is necessary to cell survival, and its deregulation is involved in relevant processes, such as cancer and neurodegenerative disorders. Great efforts have been made to elucidate the nature of polyamine homeostasis, giving rise to relevant information concerning the behavior of the different components of polyamine metabolism, and a great amount of information has been generated. However, a complex regulation at transcriptional, translational, and metabolic levels as well as the strong relationship between polyamines and essential cell processes make it difficult to discriminate the role of polyamine regulation itself from the whole cell response when an experimental approach is given in vivo. To overcome this limitation, a bottom-up approach to model mathematically metabolic pathways could allow us to elucidate the systemic behavior from individual kinetic and molecular properties. In this paper, we propose a mathematical model of polyamine metabolism from kinetic constants and both metabolite and enzyme levels extracted from bibliographic sources. This model captures the tendencies observed in transgenic mice for the so-called key enzymes of polyamine metabolism, ornithine decarboxylase, S-adenosylmethionine decarboxylase and spermine spermidine N-acetyl transferase. Furthermore, the model shows a relevant role of S-adenosylmethionine and acetyl-CoA availability in polyamine homeostasis, which are not usually considered in systemic experimental studies. link: http://identifiers.org/pubmed/16709566

Parameters:

NameDescription
Kmsamdc=50.0 microM; Kapsamdc=0.5 microM; Kiasamdc=2.5 microM; Kissamdc=500.0 microM; Vmaxsamdc = 0.367465856805639 uMperminReaction: SAM => A; S, P, Rate Law: cytosol*Vmaxsamdc/(1+S/Kissamdc)*SAM/(Kmsamdc*(1+Kapsamdc/P+A/Kiasamdc)+SAM)
Kcoa = 0.012 perminReaction: AcCoA => CoA, Rate Law: cytosol*Kcoa*AcCoA
Kpefflux=0.01 perminReaction: P =>, Rate Law: cytosol*Kpefflux*P
Vmpao=10.35 uMpermin; Kmdpao=50.0 microM; Kmspao=15.0 microM; Kmaspao=0.6 microM; Kmadpao=14.0 microMReaction: aD => P; aS, D, S, Rate Law: cytosol*Vmpao*aD/(Kmadpao*(1+aD/Kmadpao+aS/Kmaspao+D/Kmdpao+S/Kmspao))
Kidspds=100.0 microM; Vmspds=10.95 uMpermin; KaSpds=0.3 microM; Kpspds=40.0 microM; Kiaspds=0.8 microMReaction: A + P => D, Rate Law: cytosol*Vmspds*A*P/(Kiaspds*Kpspds*(1+D/Kidspds)+Kpspds*A+KaSpds*(1+D/Kidspds)*P+A*P)
Kaccoa = 0.004 perminReaction: CoA => AcCoA, Rate Law: cytosol*Kaccoa*CoA
Vmmat=0.45 uMpermin; Kmmat=41.0 microM; Kimetmat=50.0 microMReaction: Met => SAM, Rate Law: cytosol*Vmmat/(1+Kmmat/Met*(1+SAM/Kimetmat))
Kiaspms=0.06 microM; Kaspms=0.1 microM; Kisspms=25.0 microM; Vmspms=3.23 uMpermin; Kdspms=60.0 microMReaction: A + D => S, Rate Law: cytosol*Vmspms*A*D/(Kiaspms*Kdspms*(1+S/Kisspms)+Kdspms*A+Kaspms*(1+S/Kisspms)*D+A*D)
Kipodc=1300.0 microM; Kmodc=60.0 microM; Vmaxodc = 1.27905671844446 uMperminReaction: ORN => P, Rate Law: cytosol*Vmaxodc*ORN/(Kmodc*(1+P/Kipodc)+ORN)
Vmaxssat = 0.677298510125025 uMpermin; Kmdssat=130.0 microM; Kmaccoassat=1.5 microM; Kmsssat=35.0 microM; Kmcoassat=40.0 microMReaction: D + AcCoA => aD + CoA; S, Rate Law: cytosol*Vmaxssat*D*AcCoA/(Kmdssat*(1+S/Kmsssat)*Kmaccoassat*(1+CoA/Kmcoassat)+Kmaccoassat*(1+CoA/Kmcoassat)*D+Kmdssat*(1+S/Kmsssat)*AcCoA+D*AcCoA)
Kadefflux=0.01 perminReaction: aD =>, Rate Law: cytosol*Kadefflux*aD
C = 4.44 dimensionless; Vmaxssat = 0.677298510125025 uMpermin; Kmdssat=130.0 microM; Kmaccoassat=1.5 microM; Kmsssat=35.0 microM; Kmcoassat=40.0 microMReaction: S + AcCoA => aS + CoA; D, Rate Law: cytosol*1/C*Vmaxssat*S*AcCoA/(Kmsssat*(1+D/Kmdssat)*Kmaccoassat*(1+CoA/Kmcoassat)+Kmaccoassat*(1+CoA/Kmcoassat)*S+Kmsssat*(1+D/Kmdssat)*AcCoA+S*AcCoA)

States:

NameDescription
CoA[coenzyme A; CoA]
ORN[L-ornithine; L-Ornithine]
A[S-adenosylmethioninamine; S-Adenosylmethioninamine]
aS[N(1)-acetylspermine; N1-Acetylspermine]
P[putrescine; Putrescine]
SAM[S-adenosyl-L-methionine; S-Adenosyl-L-methionine]
D[spermidine; Spermidine]
S[spermine; Spermine]
AcCoA[acetyl-CoA; Acetyl-CoA]
aD[acetylspermidine; N1-Acetylspermidine]
Met[L-methionine; L-Methionine]

Rodriguez2005_denovo_pyrimidine_biosynthesis: MODEL0995500644v0.0.1

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

Details

With the emergence of multifaceted bioinformatics-derived data, it is becoming possible to merge biochemical and physiological information to develop a new level of understanding of the metabolic complexity of the cell. The biosynthetic pathway of de novo pyrimidine nucleotide metabolism is an essential capability of all free-living cells, and it occupies a pivotal position relative to metabolic processes that are involved in the macromolecular synthesis of DNA, RNA and proteins, as well as energy production and cell division. This regulatory network in all enteric bacteria involves genetic, allosteric, and physiological control systems that need to be integrated into a coordinated set of metabolic checks and balances. Allosterically regulated pathways constitute an exciting and challenging biosynthetic system to be approached from a mathematical perspective. However, to date, a mathematical model quantifying the contribution of allostery in controlling the dynamics of metabolic pathways has not been proposed. In this study, a direct, rigorous mathematical model of the de novo biosynthesis of pyrimidine nucleotides is presented. We corroborate the simulations with experimental data available in the literature and validate it with derepression experiments done in our laboratory. The model is able to faithfully represent the dynamic changes in the intracellular nucleotide pools that occur during metabolic transitions of the de novo pyrimidine biosynthetic pathway and represents a step forward in understanding the role of allosteric regulation in metabolic control. link: http://identifiers.org/pubmed/15784266

Rodríguez-Jorge2019 - Boolean model of combined TCR and TLR5 signaling for CD4 + T cell activation: MODEL1903260003v0.0.1

CD4+ T cells recognize antigens through their T cell receptors (TCRs); however, additional signals involving costimulato…

Details

CD4<sup>+</sup> T cells recognize antigens through their T cell receptors (TCRs); however, additional signals involving costimulatory receptors, for example, CD28, are required for proper T cell activation. Alternative costimulatory receptors have been proposed, including members of the Toll-like receptor (TLR) family, such as TLR5 and TLR2. To understand the molecular mechanism underlying a potential costimulatory role for TLR5, we generated detailed molecular maps and logical models for the TCR and TLR5 signaling pathways and a merged model for cross-interactions between the two pathways. Furthermore, we validated the resulting model by analyzing how T cells responded to the activation of these pathways alone or in combination, in terms of the activation of the transcriptional regulators CREB, AP-1 (c-Jun), and NF-κB (p65). Our merged model accurately predicted the experimental results, showing that the activation of TLR5 can play a similar role to that of CD28 activation with respect to AP-1, CREB, and NF-κB activation, thereby providing insights regarding the cross-regulation of these pathways in CD4+ T cells. link: http://identifiers.org/pubmed/30992399

Rodríguez-Jorge2019 - Boolean model of TCR signaling for CD4 + T cell activation: MODEL1903260001v0.0.1

CD4+ T cells recognize antigens through their T cell receptors (TCRs); however, additional signals involving costimulato…

Details

CD4<sup>+</sup> T cells recognize antigens through their T cell receptors (TCRs); however, additional signals involving costimulatory receptors, for example, CD28, are required for proper T cell activation. Alternative costimulatory receptors have been proposed, including members of the Toll-like receptor (TLR) family, such as TLR5 and TLR2. To understand the molecular mechanism underlying a potential costimulatory role for TLR5, we generated detailed molecular maps and logical models for the TCR and TLR5 signaling pathways and a merged model for cross-interactions between the two pathways. Furthermore, we validated the resulting model by analyzing how T cells responded to the activation of these pathways alone or in combination, in terms of the activation of the transcriptional regulators CREB, AP-1 (c-Jun), and NF-κB (p65). Our merged model accurately predicted the experimental results, showing that the activation of TLR5 can play a similar role to that of CD28 activation with respect to AP-1, CREB, and NF-κB activation, thereby providing insights regarding the cross-regulation of these pathways in CD4+ T cells. link: http://identifiers.org/pubmed/30992399

Rohwer2000_Phosphotransferase_System: BIOMD0000000038v0.0.1

The kinetic parameters in vitro of the components of the phosphoenolpyruvate:glycose phosphotransferase system (PTS) in…

Details

SBML level 2 code generated for the JWS Online project by Jacky Snoep using PySCeS

Run this model online at http://jjj.biochem.sun.ac.za

To cite JWS Online please refer to: Olivier, B.G. and Snoep, J.L. (2004) Web-based modelling using JWS Online, Bioinformatics, 20:2143-2144

Parameters:

NameDescription
k10r=0.0054; k10f=4800.0Reaction: EIICBPGlc => EIICB + GlcP, Rate Law: compartment*(k10f*EIICBPGlc-k10r*EIICB*GlcP)
k1r=480000.0; k1f=1960.0Reaction: PEP + EI => PyrPI, Rate Law: compartment*(k1f*PEP*EI-k1r*PyrPI)
k3f=14000.0; k3r=14000.0Reaction: HPr + EIP => EIPHPr, Rate Law: compartment*(k3f*EIP*HPr-k3r*EIPHPr)
k5r=21960.0; k5f=21960.0Reaction: HPrP + EIIA => HPrPIIA, Rate Law: compartment*(k5f*HPrP*EIIA-k5r*HPrPIIA)
k9f=260.0; k9r=389.0Reaction: EIICBP + Glc => EIICBPGlc, Rate Law: compartment*(k9f*EIICBP*Glc-k9r*EIICBPGlc)
k6f=4392.0; k6r=3384.0Reaction: HPrPIIA => EIIAP + HPr, Rate Law: compartment*(k6f*HPrPIIA-k6r*HPr*EIIAP)
k8f=2640.0; k8r=960.0Reaction: EIIAPIICB => EIICBP + EIIA, Rate Law: compartment*(k8f*EIIAPIICB-k8r*EIIA*EIICBP)
k7f=880.0; k7r=880.0Reaction: EIICB + EIIAP => EIIAPIICB, Rate Law: compartment*(k7f*EIIAP*EIICB-k7r*EIIAPIICB)
k4f=84000.0; k4r=3360.0Reaction: EIPHPr => HPrP + EI, Rate Law: compartment*(k4f*EIPHPr-k4r*EI*HPrP)
k2r=294.0; k2f=108000.0Reaction: PyrPI => EIP + Pyr, Rate Law: compartment*(k2f*PyrPI-k2r*Pyr*EIP)

States:

NameDescription
EIIAPIICB[PTS system glucose-specific EIICB component; PTS system glucose-specific EIIA component]
PyrPI[Phosphoenolpyruvate-protein phosphotransferase; Phosphoenolpyruvate; phosphoenolpyruvate; phosphoenolpyruvate; Phosphoenolpyruvate-protein phosphotransferase; Protein histidine]
EIICB[PTS system glucose-specific EIICB component]
EI[Phosphoenolpyruvate-protein phosphotransferase]
EIIA[PTS system glucose-specific EIIA component]
GlcP[D-glucose 6-phosphate; D-Glucose 6-phosphate]
EIP[Phosphoenolpyruvate-protein phosphotransferase]
HPr[Phosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPr]
HPrP[Phosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPr; Protein histidine]
Pyr[pyruvate; Pyruvate]
HPrPIIA[Phosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPr; PTS system glucose-specific EIIA component]
Glc[glucose; C00293]
EIIAP[PTS system glucose-specific EIIA component]
PEP[phosphoenolpyruvate; Phosphoenolpyruvate]
EIPHPr[Phosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPrPhosphocarrier protein HPr; Phosphoenolpyruvate-protein phosphotransferase]
EIICBP[PTS system glucose-specific EIICB component]
EIICBPGlc[glucose; PTS system glucose-specific EIICB component; C00293]

Rohwer2001_Sucrose: BIOMD0000000023v0.0.1

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

Details

Sucrose accumulation in developing sugar cane (Saccharum officinarum) is accompanied by a continuous synthesis and cleavage of sucrose in the storage tissues. Despite numerous studies, the factors affecting sucrose accumulation are still poorly understood, and no consistent pattern has emerged which pinpoints certain enzyme activities as important controlling steps. Here, we develop an approach based on pathway analysis and kinetic modelling to assess the biochemical control of sucrose accumulation and futile cycling in sugar cane. By using the concept of elementary flux modes, all possible routes of futile cycling of sucrose were enumerated in the metabolic system. The available kinetic data for the pathway enzymes were then collected and assembled in a kinetic model of sucrose accumulation in sugar cane culm tissue. Although no data were fitted, the model agreed well with independent experimental results: in no case was the difference between calculated and measured fluxes and concentrations greater than 2-fold. The model thus validated was then used to assess different enhancement strategies for increasing sucrose accumulation. First, the control coefficient of each enzyme in the system on futile cycling of sucrose was calculated. Secondly, the activities of those enzymes with the numerically largest control coefficients were varied over a 5-fold range to determine the effect on the degree of futile cycling, the conversion efficiency from hexoses into sucrose, and the net sucrose accumulation rate. In view of the modelling results, overexpression of the fructose or glucose transporter or the vacuolar sucrose import protein, as well as reduction of cytosolic neutral invertase levels, appear to be the most promising targets for genetic manipulation. This offers a more directed improvement strategy than cumbersome gene-by-gene manipulation. The kinetic model can be viewed and interrogated on the World Wide Web at http://jjj.biochem.sun.ac.za. link: http://identifiers.org/pubmed/11513743

Parameters:

NameDescription
Km6UDP=0.3; Ki6Suc6P=0.07; Km6F6P=0.6; Ki6F6P=0.4; Keq6=10.0; Vmax6r=0.2; Km6UDPGlc=1.8; Km6Suc6P=0.1; Ki6Pi=3.0; Vmax6f=0.379; Ki6UDPGlc=1.4Reaction: HexP => UDP + Suc6P; phos, Rate Law: compartment*Vmax6f*(0.0575*HexP*0.8231*HexP-Suc6P*UDP/Keq6)/(0.0575*HexP*0.8231*HexP*(1+Suc6P/Ki6Suc6P)+Km6F6P*(1+phos/Ki6Pi)*(0.8231*HexP+Ki6UDPGlc)+Km6UDPGlc*0.0575*HexP+Vmax6f/(Vmax6r*Keq6)*(Km6UDP*Suc6P*(1+0.8231*HexP/Ki6UDPGlc)+UDP*(Km6Suc6P*(1+Km6UDPGlc*0.0575*HexP/(Ki6UDPGlc*Km6F6P*(1+phos/Ki6Pi)))+Suc6P*(1+0.0575*HexP/Ki6F6P))))
Km3ATP=0.25; Ki3G6P=0.1; Km3Glc=0.07; Km4Fru=10.0; Vmax3=0.197; Ki4F6P=10.0Reaction: ATP + Glc => HexP + ADP; Fru, Rate Law: compartment*Vmax3*Glc/Km3Glc*ATP/Km3ATP/((1+ATP/Km3ATP)*(1+Glc/Km3Glc+Fru/Km4Fru+0.113*HexP/Ki3G6P+0.0575*HexP/Ki4F6P))
Km1Fruex=0.2; Ki1Fru=1.0; Vmax1=0.286Reaction: Fruex => Fru, Rate Law: compartment*Vmax1*Fruex/(Km1Fruex*(1+Fru/Ki1Fru)+Fruex)
Ki9Glc=15.0; Vmax9=0.372; Km9Suc=10.0; Ki9Fru=15.0Reaction: Suc => Fru + Glc, Rate Law: compartment*Vmax9/(1+Glc/Ki9Glc)*Suc/(Km9Suc*(1+Fru/Ki9Fru)+Suc)
Km11Suc=100.0; Vmax11=1.0Reaction: Suc => Sucvac, Rate Law: compartment*Vmax11*Suc/(Km11Suc+Suc)
Vmax2=0.286; Ki2Glc=1.0; Km2Glcex=0.2Reaction: Glcex => Glc, Rate Law: compartment*Vmax2*Glcex/(Km2Glcex*(1+Glc/Ki2Glc)+Glcex)
Ki5Fru=12.0; Km5Fru=0.1; Vmax5=0.164; Km5ATP=0.085; Ki5ADP=2.0Reaction: Fru + ATP => HexP + ADP, Rate Law: compartment*Vmax5/(1+Fru/Ki5Fru)*Fru/Km5Fru*ATP/Km5ATP/(1+Fru/Km5Fru+ATP/Km5ATP+Fru*ATP/(Km5Fru*Km5ATP)+ADP/Ki5ADP)
Vmax4=0.197; Ki3G6P=0.1; Km3Glc=0.07; Km4Fru=10.0; Km4ATP=0.25; Ki4F6P=10.0Reaction: Fru + ATP => HexP + ADP; Glc, Rate Law: compartment*Vmax4*Fru/Km4Fru*ATP/Km4ATP/((1+ATP/Km4ATP)*(1+Glc/Km3Glc+Fru/Km4Fru+0.113*HexP/Ki3G6P+0.0575*HexP/Ki4F6P))
Ki8Suc=40.0; Vmax8f=0.677; Km8Suc=50.0; Keq8=5.0; Ki8Fru=4.0; Ki8UDP=0.3; Km8UDP=0.3; Km8UDPGlc=0.3; Vmax8r=0.3; Km8Fru=4.0Reaction: HexP + Fru => Suc + UDP, Rate Law: compartment*(-Vmax8f)*(Suc*UDP-Fru*0.8231*HexP/Keq8)/(Suc*UDP*(1+Fru/Ki8Fru)+Km8Suc*(UDP+Ki8UDP)+Km8UDP*Suc+Vmax8f/(Vmax8r*Keq8)*(Km8UDPGlc*Fru*(1+UDP/Ki8UDP)+0.8231*HexP*(Km8Fru*(1+Km8UDP*Suc/(Ki8UDP*Km8Suc))+Fru*(1+Suc/Ki8Suc))))
Vmax7=0.5; Km7Suc6P=0.1Reaction: Suc6P => Suc + phos, Rate Law: compartment*Vmax7*Suc6P/(Km7Suc6P+Suc6P)
Km10F6P=0.2; Vmax10=0.1Reaction: HexP => glycolysis, Rate Law: compartment*Vmax10*0.0575*HexP/(Km10F6P+0.0575*HexP)

States:

NameDescription
Glcex[D-glucose; D-Glucose]
ATP[ATP; ATP]
Sucvac[sucrose; Sucrose]
glycolysis[beta-D-Fructose 1,6-bisphosphate; beta-D-fructofuranose 1,6-bisphosphate]
HexP[beta-D-glucose 1-phosphate; keto-D-fructose 6-phosphate; D-glucose 6-phosphate; UDP-glucose; alpha-D-Glucose 6-phosphate; D-Fructose 6-phosphate; D-Glucose 1-phosphate; UDP-D-glucose]
Suc6P[sucrose 6(F)-phosphate; Sucrose 6'-phosphate]
UDP[UDP; UDP]
Glc[D-glucose; D-Glucose]
Fruex[D-fructose; D-Fructose]
ADP[ADP; ADP]
Fru[D-fructose; D-Fructose]
Suc[sucrose; Sucrose]
phos[phosphate(3-); Orthophosphate]

Romano-Nguyen2014 - MST2 and Raf1 Crosstalk: MODEL1506070001v0.0.1

Originally created by libAntimony v1.3 (using libSBML 4.1.0-b2)

Details

Signal transduction requires the coordination of activities between different pathways. In mammalian cells, Raf-1 regulates the MST-LATS and MEK-ERK pathways. We found that a complex circuitry of competing protein interactions coordinates the crosstalk between the ERK and MST pathways. Combining mathematical modelling and experimental validation we show that competing protein interactions can cause steep signalling switches through phosphorylation-induced changes in binding affinities. These include Akt phosphorylation of MST2 and a feedback phosphorylation of Raf-1 Ser 259 by LATS1, which enables Raf-1 to suppress both MST2 and MEK signalling. Mutation of Raf-1 Ser 259 stimulates both pathways, simultaneously driving apoptosis and proliferation, whereas concomitant MST2 downregulation switches signalling to cell proliferation, transformation and survival. Thus, competing protein interactions provide a versatile regulatory mechanism for signal distribution through the dynamic integration of graded signals into switch-like responses. link: http://identifiers.org/pubmed/24929361

Romond1999_CellCycle: BIOMD0000000207v0.0.1

The model reproduces Fig 3 of the paper. Model successfully reproduced using MathSBML and Jarnac. To the extent possibl…

Details

The animal cell cycle is controlled by the periodic variation of two cyclin-dependent protein kinases, cdk1 and cdk2, which govern the entry into the M (mitosis) and S (DNA replication) phases, respectively. The ordered progression between these phases is achieved thanks to the existence of checkpoint mechanisms based on mutual inhibition of these processes. Here we study a simple theoretical model for oscillations in cdk1 and cdk2 activity, involving mutual inhibition of the two oscillators. Each minimal oscillator is described by a three-variable cascade involving a cdk, together with the associated cyclin and cyclin-degrading enzyme. The dynamics of this skeleton model of coupled oscillators is determined as a function of the strength of their mutual inhibition. The most common mode of dynamic behavior, obtained under conditions of strong mutual inhibition, is that of alternating oscillations in cdk1 and cdk2, which correspond to the physiological situation of the ordered recurrence of the M and S phases. In addition, for weaker inhibition we obtain evidence for a variety of dynamic phenomena such as complex periodic oscillations, chaos, and the coexistence between multiple periodic or chaotic attractors. We discuss the conditions of occurrence of these various modes of oscillatory behavior, as well as their possible physiological significance. link: http://identifiers.org/pubmed/10415827

Parameters:

NameDescription
vi1 = 0.05 uM_min_1; Kim1 = 0.03 dimensionlessReaction: => C1; M2, Rate Law: compartment*vi1*Kim1/(Kim1+M2)
vd1 = 0.025 uM_min_1; K_d1 = 0.02 uMReaction: C1 => ; X1, Rate Law: compartment*vd1*X1*C1/(K_d1+C1)
K1 = 0.01 dimensionless; V1 = 0.0 min_1; V2 = 0.15 min_1; K2 = 0.01 dimensionlessReaction: M1 = V1*(1-M1)/(K1+(1-M1))-V2*M1/(K2+M1), Rate Law: V1*(1-M1)/(K1+(1-M1))-V2*M1/(K2+M1)
H4 = 0.01 dimensionless; U3 = 0.0 min_1; H3 = 0.01 dimensionless; U4 = 0.05 min_1Reaction: X2 = U3*(1-X2)/(H3+(1-X2))-U4*X2/(H4+X2), Rate Law: U3*(1-X2)/(H3+(1-X2))-U4*X2/(H4+X2)
kd2 = 0.001 min_1Reaction: C2 =>, Rate Law: compartment*kd2*C2
V4 = 0.05 min_1; K4 = 0.01 dimensionless; V3 = 0.0 min_1; K3 = 0.01 dimensionlessReaction: X1 = V3*(1-X1)/(K3+(1-X1))-V4*X1/(K4+X1), Rate Law: V3*(1-X1)/(K3+(1-X1))-V4*X1/(K4+X1)
vd2 = 0.025 uM_min_1; K_d2 = 0.02 uMReaction: C2 => ; X2, Rate Law: compartment*vd2*X2*C2/(K_d2+C2)
vi2 = 0.05 uM_min_1; Kim2 = 0.03 dimensionlessReaction: => C2; M1, Rate Law: compartment*vi2*Kim2/(Kim2+M1)
kd1 = 0.001 min_1Reaction: C1 =>, Rate Law: compartment*kd1*C1
U1 = 0.0 min_1; U2 = 0.15 min_1; H2 = 0.01 dimensionless; H1 = 0.01 dimensionlessReaction: M2 = U1*(1-M2)/(H1+(1-M2))-U2*M2/(H2+M2), Rate Law: U1*(1-M2)/(H1+(1-M2))-U2*M2/(H2+M2)

States:

NameDescription
C1[G2/mitotic-specific cyclin-B3]
C2[G1/S-specific cyclin-E1]
M2[Cyclin-dependent kinase 2]
M1[Cyclin-dependent kinase 1]
X2[SUMO-activating enzyme subunit 2]
X1[SUMO-activating enzyme subunit 2]

Rosas2015 - Caffeine-induced luminal SR calcium changes: BIOMD0000000601v0.0.1

This SBML model reproduced the calcium release from SR by application of 20 mM or 2mM caffeine, described in the paper.…

Details

The process of Ca2+ release from sarcoplasmic reticulum (SR) comprises 4 phases in smooth muscle cells. Phase 1 is characterized by a large increase of the intracellular Ca2+ concentration ([Ca2+]i) with a minimal reduction of the free luminal SR Ca2+. Importantly, active SR Ca2+ ATPases (SERCA pumps) are necessary for phase 1 to occur. This situation cannot be explained by the standard kinetics that involves a fixed amount of luminal Ca2+ binding sites. A new mathematical model was developed that assumes an increasing SR Ca2+ buffering capacity in response to an increase of the luminal SR [Ca2+] that is called Kinetics-on-Demand (KonD) model. This approach can explain both phase 1 and the refractory period associated with a recovered [Ca2+]FSR. Additionally, our data suggest that active SERCA pumps are a requisite for KonD to be functional; otherwise luminal SR Ca2+ binding proteins switch to standard kinetics. The importance of KonD Ca2+ binding properties is twofold: a more efficient Ca2+ release process and that [Ca2+]FSR and Ca2+-bound to SR proteins ([Ca2+]BSR) can be regulated separately allowing for Ca2+ release to occur (provided by Ca2+-bound to luminal Ca2+ binding proteins) without an initial reduction of the [Ca2+]FSR. link: http://identifiers.org/pubmed/26390403

Parameters:

NameDescription
parameter_23 = 0.0103445237903673; parameter_6 = 1.7; parameter_2 = 65.0; parameter_5 = 0.052Reaction: mwd805cc43_4a96_472f_a894_c119a6aa895f + mw447078ee_8bc8_4358_abcd_ade10dba93b0 + mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6 => mw40a96ef6_32da_46d1_9712_4f53f60bad43 + mw447078ee_8bc8_4358_abcd_ade10dba93b0 + mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6, Rate Law: parameter_2*parameter_5^parameter_6*parameter_23*(mw447078ee_8bc8_4358_abcd_ade10dba93b0-mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6)
parameter_3 = 1.125E-5; parameter_8 = 2.0; parameter_7 = 3.0E-7Reaction: mw40a96ef6_32da_46d1_9712_4f53f60bad43 + mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6 => mwd805cc43_4a96_472f_a894_c119a6aa895f + mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6, Rate Law: parameter_3*mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6^parameter_8/(parameter_7^parameter_8+mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6^parameter_8)
parameter_9 = 100.0Reaction: mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6 = mw40a96ef6_32da_46d1_9712_4f53f60bad43/parameter_9, Rate Law: missing
parameter_14 = 1.515E-4; parameter_26 = 0.0018165Reaction: mw447078ee_8bc8_4358_abcd_ade10dba93b0 = (parameter_26-(parameter_26^2-4*mwd805cc43_4a96_472f_a894_c119a6aa895f*parameter_14)^(1/2))/2, Rate Law: missing
parameter_1 = 35.0; parameter_9 = 100.0; parameter_4 = 7.5E-6Reaction: mw40a96ef6_32da_46d1_9712_4f53f60bad43 + mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6 => mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6, Rate Law: mw44539b83_caa2_4da5_bae0_a8dcf7439431*parameter_1*(mwe1a0a651_d2d5_4f75_8d45_9336c60eb9a6-parameter_4/parameter_9)/mw44539b83_caa2_4da5_bae0_a8dcf7439431

States:

NameDescription
mwd805cc43 4a96 472f a894 c119a6aa895f[calcium(2+)]
mw447078ee 8bc8 4358 abcd ade10dba93b0[calcium(2+)]
mwe1a0a651 d2d5 4f75 8d45 9336c60eb9a6[calcium(2+)]
mw40a96ef6 32da 46d1 9712 4f53f60bad43[calcium(2+)]

Rouhimoghadam2018 - GPR30/PI3K/MAPK/STAT signaling pathway in normal and cancer cells: MODEL2002250001v0.0.1

In the current study, we aimed to simulate the GPR30/PI3K/MAPK/STAT signaling pathway in normal and cancer cells by the…

Details

Tamoxifen (Nolvadex) is one of the most widely used and effective therapeutic agent for breast cancer. It benefits nearly 75% of patients with estrogen receptor (ER)-positive breast cancer that receive this drug. Its effectiveness is mainly attributed to its capacity to function as an ER antagonist, blocking estrogen binding sites on the receptor, and inhibiting the proliferative action of the receptor-hormone complex. Although, tamoxifen can induce apoptosis in breast cancer cells via upregulation of pro-apoptotic factors, it can also promote uterine hyperplasia in some women. Thus, tamoxifen as a multi-functional drug could have different effects on cells based on the utilization of effective concentrations or availability of specific co-factors. Evidence that tamoxifen functions as a GPR30 (G-Protein Coupled Receptor 30) agonist activating adenylyl cyclase and EGFR (Epidermal Growth Factor Receptor) intracellular signaling networks, provides yet another means of explaining the multi-functionality of tamoxifen. Here ordinary differential equation (ODE) modeling, RNA sequencing and real time qPCR analysis were utilized to establish the necessary data for gene network mapping of tamoxifen-stimulated MCF-7 cells, which express the endogenous ER and GPR30. The gene set enrichment analysis and pathway analysis approaches were used to categorize transcriptionally upregulated genes in biological processes. Of the 2,713 genes that were significantly upregulated following a 48 h incubation with 250 μM tamoxifen, most were categorized as either growth-related or pro-apoptotic intermediates that fit into the Tp53 and/or MAPK signaling pathways. Collectively, our results display that the effects of tamoxifen on the breast cancer MCF-7 cell line are mediated by the activation of important signaling pathways including Tp53 and MAPKs to induce apoptosis. link: http://identifiers.org/pubmed/30050469

Rouhimoghadam2018 - GPR30/PI3K/MAPK/STAT signaling pathway in normal and cancer cells: MODEL1807040001v0.0.1

In the current study, we aimed to simulate the GPR30/PI3K/MAPK/STAT signaling pathway in normal and cancer cells by the…

Details

Tamoxifen (Nolvadex) is one of the most widely used and effective therapeutic agent for breast cancer. It benefits nearly 75% of patients with ER-positive breast cancer that receive this drug. Its effectiveness is mainly attributed to its capacity to function as an estrogen receptor (ER) antagonist, blocking estrogen binding sites on the receptor, and inhibiting the proliferative action of the receptor-hormone complex. Although, tamoxifen can induce apoptosis in breast cancer cells via upregulation of pro-apoptotic factors, it can also promote uterine hyperplasia in some women. Thus, tamoxifen as a multi-functional drug could have different effects on cells based on the utilization of effective concentrations or availability of specific co-factors. Evidence that tamoxifen functions as a GPR30 (G-Protein Coupled Receptor 30) agonist activating adenylyl cyclase and EGFR (Epidermal Growth Factor Receptor) intracellular signaling networks, provides yet another means of explaining the multi-functionality of tamoxifen. Here ordinary differential equation (ODE) modeling, RNA sequencing and real time qPCR analysis were utilized to establish the necessary data for gene network mapping of tamoxifen-stimulated MCF-7 cells, which express the endogenous ER and GPR30. The gene set enrichment analysis and pathway analysis approaches were used to categorize transcriptionally upregulated genes in biological processes. Of the 2,713 genes that were significantly upregulated following a 48 h incubation with 250 μM tamoxifen, most were categorized as either growth-related or pro-apoptotic intermediates that fit into the Tp53 and/or MAPK signaling pathways. Collectively, our results display that the effects of tamoxifen on the breast cancer MCF-7 cell line are mediated by the activation of important signaling pathways including Tp53 and MAPKs to induce apoptosis. link: http://identifiers.org/doi/10.3389/fphys.2018.00907

Rovers1995_Photsynthetic_Oscillations: BIOMD0000000292v0.0.1

This is the model described in the article: **Photosynthetic oscillations and the interdependence of photophosphorylat…

Details

A simple mathematical model of photosynthetic carbon metabolism as driven by ATP and NADPH has been formulated to analyse photosynthetic oscillations. Two essential assumptions of this model are: (i) reduction of 3-phosphoglycerate to triosephosphate in the Clavin cycle is limited by ATP, not by NADPH, and (ii) photophosphorylation is affected by the availability of both ADP and NADP, while electron transport is limited by NADP only. The model produces oscillations of observed damping and period in ATP and NADP concentrations which are about 180 degrees out of phase, while three alternative proposals regarding coupling of electron transport and photophosphorylation do not produce oscillatory model solutions. The phases of ATP and NADPH are in reasonable agreement with the available experimental data. The model (which assumes that redox control of photophosphorylation is part of the oscillatory mechanism) is compared with an alternative proposal (that oscillations are due to interdependence of turnover of adenylates and Calvin cycle intermediates). From the similarity of the mathematical structures of both models it is inviting to speculate that both models are partial aspects of 'the oscillatory mechanism'. link: http://identifiers.org/pubmed/7772723

Parameters:

NameDescription
k3 = 0.3 per_mM_per_sReaction: ADP => ATP; NADP, Rate Law: c*k3*ADP*NADP
k2 = 0.625 per_sReaction: Y + NADPH + ATP => X + ADP + NADP, Rate Law: c*k2*ATP
k1 = 0.123 per_sReaction: X + ATP => Y + ADP, Rate Law: c*k1*X
N0 = 1.2 mMReaction: NADP = N0-NADPH, Rate Law: missing
k4 = 0.614 per_sReaction: NADP => NADPH, Rate Law: c*k4*NADP
A0 = 2.5 mMReaction: ADP = A0-ATP, Rate Law: missing

States:

NameDescription
Y[3-phospho-D-glyceric acid; D-ribulose 1,5-bisphosphate; 3-Phospho-D-glycerate; D-Ribulose 1,5-bisphosphate]
NADPH[NADPH; NADPH]
ATP[ATP; ATP]
NADP[NADP(+); NADP+]
X[dihydroxyacetone phosphate; aldehydo-D-ribose 5-phosphate; D-ribulose 5-phosphate; keto-D-fructose 1,6-bisphosphate; keto-D-fructose 6-phosphate; D-erythrose 4-phosphate; sedoheptulose 1,7-bisphosphate; sedoheptulose 7-phosphate; D-glyceraldehyde 3-phosphate; Glycerone phosphate; D-Ribose 5-phosphate; D-Ribulose 5-phosphate; Sedoheptulose 7-phosphate; Sedoheptulose 1,7-bisphosphate; D-Erythrose 4-phosphate; D-Fructose 6-phosphate; D-Fructose 1,6-bisphosphate; D-Glyceraldehyde 3-phosphate]
ADP[ADP; ADP]

Roy2019 - A vivid cytokines interaction model on psoriasis with the effect of impulse biologic therapy: MODEL1911070001v0.0.1

This is a mathematical model describing keratinocyte proliferation dynamics as influenced by immune cells and cytokines…

Details

Psoriasis is a chronic skin condition that produces plaques of condensed, scaling skin due to excessively rapid proliferation of keratinocytes. During the disease progression, keratinocyte proliferation is influenced by many immune cells and cytokines. This article deals with a five dimensional deterministic model, which has been derived using quasi-steady-state approximation for describing the dynamics of psoriasis in various cytokines environment. Equilibrium analysis of the system shows that either the system converges to a stable steady state or exhibits a periodic oscillation depending upon system parameters. Finally, introducing a one dimensional impulsive system, we have determined the perfect dose and perfect dosing interval for biologic (TNF-α inhibitor) therapy to control the hyper-proliferation of keratinocytes. We have studied the effect of TNF-α inhibitor by considering both perfect and imperfect dosing during the inductive phase. The maximum possible number of drug holidays and the minimal number of doses that must subsequently be taken while avoiding drug resistance have been calculated for imperfect dosing. Since, psoriasis is non-curable but treatable disease, so the aim is to investigate the minimum dose with highest efficacy and proper dosing interval of TNF-α inhibitor for a psoriatic patient. Through numerical simulations, we have given a detailed prediction about the maximum drug holidays, tolerable for a patient, without loss of previous drug effects. Our theoretical predictions and numerical outcomes may be useful in guiding the design of future clinical trials. link: http://identifiers.org/pubmed/30980871

Rozendaal2018 - Model Integrating Glucose and Lipid Dynamics: MODEL1803200001v0.0.1

https://journals.plos.org/ploscompbiol/article?id=10.1371/journal.pcbi.1006145

Details

The Metabolic Syndrome (MetS) is a complex, multifactorial disorder that develops slowly over time presenting itself with large differences among MetS patients. We applied a systems biology approach to describe and predict the onset and progressive development of MetS, in a study that combined in vivo and in silico models. A new data-driven, physiological model (MINGLeD: Model INtegrating Glucose and Lipid Dynamics) was developed, describing glucose, lipid and cholesterol metabolism. Since classic kinetic models cannot describe slowly progressing disorders, a simulation method (ADAPT) was used to describe longitudinal dynamics and to predict metabolic concentrations and fluxes. This approach yielded a novel model that can describe long-term MetS development and progression. This model was integrated with longitudinal in vivo data that was obtained from male APOE*3-Leiden.CETP mice fed a high-fat, high-cholesterol diet for three months and that developed MetS as reflected by classical symptoms including obesity and glucose intolerance. Two distinct subgroups were identified: those who developed dyslipidemia, and those who did not. The combination of MINGLeD with ADAPT could correctly predict both phenotypes, without making any prior assumptions about changes in kinetic rates or metabolic regulation. Modeling and flux trajectory analysis revealed that differences in liver fluxes and dietary cholesterol absorption could explain this occurrence of the two different phenotypes. In individual mice with dyslipidemia dietary cholesterol absorption and hepatic turnover of metabolites, including lipid fluxes, were higher compared to those without dyslipidemia. Predicted differences were also observed in gene expression data, and consistent with the emergence of insulin resistance and hepatic steatosis, two well-known MetS co-morbidities. Whereas MINGLeD specifically models the metabolic derangements underlying MetS, the simulation method ADAPT is generic and can be applied to other diseases where dynamic modeling and longitudinal data are available. link: http://identifiers.org/pubmed/29879115

Rozi2003_GlycogenPhosphorylase_Activation: BIOMD0000000100v0.0.1

The model reproduces the temporal evolution of Glycogen phosphorylase for a vale of Vm5=30 as depicted in Fig 1a of the…

Details

Taking into account the Ca(2+)-stimulated degradation of inositol 1,4,5-trisphosphate (IP(3)) by a 3-kinase, we have theoretically explored the effects of both simple and complex Ca(2+) oscillations on the regulation of a phosphorylation-dephosphorylation cycle process involved in glycogen degradation by glycogen phosphorylase a-form, respectively. For the case of simple Ca(2+) oscillations, the roles of cytosolic Ca(2+) oscillations in the regulation of active phosphorylase depend upon the maximum rate of IP(3) degradation by the 3-kinase, V(M5). In particular, the smaller the values of V(M5) are, the lower the effective Ca(2+) threshold for the activation of glycogen phosphorylase will be. For the case of complex Ca(2+) oscillations, the average level of fraction of active phosphorylase is nearly independent from the level of stimulation increasing in the bursting oscillatory domain. Both simple and complex Ca(2+) oscillations can contribute to increase the efficiency and specificity of cellular signalling, and some theoretical results of activation of glycogen phosphorylase regulated by Ca(2+) oscillations are close to the experimental results for gene expression in lymphocytes. link: http://identifiers.org/pubmed/14556891

Parameters:

NameDescription
Ky = 0.2 uM; m = 2.0 dimensionless; Kz = 0.5 uM; Ka = 0.2 uM; Vm3 = 20.0 uM_per_minReaction: Y => Z; A, Rate Law: intravesicular*Vm3*A^4*Y^2*Z^m/((Ka^4+A^4)*(Ky^2+Y^2)*(Kz^m+Z^m))
alpha = 9.0 dimensionless; Ka1 = 10000.0 uM; Kp2 = 0.2; Ka2 = 10000.0 uM; G = 10000.0 uM; Vpm2 = 0.6 min_invReaction: GP =>, Rate Law: cytosol*Vpm2*(1+alpha*G/(Ka1+G))*GP/(Kp2/(1+G/Ka2)+GP)
epsilon = 0.1 min_invReaction: A =>, Rate Law: cytosol*epsilon*A
Vm2 = 6.0 uM_per_min; K2 = 0.1 uMReaction: Z => Y, Rate Law: cytosol*Vm2*Z^2/(K2^2+Z^2)
K5 = 1.0 uM; p = 2.0 dimensionless; n = 4.0 dimensionless; Kd = 0.4 uM; Vm5 = 30.0 uM_per_minReaction: A => ; Z, Rate Law: cytosol*Vm5*A^p*Z^n/((K5^p+A^p)*(Kd^n+Z^n))
v0 = 2.0 uM_per_min; v1 = 2.0 uM_per_min; beta = 0.5 dimensionlessReaction: EC => Z, Rate Law: extracellular*(v0+v1*beta)
Kf = 1.0 min_invReaction: Y => Z, Rate Law: intravesicular*Kf*Y
K = 10.0 min_invReaction: Z => EC, Rate Law: cytosol*K*Z
beta = 0.5 dimensionless; V4 = 2.0 uM_per_minReaction: => A, Rate Law: cytosol*beta*V4
gamma = 9.0 dimensionless; Ka6 = 0.5 uM; K1 = 0.1; Vpm1 = 1.5 min_inv; Ka5 = 0.5 uMReaction: => GP; Z, Rate Law: cytosol*Vpm1*(1+gamma*Z^4/(Ka5^4+Z^4))*(1-GP)/((K1/(1+Z^4/Ka6^4)+1)-GP)

States:

NameDescription
Y[calcium(2+); Calcium cation]
Z[calcium(2+); Calcium cation]
A[1D-myo-inositol 1,4,5-trisphosphate; D-myo-Inositol 1,4,5-trisphosphate]
EC[calcium(2+); Calcium cation]
GP[Glycogen phosphorylase, brain form]

Rutkis2013 - Entner-Doudoroff pathway in Z.mobilis: MODEL1409050000v0.0.1

The model corresponds to the third columns (model simulations) in tables 3 and 5 of publication (PMID: 24085837, DOI: 10…

Details

Zymomonas mobilis, an ethanol-producing bacterium, possesses the Entner-Doudoroff (E-D) pathway, pyruvate decarboxylase and two alcohol dehydrogenase isoenzymes for the fermentative production of ethanol and carbon dioxide from glucose. Using available kinetic parameters, we have developed a kinetic model that incorporates the enzymic reactions of the E-D pathway, both alcohol dehydrogenases, transport reactions and reactions related to ATP metabolism. After optimizing the reaction parameters within likely physiological limits, the resulting kinetic model was capable of simulating glycolysis in vivo and in cell-free extracts with good agreement with the fluxes and steady-state intermediate concentrations reported in previous experimental studies. In addition, the model is shown to be consistent with experimental results for the coupled response of ATP concentration and glycolytic flux to ATPase inhibition. Metabolic control analysis of the model revealed that the majority of flux control resides not inside, but outside the E-D pathway itself, predominantly in ATP consumption, demonstrating why past attempts to increase the glycolytic flux through overexpression of glycolytic enzymes have been unsuccessful. Co-response analysis indicates how homeostasis of ATP concentrations starts to deteriorate markedly at the highest glycolytic rates. This kinetic model has potential for application in Z. mobilis metabolic engineering and, since there are currently no E-D pathway models available in public databases, it can serve as a basis for the development of models for other micro-organisms possessing this type of glycolytic pathway. link: http://identifiers.org/pubmed/24085837

Rutkis2013 - Entner-Doudoroff pathway in Z.mobilis (cell-free): MODEL1409050001v0.0.1

The model corresponds to the Table 4 and the last column of table 5 of publication (PMID: 24085837, DOI: 10.1099/mic.0.0…

Details

Zymomonas mobilis, an ethanol-producing bacterium, possesses the Entner-Doudoroff (E-D) pathway, pyruvate decarboxylase and two alcohol dehydrogenase isoenzymes for the fermentative production of ethanol and carbon dioxide from glucose. Using available kinetic parameters, we have developed a kinetic model that incorporates the enzymic reactions of the E-D pathway, both alcohol dehydrogenases, transport reactions and reactions related to ATP metabolism. After optimizing the reaction parameters within likely physiological limits, the resulting kinetic model was capable of simulating glycolysis in vivo and in cell-free extracts with good agreement with the fluxes and steady-state intermediate concentrations reported in previous experimental studies. In addition, the model is shown to be consistent with experimental results for the coupled response of ATP concentration and glycolytic flux to ATPase inhibition. Metabolic control analysis of the model revealed that the majority of flux control resides not inside, but outside the E-D pathway itself, predominantly in ATP consumption, demonstrating why past attempts to increase the glycolytic flux through overexpression of glycolytic enzymes have been unsuccessful. Co-response analysis indicates how homeostasis of ATP concentrations starts to deteriorate markedly at the highest glycolytic rates. This kinetic model has potential for application in Z. mobilis metabolic engineering and, since there are currently no E-D pathway models available in public databases, it can serve as a basis for the development of models for other micro-organisms possessing this type of glycolytic pathway. link: http://identifiers.org/pubmed/24085837

S


Saa2016 - Mammalian methionine cycle - approximate bayesian computation: MODEL1603150000v0.0.1

Construction of feasible and accurate kinetic models of metabolism: A Bayesian approach Pedro A. Saa, Lars K. Nielsen*…

Details

Kinetic models are essential to quantitatively understand and predict the behaviour of metabolic networks. Detailed and thermodynamically feasible kinetic models of metabolism are inherently difficult to formulate and fit. They have a large number of heterogeneous parameters, are non-linear and have complex interactions. Many powerful fitting strategies are ruled out by the intractability of the likelihood function. Here, we have developed a computational framework capable of fitting feasible and accurate kinetic models using Approximate Bayesian Computation. This framework readily supports advanced modelling features such as model selection and model-based experimental design. We illustrate this approach on the tightly-regulated mammalian methionine cycle. Sampling from the posterior distribution, the proposed framework generated thermodynamically feasible parameter samples that converged on the true values, and displayed remarkable prediction accuracy in several validation tests. Furthermore, a posteriori analysis of the parameter distributions enabled appraisal of the systems properties of the network (e.g., control structure) and key metabolic regulations. Finally, the framework was used to predict missing allosteric interactions. link: http://identifiers.org/pubmed/27417285

Saad2017 - immune checkpoint and BCG in superficial bladder cancer: BIOMD0000000746v0.0.1

The paper describes a model on the Dynamics of Immune Checkpoints, Immune System, and BCG in the Treatment of Superficia…

Details

This paper aims to study the dynamics of immune suppressors/checkpoints, immune system, and BCG in the treatment of superficial bladder cancer. Programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA4), and transforming growth factor-beta (TGF-β) are some of the examples of immune suppressors/checkpoints. They are responsible for deactivating the immune system and enhancing immunological tolerance. Moreover, they categorically downregulate and suppress the immune system by preventing and blocking the activation of T-cells, which in turn decreases autoimmunity and enhances self-tolerance. In cancer immunotherapy, the immune checkpoints/suppressors prevent and block the immune cells from attacking, spreading, and killing the cancer cells, which leads to cancer growth and development. We formulate a mathematical model that studies three possible dynamics of the treatment and establish the effects of the immune checkpoints on the immune system and the treatment at large. Although the effect cannot be seen explicitly in the analysis of the model, we show it by numerical simulations. link: http://identifiers.org/pubmed/29312460

Parameters:

NameDescription
delta = 151932.0 1/dReaction: => P, Rate Law: bladder_cancer_tme*delta
u2 = 0.1 1/dReaction: B =>, Rate Law: bladder_cancer_tme*u2*B
alpha2 = 3.45E-10 1/dReaction: E => ; C, Rate Law: bladder_cancer_tme*alpha2*E*C
k = 2000.0 1; a2 = 0.052 1/dReaction: => E; B, P, Rate Law: bladder_cancer_tme*a2*B*E/(P+k)
r = 0.0033 1/dReaction: => C, Rate Law: bladder_cancer_tme*r*C
alpha1 = 1.1E-7 1/d; k = 2000.0 1Reaction: C => ; E, P, Rate Law: bladder_cancer_tme*alpha1*E*C/(P+k)
u3 = 166.32 1/dReaction: P =>, Rate Law: bladder_cancer_tme*u3*P
alpha3 = 1.25E-7 1/dReaction: B => ; E, Rate Law: bladder_cancer_tme*alpha3*E*B
b = 650000.0 1/dReaction: => B, Rate Law: bladder_cancer_tme*b
k = 2000.0 1; a1 = 0.25 1/dReaction: => E; C, P, Rate Law: bladder_cancer_tme*a1*C*E/(P+k)
u1 = 0.041 1/dReaction: E =>, Rate Law: bladder_cancer_tme*u1*E

States:

NameDescription
B[Mycobacterium bovis BCG]
C[0000362; malignant cell]
P[Transforming growth factor beta-1; Cytotoxic T-lymphocyte protein 4; Programmed cell death protein 1]
E[Effector Immune Cell]

Sachse2008_FibroblastInteractingMyocytes: MODEL7914759868v0.0.1

This a model from the article: Electrophysiological modeling of fibroblasts and their interaction with myocytes. Sac…

Details

Experimental studies have shown that cardiac fibroblasts are electrically inexcitable, but can contribute to electrophysiology of myocardium in various manners. The aim of this computational study was to give insights in the electrophysiological role of fibroblasts and their interaction with myocytes. We developed a mathematical model of fibroblasts based on data from whole-cell patch clamp and polymerase chain reaction (PCR) studies. The fibroblast model was applied together with models of ventricular myocytes to assess effects of heterogeneous intercellular electrical coupling. We investigated the modulation of action potentials of a single myocyte varying the number of coupled fibroblasts and intercellular resistance. Coupling to fibroblasts had only a minor impact on the myocyte's resting and peak transmembrane voltage, but led to significant changes of action potential duration and upstroke velocity. We examined the impact of fibroblasts on conduction in one-dimensional strands of myocytes. Coupled fibroblasts reduced conduction and upstroke velocity. We studied electrical bridging between ventricular myocytes via fibroblast insets for various coupling resistors. The simulations showed significant conduction delays up to 20.3 ms. In summary, the simulations support strongly the hypothesis that coupling of fibroblasts to myocytes modulates electrophysiology of cardiac cells and tissues. link: http://identifiers.org/pubmed/17999190

Sackmann2006 - mating pheromone response pathway of S.cerevisiae: MODEL1403040000v0.0.1

Sackmann2006 - mating pheromone response pathway of S.cerevisiaeThis model is described in the article: [Application of…

Details

BACKGROUND: Signal transduction pathways are usually modelled using classical quantitative methods, which are based on ordinary differential equations (ODEs). However, some difficulties are inherent in this approach. On the one hand, the kinetic parameters involved are often unknown and have to be estimated. With increasing size and complexity of signal transduction pathways, the estimation of missing kinetic data is not possible. On the other hand, ODEs based models do not support any explicit insights into possible (signal-) flows within the network. Moreover, a huge amount of qualitative data is available due to high-throughput techniques. In order to get information on the systems behaviour, qualitative analysis techniques have been developed. Applications of the known qualitative analysis methods concern mainly metabolic networks. Petri net theory provides a variety of established analysis techniques, which are also applicable to signal transduction models. In this context special properties have to be considered and new dedicated techniques have to be designed. METHODS: We apply Petri net theory to model and analyse signal transduction pathways first qualitatively before continuing with quantitative analyses. This paper demonstrates how to build systematically a discrete model, which reflects provably the qualitative biological behaviour without any knowledge of kinetic parameters. The mating pheromone response pathway in Saccharomyces cerevisiae serves as case study. RESULTS: We propose an approach for model validation of signal transduction pathways based on the network structure only. For this purpose, we introduce the new notion of feasible t-invariants, which represent minimal self-contained subnets being active under a given input situation. Each of these subnets stands for a signal flow in the system. We define maximal common transition sets (MCT-sets), which can be used for t-invariant examination and net decomposition into smallest biologically meaningful functional units. CONCLUSION: The paper demonstrates how Petri net analysis techniques can promote a deeper understanding of signal transduction pathways. The new concepts of feasible t-invariants and MCT-sets have been proven to be useful for model validation and the interpretation of the biological system behaviour. Whereas MCT-sets provide a decomposition of the net into disjunctive subnets, feasible t-invariants describe subnets, which generally overlap. This work contributes to qualitative modelling and to the analysis of large biological networks by their fully automatic decomposition into biologically meaningful modules. link: http://identifiers.org/pubmed/17081284

Saeidi2012 - Quorum sensing device that produces GFP: BIOMD0000000438v0.0.1

Saeidi2012 - Quorum sensing device that produces GFPSaeidi et al. (2012) has modelled a quorum sensing device that prod…

Details

Modeling of biological parts is of crucial importance as it enables the in silico study of synthetic biological systems prior to the actual construction of genetic circuits, which can be time consuming and costly. Because standard biological parts are utilized to build the synthetic systems, it is important that each of these standard parts is well characterized and has a corresponding mathematical model that could simulate the characteristics of the part. These models could be used in computer aided design (CAD) tools during the design stage to facilitate the building of the model of biological systems. This paper describes the development of a mathematical model that is able to simulate both the dynamic and static performance of a biological device created using standard parts. We modeled an example quorum sensing device that produces green fluorescent protein (GFP) as reporter in the presence of Acyl Homoserine Lactone (AHL). The parameters of the model were estimated using experimental results. The simulation results show that the model was able to simulate behavior similar to experimental results. Since it is important that these models and the content in the models can be searchable and readable by machines, standard SBML (system biology markup language) format was used to store the models. All parts and reactions are fully annotated to enable easy searching, and the models follow the Minimum Information Requested In the Annotation of Models (MIRIAM) compliance as well as the Minimum Information About a Simulation Experiment (MIASE). link: http://identifiers.org/doi/10.1016/j.ces.2012.12.016

Parameters:

NameDescription
n2=2.0 substance; K12=2.4E-7 substance; K7=0.004051 substance; K8=0.009567 substance; n1=2.0 substance; K9=9.742E-8 substance; K11=1.0E-14 substance; k10=6.5E-16 substanceReaction: s17 => s45; s17, s45, Rate Law: (K7+K8*s17^n1/(K9^n1+s17^n1))*((k10+K11*s17^n2/(K12^n2+s17^n2))-s45)
k2=35.7 substanceReaction: s2 => s19; s2, Rate Law: k2*s2
k5=1960000.0 substance; k6=10.2 substanceReaction: s42 + s16 => s17; s16, s42, s17, Rate Law: s16*s42*k5-k6*s17
Y2=0.0696 substanceReaction: s19 => s3; s19, Rate Law: Y2*s19
k3=9600000.0 substance; k4=0.0 substanceReaction: s19 + s4 => s42; s19, s4, s42, Rate Law: s19*s4*k3-k4*s42
k1=3.734 substance; Y1=0.348 substanceReaction: s1 => s2; s1, s2, Rate Law: k1*s1-Y1*s2
Y3=2.832E-4 substanceReaction: s4 => s5; s4, Rate Law: Y3*s4

States:

NameDescription
s1Ptet-LasR
s42[transcription factor complex]
s5sa6_degraded
s17[DNA-directed RNA polymerase complex]
s2[messenger RNA]
s4[N-(3-oxododecanoyl)-D-homoserine lactone]
s19[Transcriptional activator protein LasR]
s16pLuxR
s3sa3_degraded
s45[Green fluorescent protein]

Saha2011 - Genome-scale metabolic network of Arabidopsis thaliana (iRS1597): MODEL1507180011v0.0.1

Saha2011 - Genome-scale metabolic network of Arabidopsis thaliana (iRS1597)This model is described in the article: [Zea…

Details

The scope and breadth of genome-scale metabolic reconstructions have continued to expand over the last decade. Herein, we introduce a genome-scale model for a plant with direct applications to food and bioenergy production (i.e., maize). Maize annotation is still underway, which introduces significant challenges in the association of metabolic functions to genes. The developed model is designed to meet rigorous standards on gene-protein-reaction (GPR) associations, elementally and charged balanced reactions and a biomass reaction abstracting the relative contribution of all biomass constituents. The metabolic network contains 1,563 genes and 1,825 metabolites involved in 1,985 reactions from primary and secondary maize metabolism. For approximately 42% of the reactions direct literature evidence for the participation of the reaction in maize was found. As many as 445 reactions and 369 metabolites are unique to the maize model compared to the AraGEM model for A. thaliana. 674 metabolites and 893 reactions are present in Zea mays iRS1563 that are not accounted for in maize C4GEM. All reactions are elementally and charged balanced and localized into six different compartments (i.e., cytoplasm, mitochondrion, plastid, peroxisome, vacuole and extracellular). GPR associations are also established based on the functional annotation information and homology prediction accounting for monofunctional, multifunctional and multimeric proteins, isozymes and protein complexes. We describe results from performing flux balance analysis under different physiological conditions, (i.e., photosynthesis, photorespiration and respiration) of a C4 plant and also explore model predictions against experimental observations for two naturally occurring mutants (i.e., bm1 and bm3). The developed model corresponds to the largest and more complete to-date effort at cataloguing metabolism for a plant species. link: http://identifiers.org/pubmed/21755001

Saha2011- Genome-scale metabolic network of Zea mays (iRS1563): MODEL1507180064v0.0.1

Saha2011- Genome-scale metabolic network of Zea mays (iRS1563)This model is described in the article: [Zea mays iRS1563…

Details

The scope and breadth of genome-scale metabolic reconstructions have continued to expand over the last decade. Herein, we introduce a genome-scale model for a plant with direct applications to food and bioenergy production (i.e., maize). Maize annotation is still underway, which introduces significant challenges in the association of metabolic functions to genes. The developed model is designed to meet rigorous standards on gene-protein-reaction (GPR) associations, elementally and charged balanced reactions and a biomass reaction abstracting the relative contribution of all biomass constituents. The metabolic network contains 1,563 genes and 1,825 metabolites involved in 1,985 reactions from primary and secondary maize metabolism. For approximately 42% of the reactions direct literature evidence for the participation of the reaction in maize was found. As many as 445 reactions and 369 metabolites are unique to the maize model compared to the AraGEM model for A. thaliana. 674 metabolites and 893 reactions are present in Zea mays iRS1563 that are not accounted for in maize C4GEM. All reactions are elementally and charged balanced and localized into six different compartments (i.e., cytoplasm, mitochondrion, plastid, peroxisome, vacuole and extracellular). GPR associations are also established based on the functional annotation information and homology prediction accounting for monofunctional, multifunctional and multimeric proteins, isozymes and protein complexes. We describe results from performing flux balance analysis under different physiological conditions, (i.e., photosynthesis, photorespiration and respiration) of a C4 plant and also explore model predictions against experimental observations for two naturally occurring mutants (i.e., bm1 and bm3). The developed model corresponds to the largest and more complete to-date effort at cataloguing metabolism for a plant species. link: http://identifiers.org/pubmed/21755001

Sakmann2000_SodiumCurrent_ModelA: MODEL1006230021v0.0.1

This a model from the article: Distribution of a persistent sodium current across the ventricular wall in guinea pigs.…

Details

A tetrodotoxin-sensitive persistent sodium current, I(pNa), was found in guinea pig ventricular myocytes by whole-cell patch clamping. This current was characterized in cells derived from the basal left ventricular subendocardium, midmyocardium, and subepicardium. Midmyocardial cells show a statistically significant (P<0.05) smaller I(pNa) than subendocardial and subepicardial myocytes. There was no significant difference in I(pNa) current density between subepicardial and subendocardial cells. Computer modeling studies support a role of this current in the dispersion of action potential duration across the ventricular wall. link: http://identifiers.org/pubmed/11073887