SBMLBioModels: D - F

D


dAlcantara2003_SynapticPlasticity: MODEL8938094216v0.0.1

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

Details

Postsynaptic Ca2+ signals of different amplitudes and durations are able to induce either long-lasting potentiation (LPT) or depression (LTD). The bidirectional character of synaptic plasticity may result at least in part from an increased or decreased responsiveness of the glutamatergic alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPA-R) due to the modification of conductance and/or channel number, and controlled by the balance between the activities of phosphorylation and dephosphorylation pathways. AMPA-R depression can be induced by a long-lived Ca2+ signal of moderate amplitude favouring the activation of the dephosphorylation pathway, whereas a shorter but higher Ca2+ signal would induce AMPA-R potentiation resulting from the preferential activation of the phosphorylation pathway. Within the framework of a model involving calcium/calmodulin-dependent protein kinase II (CaMKII), calcineurin (PP2B) and type 1 protein phosphatase (PP1), we aimed at delineating the conditions allowing a biphasic U-shaped relationship between AMPA-R and Ca2+ signal amplitude, and thus bidirectional plasticity. Our theoretical analysis shows that such a property may be observed if the phosphorylation pathway: (i) displays higher cooperativity in its Ca2+-dependence than the dephosphorylation pathway; (ii) displays a basal Ca2+-independent activity; or (iii) is directly inhibited by the dephosphorylation pathway. Because the experimentally observed inactivation of CaMKII by PP1 accounts for this latter characteristic, we aimed at verifying whether a realistic model using reported parameters values can simulate the induction of either LTP or LTD, depending on the time and amplitude characteristics of the Ca2+ signal. Our simulations demonstrate that the experimentally observed bidirectional nature of Ca2+-dependent synaptic plasticity could be the consequence of the PP1-mediated inactivation of CaMKII. link: http://identifiers.org/pubmed/12823459

DallaMan2007_MealModel_GlucoseInsulinSystem: BIOMD0000000379v0.0.1

This a model from the article: Meal simulation model of the glucose-insulin system. Dalla Man C, Rizza RA, Cobelli…

Details

A simulation model of the glucose-insulin system in the postprandial state can be useful in several circumstances, including testing of glucose sensors, insulin infusion algorithms and decision support systems for diabetes. Here, we present a new simulation model in normal humans that describes the physiological events that occur after a meal, by employing the quantitative knowledge that has become available in recent years. Model parameters were set to fit the mean data of a large normal subject database that underwent a triple tracer meal protocol which provided quasi-model-independent estimates of major glucose and insulin fluxes, e.g., meal rate of appearance, endogenous glucose production, utilization of glucose, insulin secretion. By decomposing the system into subsystems, we have developed parametric models of each subsystem by using a forcing function strategy. Model results are shown in describing both a single meal and normal daily life (breakfast, lunch, dinner) in normal. The same strategy is also applied on a smaller database for extending the model to type 2 diabetes. link: http://identifiers.org/pubmed/17926672

Parameters:

NameDescription
k_2 = 0.079; U_id = 0.748772844504839; k_1 = 0.065Reaction: G_t = ((-U_id)+k_1*G_p)-k_2*G_t, Rate Law: ((-U_id)+k_1*G_p)-k_2*G_t
k_empt = 0.0554800817258192; k_gri = 0.0558Reaction: Q_sto2 = (-k_empt)*Q_sto2+k_gri*Q_sto1, Rate Law: (-k_empt)*Q_sto2+k_gri*Q_sto1
m_4 = 0.194; m_1 = 0.19; m_2 = 0.484Reaction: I_p = ((-m_2)*I_p-m_4*I_p)+m_1*I_l, Rate Law: ((-m_2)*I_p-m_4*I_p)+m_1*I_l
S = 1.8; m_1 = 0.19; m_3 = 0.276120406260733; m_2 = 0.484Reaction: I_l = ((-m_1)*I_l-m_3*I_l)+m_2*I_p+S, Rate Law: ((-m_1)*I_l-m_3*I_l)+m_2*I_p+S
I = 25.0; k_i = 0.0079Reaction: I_1 = (-k_i)*(I_1-I), Rate Law: (-k_i)*(I_1-I)
beta = 0.11; G_b = 95.0; G = 94.6808510638298; alpha = 0.05Reaction: Y = (-alpha)*(Y-beta*(G-G_b)), Rate Law: (-alpha)*(Y-beta*(G-G_b))
k_gri = 0.0558Reaction: Q_sto1 = (-k_gri)*Q_sto1, Rate Law: (-k_gri)*Q_sto1
k_abs = 0.057; k_empt = 0.0554800817258192Reaction: Q_gut = (-k_abs)*Q_gut+k_empt*Q_sto2, Rate Law: (-k_abs)*Q_gut+k_empt*Q_sto2
k_2 = 0.079; E = 0.0; EGP = 1.87872; Ra = 0.0; k_1 = 0.065; U_ii = 1.0Reaction: G_p = ((((EGP+Ra)-E)-U_ii)-k_1*G_p)+k_2*G_t, Rate Law: ((((EGP+Ra)-E)-U_ii)-k_1*G_p)+k_2*G_t
k_i = 0.0079Reaction: I_d = (-k_i)*(I_d-I_1), Rate Law: (-k_i)*(I_d-I_1)
gamma = 0.5; S_po = 1.76784893617021Reaction: I_po = (-gamma)*I_po+S_po, Rate Law: (-gamma)*I_po+S_po
I = 25.0; p_2U = 0.0331; I_b = 25.0Reaction: X = (-p_2U)*X+p_2U*(I-I_b), Rate Law: (-p_2U)*X+p_2U*(I-I_b)

States:

NameDescription
X[Insulin]
I po[Insulin]
G p[glucose]
Q sto1[glucose]
I p[Insulin]
I 1[Insulin]
Q gut[glucose]
YY
Q sto2[glucose]
G t[glucose]
I l[Insulin]
I dI_d

DallePezze2012 - TSC-independent mTORC2 regulation: BIOMD0000000581v0.0.1

DallePezze2012 - TSC-independent mTORC2 regulationThis model is described in the article: [A dynamic network model of m…

Details

The kinase mammalian target of rapamycin (mTOR) exists in two multiprotein complexes (mTORC1 and mTORC2) and is a central regulator of growth and metabolism. Insulin activation of mTORC1, mediated by phosphoinositide 3-kinase (PI3K), Akt, and the inhibitory tuberous sclerosis complex 1/2 (TSC1-TSC2), initiates a negative feedback loop that ultimately inhibits PI3K. We present a data-driven dynamic insulin-mTOR network model that integrates the entire core network and used this model to investigate the less well understood mechanisms by which insulin regulates mTORC2. By analyzing the effects of perturbations targeting several levels within the network in silico and experimentally, we found that, in contrast to current hypotheses, the TSC1-TSC2 complex was not a direct or indirect (acting through the negative feedback loop) regulator of mTORC2. Although mTORC2 activation required active PI3K, this was not affected by the negative feedback loop. Therefore, we propose an mTORC2 activation pathway through a PI3K variant that is insensitive to the negative feedback loop that regulates mTORC1. This putative pathway predicts that mTORC2 would be refractory to Akt, which inhibits TSC1-TSC2, and, indeed, we found that mTORC2 was insensitive to constitutive Akt activation in several cell types. Our results suggest that a previously unknown network structure connects mTORC2 to its upstream cues and clarifies which molecular connectors contribute to mTORC2 activation. link: http://identifiers.org/pubmed/22457331

Parameters:

NameDescription
k1=0.999989Reaction: species_2 + species_6 => species_11 + species_6; species_2, species_6, Rate Law: compartment_2*k1*species_2*species_6
k1=4.50769Reaction: species_3 + species_22 => species_4 + species_22; species_3, species_22, Rate Law: compartment_2*k1*species_3*species_22
k1=0.0253763Reaction: species_20 + species_41 => species_21; species_20, species_41, Rate Law: compartment_1*k1*species_20*species_41
k1=0.073093Reaction: species_9 + species_2 => species_12 + species_2; species_9, species_2, Rate Law: compartment_2*k1*species_9*species_2
k1=1.00001E-4Reaction: species_9 + species_4 => species_10 + species_4; species_9, species_4, Rate Law: compartment_2*k1*species_9*species_4
k1=0.00812537Reaction: species_8 => species_6; species_8, Rate Law: compartment_2*k1*species_8
k1=0.0513784Reaction: species_11 + species_28 => species_2; species_11, species_28, Rate Law: compartment_2*k1*species_11*species_28
k1=2.32165E-4Reaction: species_16 => species_18; species_16, Rate Law: compartment_2*k1*species_16
k1=0.0309731Reaction: species_15 => species_20; species_15, Rate Law: compartment_1*k1*species_15
k1=7.52842Reaction: species_4 => species_3; species_4, Rate Law: compartment_2*k1*species_4
k1=0.0239178Reaction: species_9 + species_3 => species_10 + species_3; species_9, species_3, Rate Law: compartment_2*k1*species_9*species_3
k1=0.00573896Reaction: species_47 + species_2 => species_17 + species_2; species_47, species_2, Rate Law: compartment_2*k1*species_47*species_2
k1=0.00627315Reaction: species_6 + species_3 => species_8 + species_3; species_6, species_3, Rate Law: compartment_2*k1*species_6*species_3
k1=1.0Reaction: species_42 + species_17 => species_19 + species_17; species_42, species_17, Rate Law: compartment_2*k1*species_42*species_17
k1=0.403706Reaction: species_12 => species_9; species_12, Rate Law: compartment_2*k1*species_12
k1=0.0255714Reaction: species_22 => species_5; species_22, Rate Law: compartment_2*k1*species_22
k1=0.0999968Reaction: species_1 => species_42; species_1, Rate Law: compartment_2*k1*species_1
k1=1.00039E-4Reaction: species_6 + species_4 => species_8 + species_4; species_6, species_4, Rate Law: compartment_2*k1*species_6*species_4
k1=0.699505Reaction: species_27 + species_7 => species_3 + species_7; species_27, species_7, Rate Law: compartment_2*k1*species_27*species_7
k1=0.0318902Reaction: species_5 + species_16 => species_22 + species_16; species_5, species_16, Rate Law: compartment_2*k1*species_5*species_16
k1=5.90372Reaction: species_3 + species_14 => species_4 + species_14; species_3, species_14, Rate Law: compartment_2*k1*species_3*species_14
k1=0.00328283Reaction: species_7 => species_42; species_7, Rate Law: compartment_2*k1*species_7
k1=0.00528455Reaction: species_17 => species_47; species_17, Rate Law: compartment_2*k1*species_17
k1=1.0E-4Reaction: species_7 + species_17 => species_19 + species_17; species_7, species_17, Rate Law: compartment_2*k1*species_7*species_17
k1=0.149328Reaction: species_21 => species_15; species_21, Rate Law: compartment_1*k1*species_21
k1=4.0739Reaction: species_3 => species_27; species_3, Rate Law: compartment_2*k1*species_3
k1=0.999985Reaction: species_18 + species_21 => species_16 + species_21; species_18, species_21, Rate Law: k1*species_18*species_21
k1=0.1Reaction: species_13 + species_21 => species_14 + species_21; species_13, species_21, Rate Law: k1*species_13*species_21
k1=0.999991Reaction: species_10 => species_9; species_10, Rate Law: compartment_2*k1*species_10
k1=0.134664Reaction: species_42 + species_21 => species_7 + species_21; species_42, species_21, Rate Law: k1*species_42*species_21

States:

NameDescription
species 9[Proline-rich AKT1 substrate 1]
species 27[RAC-alpha serine/threonine-protein kinase]
species 1Sink
species 18[Phosphatidylinositol 3-kinase regulatory subunit alpha]
species 4[RAC-alpha serine/threonine-protein kinase]
species 16[Phosphatidylinositol 3-kinase regulatory subunit alpha]
species 20[Insulin receptor]
species 28Amino_Acids
species 47[Ribosomal protein S6 kinase beta-1]
species 21[Insulin receptor]
species 8[Tuberin; Hamartin]
species 17[Ribosomal protein S6 kinase beta-1]
species 12[Proline-rich AKT1 substrate 1]
species 5[Serine/threonine-protein kinase mTOR]
species 15[Insulin receptor]
species 2[Serine/threonine-protein kinase mTOR]
species 42[Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform; Insulin receptor substrate 1]
species 6[Hamartin; Tuberin]
species 19[Insulin receptor substrate 1; Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform]
species 10[Proline-rich AKT1 substrate 1]
species 11[Serine/threonine-protein kinase mTOR]
species 14[DNA-dependent protein kinase catalytic subunit; Serine-protein kinase ATM; Serine/threonine-protein kinase PAK 1; MAP kinase-activated protein kinase 2]
species 22[Serine/threonine-protein kinase mTOR]
species 3[RAC-alpha serine/threonine-protein kinase]
species 7[Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform; Insulin receptor substrate 1]
species 41[Insulin]
species 13[MAP kinase-activated protein kinase 2; DNA-dependent protein kinase catalytic subunit; Serine-protein kinase ATM; Serine/threonine-protein kinase PAK 1]

DallePezze2014 - Cellular senescene-induced mitochondrial dysfunction: BIOMD0000000582v0.0.1

DallePazze2014 - Cellular senescene-induced mitochondrial dysfunctionThis model is described in the article: [Dynamic m…

Details

Cellular senescence, a state of irreversible cell cycle arrest, is thought to help protect an organism from cancer, yet also contributes to ageing. The changes which occur in senescence are controlled by networks of multiple signalling and feedback pathways at the cellular level, and the interplay between these is difficult to predict and understand. To unravel the intrinsic challenges of understanding such a highly networked system, we have taken a systems biology approach to cellular senescence. We report a detailed analysis of senescence signalling via DNA damage, insulin-TOR, FoxO3a transcription factors, oxidative stress response, mitochondrial regulation and mitophagy. We show in silico and in vitro that inhibition of reactive oxygen species can prevent loss of mitochondrial membrane potential, whilst inhibition of mTOR shows a partial rescue of mitochondrial mass changes during establishment of senescence. Dual inhibition of ROS and mTOR in vitro confirmed computational model predictions that it was possible to further reduce senescence-induced mitochondrial dysfunction and DNA double-strand breaks. However, these interventions were unable to abrogate the senescence-induced mitochondrial dysfunction completely, and we identified decreased mitochondrial fission as the potential driving force for increased mitochondrial mass via prevention of mitophagy. Dynamic sensitivity analysis of the model showed the network stabilised at a new late state of cellular senescence. This was characterised by poor network sensitivity, high signalling noise, low cellular energy, high inflammation and permanent cell cycle arrest suggesting an unsatisfactory outcome for treatments aiming to delay or reverse cellular senescence at late time points. Combinatorial targeted interventions are therefore possible for intervening in the cellular pathway to senescence, but in the cases identified here, are only capable of delaying senescence onset. link: http://identifiers.org/pubmed/25166345

Parameters:

NameDescription
scale_Mito_Membr_Pot_obs = 1.0Reaction: Mito_Membr_Pot_obs = scale_Mito_Membr_Pot_obs*(Mito_membr_pot_new+Mito_membr_pot_old), Rate Law: missing
DNA_damaged_by_irradiation = 9237.72311545872Reaction: => DNA_damage; Irradiation, Irradiation, Rate Law: Cell*DNA_damaged_by_irradiation*Irradiation
mitophagy_activ_by_FoxO3a_n_AMPK_pT172 = 1319.84219165251Reaction: => Mitophagy; FoxO3a, AMPK_pT172, AMPK_pT172, FoxO3a, Rate Law: Cell*mitophagy_activ_by_FoxO3a_n_AMPK_pT172*FoxO3a*AMPK_pT172
IKKbeta_inactiv = 1.0Reaction: IKKbeta => Nil; IKKbeta, Rate Law: Cell*IKKbeta_inactiv*IKKbeta
mTORC1_pS2448_dephos_by_AMPK_pT172 = 191.297262771509Reaction: mTORC1_pS2448 => mTORC1; AMPK_pT172, AMPK_pT172, mTORC1_pS2448, Rate Law: Cell*mTORC1_pS2448_dephos_by_AMPK_pT172*mTORC1_pS2448*AMPK_pT172
mito_biogenesis_by_mTORC1_pS2448 = 0.0133620123598202Reaction: Mito_mass_turnover => Mito_mass_new; mTORC1_pS2448, Mito_mass_turnover, mTORC1_pS2448, Rate Law: Cell*mito_biogenesis_by_mTORC1_pS2448*Mito_mass_turnover*mTORC1_pS2448
mito_membr_pot_new_dec = 1094.58423149719Reaction: Mito_membr_pot_new => Nil; Mito_membr_pot_new, Rate Law: Cell*mito_membr_pot_new_dec*Mito_membr_pot_new
scale_Akt_pS473_obs = 1.0Reaction: Akt_pS473_obs = scale_Akt_pS473_obs*Akt_pS473, Rate Law: missing
Akt_pS473_dephos_by_mTORC1_pS2448 = 0.114598191621279Reaction: Akt_pS473 => Akt; mTORC1_pS2448, Akt_pS473, mTORC1_pS2448, Rate Law: Cell*Akt_pS473_dephos_by_mTORC1_pS2448*Akt_pS473*mTORC1_pS2448
scale_CDKN1A_obs = 1.0Reaction: CDKN1A_obs = scale_CDKN1A_obs*CDKN1A, Rate Law: missing
CDKN1A_transcr_by_FoxO3a_n_DNA_damage = 0.0852182335681166Reaction: => CDKN1A; DNA_damage, FoxO3a, DNA_damage, FoxO3a, Rate Law: Cell*CDKN1A_transcr_by_FoxO3a_n_DNA_damage*DNA_damage*FoxO3a
CDKN1B_transcr_by_FoxO3a_n_DNA_damage = 0.0920526565951487Reaction: => CDKN1B; DNA_damage, FoxO3a, DNA_damage, FoxO3a, Rate Law: Cell*CDKN1B_transcr_by_FoxO3a_n_DNA_damage*DNA_damage*FoxO3a
Akt_S473_phos_by_insulin = 0.588783148144923Reaction: Akt => Akt_pS473; Insulin, Akt, Insulin, Rate Law: Cell*Akt_S473_phos_by_insulin*Akt*Insulin
AMPK_pT172_dephos_by_Mito_membr_pot_old = 1.00000000000003E-6Reaction: AMPK_pT172 => AMPK; Mito_membr_pot_old, AMPK_pT172, Mito_membr_pot_old, Rate Law: Cell*AMPK_pT172_dephos_by_Mito_membr_pot_old*AMPK_pT172*Mito_membr_pot_old
AMPK_T172_phos = 0.355183987378767Reaction: AMPK => AMPK_pT172; AMPK, Rate Law: Cell*AMPK_T172_phos*AMPK
sen_ass_beta_gal_dec = 0.154821166783837Reaction: SA_beta_gal => ; SA_beta_gal, Rate Law: Cell*sen_ass_beta_gal_dec*SA_beta_gal
ROS_prod_by_Mito_membr_pot_old = 772.829490967078Reaction: => ROS; Mito_membr_pot_old, Mito_membr_pot_old, Rate Law: Cell*ROS_prod_by_Mito_membr_pot_old*Mito_membr_pot_old
mTORC1_S2448_phos_by_AA_n_Akt_pS473 = 162.471039450073Reaction: mTORC1 => mTORC1_pS2448; Amino_Acids, Akt_pS473, Akt_pS473, Amino_Acids, mTORC1, Rate Law: Cell*mTORC1_S2448_phos_by_AA_n_Akt_pS473*mTORC1*Amino_Acids*Akt_pS473
scale_CDKN1B_obs = 1.0Reaction: CDKN1B_obs = scale_CDKN1B_obs*CDKN1B, Rate Law: missing
mitophagy_new = 0.22465992989378Reaction: Mito_mass_new => Mito_mass_turnover; Mitophagy, Mito_mass_new, Mitophagy, Rate Law: Cell*mitophagy_new*Mito_mass_new*Mitophagy
mitophagy_old = 0.00122607614891116Reaction: Mito_mass_old => Mito_mass_turnover; Mitophagy, Mito_mass_old, Mitophagy, Rate Law: Cell*mitophagy_old*Mito_mass_old*Mitophagy
ROS_turnover = 3.23082321168464Reaction: ROS => Nil; ROS, Rate Law: Cell*ROS_turnover*ROS
DNA_damaged_by_ROS = 0.118873655169353Reaction: => DNA_damage; ROS, ROS, Rate Law: Cell*DNA_damaged_by_ROS*ROS
scale_JNK_pT183_obs = 1.0Reaction: JNK_pT183_obs = scale_JNK_pT183_obs*JNK_pT183, Rate Law: missing
mito_membr_pot_old_inc = 0.00586017882122243Reaction: => Mito_membr_pot_old; Mito_mass_old, Mito_mass_old, Rate Law: Cell*mito_membr_pot_old_inc*Mito_mass_old
DNA_repair = 0.325724769122274Reaction: DNA_damage => Nil; DNA_damage, Rate Law: Cell*DNA_repair*DNA_damage
mito_dysfunction = 0.0270695257507146Reaction: Mito_mass_new => Mito_mass_old; CDKN1A, CDKN1A, Mito_mass_new, Rate Law: Cell*mito_dysfunction*Mito_mass_new*CDKN1A
FoxO3a_phos_by_JNK_pT183 = 0.112877630496044Reaction: FoxO3a_pS253 => FoxO3a; JNK_pT183, FoxO3a_pS253, JNK_pT183, Rate Law: Cell*FoxO3a_phos_by_JNK_pT183*FoxO3a_pS253*JNK_pT183
scale_Mitophagy_obs = 1.0Reaction: Mitophagy_obs = scale_Mitophagy_obs*Mitophagy, Rate Law: missing
JNK_pT183_inactiv = 0.0718429173444438Reaction: JNK_pT183 => JNK; JNK_pT183, Rate Law: Cell*JNK_pT183_inactiv*JNK_pT183
mito_membr_pot_old_dec = 0.954903499913184Reaction: Mito_membr_pot_old => Nil; Mito_membr_pot_old, Rate Law: Cell*mito_membr_pot_old_dec*Mito_membr_pot_old
mitophagy_inactiv_by_mTORC1_pS2448 = 645.999307230137Reaction: Mitophagy => Nil; mTORC1_pS2448, Mitophagy, mTORC1_pS2448, Rate Law: Cell*mitophagy_inactiv_by_mTORC1_pS2448*Mitophagy*mTORC1_pS2448
FoxO3a_pS253_degrad = 39.4068609318082Reaction: FoxO3a_pS253 => Nil; FoxO3a_pS253, Rate Law: Cell*FoxO3a_pS253_degrad*FoxO3a_pS253
JNK_activ_by_ROS = 0.00502329152478409Reaction: JNK => JNK_pT183; ROS, JNK, ROS, Rate Law: Cell*JNK_activ_by_ROS*JNK*ROS
scale_FoxO3a_pS253_obs = 1.0Reaction: FoxO3a_pS253_obs = scale_FoxO3a_pS253_obs*FoxO3a_pS253, Rate Law: missing
mTORC1_S2448_phos_by_AA_n_IKKbeta = 1.00008996727694E-5Reaction: mTORC1 => mTORC1_pS2448; Amino_Acids, IKKbeta, Amino_Acids, IKKbeta, mTORC1, Rate Law: Cell*mTORC1_S2448_phos_by_AA_n_IKKbeta*mTORC1*Amino_Acids*IKKbeta
mTORC1_S2448_phos_by_AA = 1.00008999860285E-6Reaction: mTORC1 => mTORC1_pS2448; Amino_Acids, Amino_Acids, mTORC1, Rate Law: Cell*mTORC1_S2448_phos_by_AA*mTORC1*Amino_Acids
CDKN1A_inactiv_by_Akt_pS473 = 0.0667971061916905Reaction: CDKN1A => Nil; Akt_pS473, Akt_pS473, CDKN1A, Rate Law: Cell*CDKN1A_inactiv_by_Akt_pS473*CDKN1A*Akt_pS473
mito_membr_pot_new_inc = 9882.02736076158Reaction: => Mito_membr_pot_new; Mito_mass_new, Mito_mass_new, Rate Law: Cell*mito_membr_pot_new_inc*Mito_mass_new
FoxO3a_synthesis = 407.307409980937Reaction: => FoxO3a, Rate Law: Cell*FoxO3a_synthesis
scale_mTOR_pS2448_obs = 1.0Reaction: mTOR_pS2448_obs = scale_mTOR_pS2448_obs*mTORC1_pS2448, Rate Law: missing
scale_AMPK_pT172_obs = 1.0Reaction: AMPK_pT172_obs = scale_AMPK_pT172_obs*AMPK_pT172, Rate Law: missing
sen_ass_beta_gal_inc_by_Mitophagy = 1.00000000000011E-6Reaction: => SA_beta_gal; Mitophagy, Mitophagy, Rate Law: Cell*sen_ass_beta_gal_inc_by_Mitophagy*Mitophagy
CDKN1B_inactiv_by_Akt_pS473 = 0.0596841598127919Reaction: CDKN1B => Nil; Akt_pS473, Akt_pS473, CDKN1B, Rate Law: Cell*CDKN1B_inactiv_by_Akt_pS473*CDKN1B*Akt_pS473
ROS_prod_by_Mito_membr_pot_new = 4.55464788075885Reaction: => ROS; Mito_membr_pot_new, Mito_membr_pot_new, Rate Law: Cell*ROS_prod_by_Mito_membr_pot_new*Mito_membr_pot_new
scale_DNA_damage_gammaH2AX_obs = 1.0Reaction: DNA_damage_gammaH2AX_obs = scale_DNA_damage_gammaH2AX_obs*DNA_damage, Rate Law: missing
scale_FoxO3a_total_obs = 1.0Reaction: FoxO3a_total_obs = scale_FoxO3a_total_obs*(FoxO3a+FoxO3a_pS253), Rate Law: missing
FoxO3a_phos_by_Akt_pS473 = 6.83511123229576Reaction: FoxO3a => FoxO3a_pS253; Akt_pS473, Akt_pS473, FoxO3a, Rate Law: Cell*FoxO3a_phos_by_Akt_pS473*FoxO3a*Akt_pS473
scale_Mito_Mass_obs = 1.0Reaction: Mito_Mass_obs = scale_Mito_Mass_obs*(Mito_mass_new+Mito_mass_old), Rate Law: missing
IKKbeta_activ_by_ROS = 1.0Reaction: => IKKbeta; ROS, ROS, Rate Law: Cell*IKKbeta_activ_by_ROS*ROS
mito_biogenesis_by_AMPK_pT172 = 5.8915457309741E-5Reaction: Mito_mass_turnover => Mito_mass_new; mTORC1_pS2448, Mito_mass_turnover, mTORC1_pS2448, Rate Law: Cell*mito_biogenesis_by_AMPK_pT172*Mito_mass_turnover*mTORC1_pS2448
AMPK_pT172_dephos_by_Mito_membr_pot_new = 0.117744691539618Reaction: AMPK_pT172 => AMPK; Mito_membr_pot_new, AMPK_pT172, Mito_membr_pot_new, Rate Law: Cell*AMPK_pT172_dephos_by_Mito_membr_pot_new*AMPK_pT172*Mito_membr_pot_new
scale_SA_beta_gal_obs = 1.0Reaction: SA_beta_gal_obs = scale_SA_beta_gal_obs*SA_beta_gal, Rate Law: missing
sen_ass_beta_gal_inc_by_ROS = 0.0701139988718817Reaction: => SA_beta_gal; ROS, ROS, Rate Law: Cell*sen_ass_beta_gal_inc_by_ROS*ROS
scale_ROS_obs = 1.0Reaction: ROS_obs = scale_ROS_obs*ROS, Rate Law: missing

States:

NameDescription
Mito mass turnover[mitochondrion]
mTORC1 pS2448[TORC1 complex]
Mito membr pot new[mitochondrion]
DNA damage[deoxyribonucleic acid]
mTOR pS2448 obsmTOR_pS2448_obs
AMPK pT172 obsAMPK_pT172_obs
Mito membr pot old[mitochondrion]
CDKN1A[Cyclin-dependent kinase inhibitor 1]
Akt[RAC-alpha serine/threonine-protein kinase]
Mitophagy[autophagy of mitochondrion]
AMPK pT172[5'-AMP-activated protein kinase subunit beta-1; 5'-AMP-activated protein kinase catalytic subunit alpha-2]
CDKN1B obsCDKN1B_obs
FoxO3a total obsFoxO3a_total_obs
ROS obsROS_obs
IKKbeta[Inhibitor of nuclear factor kappa-B kinase subunit beta]
Mito Mass obsMito_Mass_obs
Akt pS473[RAC-alpha serine/threonine-protein kinase]
Nil[empty set]
Mito Membr Pot obsMito_Membr_Pot_obs
CDKN1A obsCDKN1A_obs
CDKN1B[CDKN1B proteinCyclin-dependent kinase inhibitor 1BCyclin-dependent kinase inhibitor 1B (P27, Kip1), isoform CRA_acDNA, FLJ92816, Homo sapiens cyclin-dependent kinase inhibitor 1B (p27, Kip1)(CDKN1B), mRNA]
SA beta gal obsSA_beta_gal_obs
ROS[reactive oxygen species]
Akt pS473 obsAkt_pS473_obs
Mito mass new[mitochondrion]
Mitophagy obsMitophagy_obs
Mito mass old[mitochondrion]
Insulin[insulin (human)]
FoxO3a pS253[Forkhead box protein O3]
mTORC1[TORC1 complex]
AMPK[5'-AMP-activated protein kinase subunit beta-1; 5'-AMP-activated protein kinase catalytic subunit alpha-2]
Amino Acids[amino acid]
Irradiation[SBO:0000405]
DNA damage gammaH2AX obsDNA_damage_gammaH2AX_obs
JNK pT183[Mitogen-activated protein kinase 8]
SA beta gal[Beta-galactosidase]
FoxO3a pS253 obsFoxO3a_pS253_obs
JNK pT183 obsJNK_pT183_obs
FoxO3a[Forkhead box protein O3]
JNK[Mitogen-activated protein kinase 8]

DallePezze2016 - Activation of AMPK and mTOR by amino acids: BIOMD0000000640v0.0.1

DallePezze2016 - Activation of AMPK and mTOR by amino acids (Model 3)This model is as described in the Supplementary So…

Details

Amino acids (aa) are not only building blocks for proteins, but also signalling molecules, with the mammalian target of rapamycin complex 1 (mTORC1) acting as a key mediator. However, little is known about whether aa, independently of mTORC1, activate other kinases of the mTOR signalling network. To delineate aa-stimulated mTOR network dynamics, we here combine a computational-experimental approach with text mining-enhanced quantitative proteomics. We report that AMP-activated protein kinase (AMPK), phosphatidylinositide 3-kinase (PI3K) and mTOR complex 2 (mTORC2) are acutely activated by aa-readdition in an mTORC1-independent manner. AMPK activation by aa is mediated by Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ). In response, AMPK impinges on the autophagy regulators Unc-51-like kinase-1 (ULK1) and c-Jun. AMPK is widely recognized as an mTORC1 antagonist that is activated by starvation. We find that aa acutely activate AMPK concurrently with mTOR. We show that AMPK under aa sufficiency acts to sustain autophagy. This may be required to maintain protein homoeostasis and deliver metabolite intermediates for biosynthetic processes. link: http://identifiers.org/pubmed/27869123

Parameters:

NameDescription
AMPK_pT172_dephos = 165.704Reaction: AMPK_pT172 => AMPK, Rate Law: Cell*AMPK_pT172_dephos*AMPK_pT172
AMPK_T172_phos_by_Amino_Acids = 17.6284Reaction: AMPK => AMPK_pT172; Amino_Acids, Rate Law: Cell*AMPK_T172_phos_by_Amino_Acids*AMPK*Amino_Acids
IRS_phos_by_p70_S6K_pT229_pT389 = 0.0863775267376444Reaction: IRS => IRS_pS636; p70_S6K_pT229_pT389, Rate Law: Cell*IRS_phos_by_p70_S6K_pT229_pT389*IRS*p70_S6K_pT229_pT389
IRS_phos_by_Amino_Acids = 0.0331672Reaction: IRS => IRS_p; Amino_Acids, Rate Law: Cell*IRS_phos_by_Amino_Acids*IRS*Amino_Acids
PRAS40_pS183_dephos_second = 1.88453Reaction: PRAS40_pT246_pS183 => PRAS40_pT246, Rate Law: Cell*PRAS40_pS183_dephos_second*PRAS40_pT246_pS183
IR_beta_pY1146_dephos = 0.493514Reaction: IR_beta_pY1146 => IR_beta_refractory, Rate Law: Cell*IR_beta_pY1146_dephos*IR_beta_pY1146
mTORC2_pS2481_dephos = 1.42511Reaction: mTORC2_pS2481 => mTORC2, Rate Law: Cell*mTORC2_pS2481_dephos*mTORC2_pS2481
mTORC1_S2448_activation_by_Amino_Acids = 0.0156992Reaction: mTORC1 => mTORC1_pS2448; Amino_Acids, Rate Law: Cell*mTORC1_S2448_activation_by_Amino_Acids*mTORC1*Amino_Acids
Akt_T308_phos_by_PI3K_p_PDK1_second = 7.47345Reaction: Akt_pS473 => Akt_pT308_pS473; PI3K_p_PDK1, Rate Law: Cell*Akt_T308_phos_by_PI3K_p_PDK1_second*Akt_pS473*PI3K_p_PDK1
PRAS40_pT246_dephos_second = 11.876Reaction: PRAS40_pT246_pS183 => PRAS40_pS183, Rate Law: Cell*PRAS40_pT246_dephos_second*PRAS40_pT246_pS183
PRAS40_pS183_dephos_first = 1.8706Reaction: PRAS40_pS183 => PRAS40, Rate Law: Cell*PRAS40_pS183_dephos_first*PRAS40_pS183
IRS_p_phos_by_p70_S6K_pT229_pT389 = 0.338859859949792Reaction: IRS_p => IRS_pS636; p70_S6K_pT229_pT389, Rate Law: Cell*IRS_p_phos_by_p70_S6K_pT229_pT389*IRS_p*p70_S6K_pT229_pT389
IR_beta_phos_by_Insulin = 0.0203796Reaction: IR_beta => IR_beta_pY1146; Insulin, Rate Law: Cell*IR_beta_phos_by_Insulin*IR_beta*Insulin
p70_S6K_T229_phos_by_PI3K_p_PDK1_second = 1.00000002814509E-6Reaction: p70_S6K_pT389 => p70_S6K_pT229_pT389; PI3K_p_PDK1, Rate Law: Cell*p70_S6K_T229_phos_by_PI3K_p_PDK1_second*p70_S6K_pT389*PI3K_p_PDK1
TSC1_TSC2_pT1462_dephos = 147.239Reaction: TSC1_TSC2_pT1462 => TSC1_TSC2, Rate Law: Cell*TSC1_TSC2_pT1462_dephos*TSC1_TSC2_pT1462
IR_beta_ready = 323.611Reaction: IR_beta_refractory => IR_beta, Rate Law: Cell*IR_beta_ready*IR_beta_refractory
p70_S6K_pT389_dephos_first = 1.10036057608758Reaction: p70_S6K_pT389 => p70_S6K, Rate Law: Cell*p70_S6K_pT389_dephos_first*p70_S6K_pT389
p70_S6K_pT229_dephos_first = 1.00000012897033E-6Reaction: p70_S6K_pT229 => p70_S6K, Rate Law: Cell*p70_S6K_pT229_dephos_first*p70_S6K_pT229
PI3K_p_PDK1_dephos = 0.18913343080532Reaction: PI3K_p_PDK1 => PI3K_PDK1, Rate Law: Cell*PI3K_p_PDK1_dephos*PI3K_p_PDK1
Akt_T308_phos_by_PI3K_p_PDK1_first = 7.47437Reaction: Akt => Akt_pT308; PI3K_p_PDK1, Rate Law: Cell*Akt_T308_phos_by_PI3K_p_PDK1_first*Akt*PI3K_p_PDK1
p70_S6K_pT229_dephos_second = 0.159201353240651Reaction: p70_S6K_pT229_pT389 => p70_S6K_pT389, Rate Law: Cell*p70_S6K_pT229_dephos_second*p70_S6K_pT229_pT389
p70_S6K_T389_phos_by_mTORC1_pS2448_first = 0.00261303413778722Reaction: p70_S6K => p70_S6K_pT389; mTORC1_pS2448, Rate Law: Cell*p70_S6K_T389_phos_by_mTORC1_pS2448_first*p70_S6K*mTORC1_pS2448
PRAS40_T246_phos_by_Akt_pT308_second = 0.279401Reaction: PRAS40_pS183 => PRAS40_pT246_pS183; Akt_pT308, Akt_pT308_pS473, Rate Law: Cell*PRAS40_T246_phos_by_Akt_pT308_second*PRAS40_pS183*(Akt_pT308+Akt_pT308_pS473)
PRAS40_S183_phos_by_mTORC1_pS2448_second = 0.0683009Reaction: PRAS40_pT246 => PRAS40_pT246_pS183; mTORC1_pS2448, Rate Law: Cell*PRAS40_S183_phos_by_mTORC1_pS2448_second*PRAS40_pT246*mTORC1_pS2448
mTORC2_S2481_phos_by_PI3K_variant_p = 0.120736Reaction: mTORC2 => mTORC2_pS2481; PI3K_variant_p, Rate Law: Cell*mTORC2_S2481_phos_by_PI3K_variant_p*mTORC2*PI3K_variant_p
TSC1_TSC2_T1462_phos_by_Akt_pT308 = 1.52417Reaction: TSC1_TSC2 => TSC1_TSC2_pT1462; Akt_pT308, Akt_pT308_pS473, Rate Law: Cell*TSC1_TSC2_T1462_phos_by_Akt_pT308*TSC1_TSC2*(Akt_pT308+Akt_pT308_pS473)
p70_S6K_T389_phos_by_mTORC1_pS2448_second = 0.110720890919343Reaction: p70_S6K_pT229 => p70_S6K_pT229_pT389; mTORC1_pS2448, Rate Law: Cell*p70_S6K_T389_phos_by_mTORC1_pS2448_second*p70_S6K_pT229*mTORC1_pS2448
mTORC1_pS2448_dephos_by_TSC1_TSC2 = 0.00869774Reaction: mTORC1_pS2448 => mTORC1; TSC1_TSC2, TSC1_TSC2_pS1387, Rate Law: Cell*mTORC1_pS2448_dephos_by_TSC1_TSC2*mTORC1_pS2448*(TSC1_TSC2+TSC1_TSC2_pS1387)
Akt_S473_phos_by_mTORC2_pS2481_second = 0.159093Reaction: Akt_pT308 => Akt_pT308_pS473; mTORC2_pS2481, Rate Law: Cell*Akt_S473_phos_by_mTORC2_pS2481_second*Akt_pT308*mTORC2_pS2481
TSC1_TSC2_pS1387_dephos = 0.25319Reaction: TSC1_TSC2_pS1387 => TSC1_TSC2, Rate Law: Cell*TSC1_TSC2_pS1387_dephos*TSC1_TSC2_pS1387
mTORC2_S2481_phos_by_Amino_Acids = 0.0268658Reaction: mTORC2 => mTORC2_pS2481; Amino_Acids, Rate Law: Cell*mTORC2_S2481_phos_by_Amino_Acids*mTORC2*Amino_Acids
Akt_pS473_dephos_second = 0.380005Reaction: Akt_pT308_pS473 => Akt_pT308, Rate Law: Cell*Akt_pS473_dephos_second*Akt_pT308_pS473
Akt_pT308_dephos_second = 88.9639Reaction: Akt_pT308_pS473 => Akt_pS473, Rate Law: Cell*Akt_pT308_dephos_second*Akt_pT308_pS473
TSC1_TSC2_S1387_phos_by_AMPK_pT172 = 0.00175772Reaction: TSC1_TSC2 => TSC1_TSC2_pS1387; AMPK_pT172, Rate Law: Cell*TSC1_TSC2_S1387_phos_by_AMPK_pT172*TSC1_TSC2*AMPK_pT172
Akt_S473_phos_by_mTORC2_pS2481_first = 1.31992E-5Reaction: Akt => Akt_pS473; mTORC2_pS2481, Rate Law: Cell*Akt_S473_phos_by_mTORC2_pS2481_first*Akt*mTORC2_pS2481
PRAS40_T246_phos_by_Akt_pT308_first = 0.279344Reaction: PRAS40 => PRAS40_pT246; Akt_pT308, Akt_pT308_pS473, Rate Law: Cell*PRAS40_T246_phos_by_Akt_pT308_first*PRAS40*(Akt_pT308+Akt_pT308_pS473)
PI3K_PDK1_phos_by_IRS_p = 1.87226757782201E-4Reaction: PI3K_PDK1 => PI3K_p_PDK1; IRS_p, Rate Law: Cell*PI3K_PDK1_phos_by_IRS_p*PI3K_PDK1*IRS_p
p70_S6K_T229_phos_by_PI3K_p_PDK1_first = 0.0133520172873009Reaction: p70_S6K => p70_S6K_pT229; PI3K_p_PDK1, Rate Law: Cell*p70_S6K_T229_phos_by_PI3K_p_PDK1_first*p70_S6K*PI3K_p_PDK1
Akt_pS473_dephos_first = 0.376999Reaction: Akt_pS473 => Akt, Rate Law: Cell*Akt_pS473_dephos_first*Akt_pS473
IRS_pS636_turnover = 25.0Reaction: IRS_pS636 => IRS, Rate Law: Cell*IRS_pS636_turnover*IRS_pS636
IRS_phos_by_IR_beta_pY1146 = 2.11894Reaction: IRS => IRS_p; IR_beta_pY1146, Rate Law: Cell*IRS_phos_by_IR_beta_pY1146*IRS*IR_beta_pY1146
PRAS40_pT246_dephos_first = 11.8759Reaction: PRAS40_pT246 => PRAS40, Rate Law: Cell*PRAS40_pT246_dephos_first*PRAS40_pT246
Akt_pT308_dephos_first = 88.9654Reaction: Akt_pT308 => Akt, Rate Law: Cell*Akt_pT308_dephos_first*Akt_pT308
PRAS40_S183_phos_by_mTORC1_pS2448_first = 0.15881Reaction: PRAS40 => PRAS40_pS183; mTORC1_pS2448, Rate Law: Cell*PRAS40_S183_phos_by_mTORC1_pS2448_first*PRAS40*mTORC1_pS2448
AMPK_T172_phos = 0.490602Reaction: AMPK => AMPK_pT172; IRS_p, Rate Law: Cell*AMPK_T172_phos*AMPK*IRS_p
p70_S6K_pT389_dephos_second = 1.10215267954479Reaction: p70_S6K_pT229_pT389 => p70_S6K_pT229, Rate Law: Cell*p70_S6K_pT389_dephos_second*p70_S6K_pT229_pT389

States:

NameDescription
mTORC1 pS2448mTORC1_pS2448
mTOR pS2448 obsmTOR_pS2448_obs
Akt pT308 pS473Akt_pT308_pS473
IR beta[Insulin receptor]
AMPK pT172AMPK_pT172
IRS pS636 obsIRS_pS636_obs
PRAS40 pT246 obsPRAS40_pT246_obs
p70 S6K pT389p70_S6K_pT389
mTOR pS2481 obsmTOR_pS2481_obs
p70 S6K pT229 obsp70_S6K_pT229_obs
TSC1 TSC2 pT1462TSC1_TSC2_pT1462
PI3K p PDK1PI3K_p_PDK1
mTORC1mTORC1
Amino AcidsAmino_Acids
IRS pS636IRS_pS636
PRAS40 pT246 pS183PRAS40_pT246_pS183
TSC1 TSC2 pS1387TSC1_TSC2_pS1387
IR beta pY1146 obsIR_beta_pY1146_obs
p70 S6Kp70_S6K
AMPK pT172 obsAMPK_pT172_obs
PRAS40PRAS40
AktAkt
p70 S6K pT229 pT389p70_S6K_pT229_pT389
PRAS40 pT246PRAS40_pT246
PRAS40 pS183PRAS40_pS183
Akt pS473Akt_pS473
IRS pIRS_p
PI3K PDK1PI3K_PDK1
PRAS40 pS183 obsPRAS40_pS183_obs
IR beta refractoryIR_beta_refractory
Akt pS473 obsAkt_pS473_obs
mTORC2mTORC2
IRS[Insulin receptor substrate 1]
Akt pT308Akt_pT308
TSC1 TSC2TSC1_TSC2
InsulinInsulin
IR beta pY1146IR_beta_pY1146
AMPK[5'-AMP-activated protein kinase catalytic subunit alpha-1]
p70 S6K pT229p70_S6K_pT229
p70 S6K pT389 obsp70_S6K_pT389_obs
mTORC2 pS2481mTORC2_pS2481
TSC1 TSC2 pS1387 obsTSC1_TSC2_pS1387_obs
Akt pT308 obsAkt_pT308_obs

DallePezze2016 - Activation of AMPK and mTOR by amino acids (Model 1): MODEL1705030000v0.0.1

DallePezze2016 - Activation of AMPK and mTOR by amino acids (Model 1)This model is as described in Supplementary Softwar…

Details

Amino acids (aa) are not only building blocks for proteins, but also signalling molecules, with the mammalian target of rapamycin complex 1 (mTORC1) acting as a key mediator. However, little is known about whether aa, independently of mTORC1, activate other kinases of the mTOR signalling network. To delineate aa-stimulated mTOR network dynamics, we here combine a computational-experimental approach with text mining-enhanced quantitative proteomics. We report that AMP-activated protein kinase (AMPK), phosphatidylinositide 3-kinase (PI3K) and mTOR complex 2 (mTORC2) are acutely activated by aa-readdition in an mTORC1-independent manner. AMPK activation by aa is mediated by Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ). In response, AMPK impinges on the autophagy regulators Unc-51-like kinase-1 (ULK1) and c-Jun. AMPK is widely recognized as an mTORC1 antagonist that is activated by starvation. We find that aa acutely activate AMPK concurrently with mTOR. We show that AMPK under aa sufficiency acts to sustain autophagy. This may be required to maintain protein homoeostasis and deliver metabolite intermediates for biosynthetic processes. link: http://identifiers.org/pubmed/27869123

DallePezze2016 - Activation of AMPK and mTOR by amino acids (Model 2): MODEL1705030001v0.0.1

DallePezze2016 - Activation of AMPK and mTOR by amino acids (Model 2)This model is as described in the Supplementary So…

Details

Amino acids (aa) are not only building blocks for proteins, but also signalling molecules, with the mammalian target of rapamycin complex 1 (mTORC1) acting as a key mediator. However, little is known about whether aa, independently of mTORC1, activate other kinases of the mTOR signalling network. To delineate aa-stimulated mTOR network dynamics, we here combine a computational-experimental approach with text mining-enhanced quantitative proteomics. We report that AMP-activated protein kinase (AMPK), phosphatidylinositide 3-kinase (PI3K) and mTOR complex 2 (mTORC2) are acutely activated by aa-readdition in an mTORC1-independent manner. AMPK activation by aa is mediated by Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ). In response, AMPK impinges on the autophagy regulators Unc-51-like kinase-1 (ULK1) and c-Jun. AMPK is widely recognized as an mTORC1 antagonist that is activated by starvation. We find that aa acutely activate AMPK concurrently with mTOR. We show that AMPK under aa sufficiency acts to sustain autophagy. This may be required to maintain protein homoeostasis and deliver metabolite intermediates for biosynthetic processes. link: http://identifiers.org/pubmed/27869123

Danø2006_Glycolysis_Reduction: MODEL5952308332v0.0.1

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

Details

The complexity of full-scale metabolic models is a major obstacle for their effective use in computational systems biology. The aim of model reduction is to circumvent this problem by eliminating parts of a model that are unimportant for the properties of interest. The choice of reduction method is influenced both by the type of model complexity and by the objective of the reduction; therefore, no single method is superior in all cases. In this study we present a comparative study of two different methods applied to a 20D model of yeast glycolytic oscillations. Our objective is to obtain biochemically meaningful reduced models, which reproduce the dynamic properties of the 20D model. The first method uses lumping and subsequent constrained parameter optimization. The second method is a novel approach that eliminates variables not essential for the dynamics. The applications of the two methods result in models of eight (lumping), six (elimination) and three (lumping followed by elimination) dimensions. All models have similar dynamic properties and pin-point the same interactions as being crucial for generation of the oscillations. The advantage of the novel method is that it is algorithmic, and does not require input in the form of biochemical knowledge. The lumping approach, however, is better at preserving biochemical properties, as we show through extensive analyses of the models. link: http://identifiers.org/pubmed/17010168

Das2010 - Effect of a gamma-secretase inhibitor on Amyloid-beta dynamics: BIOMD0000000551v0.0.1

Das2010 - Effect of a gamma-secretase inhibitor on Amyloid-beta dynamicsThis model is described in the article: [Modeli…

Details

Aggregation of the small peptide amyloid beta (Aβ) into oligomers and fibrils in the brain is believed to be a precursor to Alzheimer's disease. Aβ is produced via multiple proteolytic cleavages of amyloid precursor protein (APP), mediated by the enzymes β- and γ-secretase. In this study, we examine the temporal dynamics of soluble (unaggregated) Aβ in the plasma and cerebral-spinal fluid (CSF) of rhesus monkeys treated with different oral doses of a γ-secretase inhibitor. A dose-dependent reduction of Aβ concentration was observed within hours of drug ingestion, for all doses tested. Aβ concentration in the CSF returned to its predrug level over the monitoring period. In contrast, Aβ concentration in the plasma exhibited an unexpected overshoot to as high as 200% of the predrug concentration, and this overshoot persisted as late as 72 hours post-drug ingestion. To account for these observations, we proposed and analyzed a minimal physiological model for Aβ dynamics that could fit the data. Our analysis suggests that the overshoot arises from the attenuation of an Aβ clearance mechanism, possibly due to the inhibitor. Our model predicts that the efficacy of Aβ clearance recovers to its basal (pretreatment) value with a characteristic time of >48 hours, matching the time-scale of the overshoot. These results point to the need for a more detailed investigation of soluble Aβ clearance mechanisms and their interaction with Aβ-reducing drugs. link: http://identifiers.org/pubmed/20411345

Parameters:

NameDescription
k1 = 1.13; r = 0.43; deltap = 0.55; J = 0.0; l = 1.0Reaction: P = (k1*r*C-J*r)-deltap*P*l, Rate Law: (k1*r*C-J*r)-deltap*P*l
Ki = 0.0232; k1 = 1.13; Sc = 1.16; g_t = 0.0; J = 0.0Reaction: C = (Sc/(1+g_t/Ki)-k1*C)+J, Rate Law: (Sc/(1+g_t/Ki)-k1*C)+J

States:

NameDescription
C[Amyloid beta A4 protein]
P[Amyloid beta A4 protein]

Dasgupta2020 - Reduced model of receptor clusturing and aggregation: BIOMD0000000973v0.0.1

a simple kinetic mass-action-law-based model could be utilized to adequately describe clustering in response to activat…

Details

The process of clustering of plasma membrane receptors in response to their agonist is the first step in signal transduction. The rate of the clustering process and the size of the clusters determine further cell responses. Here we aim to demonstrate that a simple 2-differential equation mathematical model is capable of quantitative description of the kinetics of 2D or 3D cluster formation in various processes. Three mathematical models based on mass action kinetics were considered and compared with each other by their ability to describe experimental data on GPVI or CR3 receptor clustering (2D) and albumin or platelet aggregation (3D) in response to activation. The models were able to successfully describe experimental data without losing accuracy after switching between complex and simple models. However, additional restrictions on parameter values are required to match a single set of parameters for the given experimental data. The extended clustering model captured several properties of the kinetics of cluster formation, such as the existence of only three typical steady states for this system: unclustered receptors, receptor dimers, and clusters. Therefore, a simple kinetic mass-action-law-based model could be utilized to adequately describe clustering in response to activation both in 2D and in 3D. link: http://identifiers.org/pubmed/32604803

David2008 - Genome-scale metabolic network of Aspergillus nidulans (iHD666): MODEL1507180016v0.0.1

David2008 - Genome-scale metabolic network of Aspergillus nidulans (iHD666)This model is described in the article: [Ana…

Details

BACKGROUND: Aspergillus nidulans is a member of a diverse group of filamentous fungi, sharing many of the properties of its close relatives with significance in the fields of medicine, agriculture and industry. Furthermore, A. nidulans has been a classical model organism for studies of development biology and gene regulation, and thus it has become one of the best-characterized filamentous fungi. It was the first Aspergillus species to have its genome sequenced, and automated gene prediction tools predicted 9,451 open reading frames (ORFs) in the genome, of which less than 10% were assigned a function. RESULTS: In this work, we have manually assigned functions to 472 orphan genes in the metabolism of A. nidulans, by using a pathway-driven approach and by employing comparative genomics tools based on sequence similarity. The central metabolism of A. nidulans, as well as biosynthetic pathways of relevant secondary metabolites, was reconstructed based on detailed metabolic reconstructions available for A. niger and Saccharomyces cerevisiae, and information on the genetics, biochemistry and physiology of A. nidulans. Thereby, it was possible to identify metabolic functions without a gene associated, and to look for candidate ORFs in the genome of A. nidulans by comparing its sequence to sequences of well-characterized genes in other species encoding the function of interest. A classification system, based on defined criteria, was developed for evaluating and selecting the ORFs among the candidates, in an objective and systematic manner. The functional assignments served as a basis to develop a mathematical model, linking 666 genes (both previously and newly annotated) to metabolic roles. The model was used to simulate metabolic behavior and additionally to integrate, analyze and interpret large-scale gene expression data concerning a study on glucose repression, thereby providing a means of upgrading the information content of experimental data and getting further insight into this phenomenon in A. nidulans. CONCLUSION: We demonstrate how pathway modeling of A. nidulans can be used as an approach to improve the functional annotation of the genome of this organism. Furthermore we show how the metabolic model establishes functional links between genes, enabling the upgrade of the information content of transcriptome data. link: http://identifiers.org/pubmed/18405346

Day2015 - Early cellular innate and adaptive immune responses to ischemia/reperfusion injury and solid organ allotransplantation: MODEL1911190001v0.0.1

This is an ordinary differential equation model of the early inflammatory response during transplantion. Descriptions ar…

Details

A mathematical model of the early inflammatory response in transplantation is formulated with ordinary differential equations. We first consider the inflammatory events associated only with the initial surgical procedure and the subsequent ischemia/reperfusion (I/R) events that cause tissue damage to the host as well as the donor graft. These events release damage-associated molecular pattern molecules (DAMPs), thereby initiating an acute inflammatory response. In simulations of this model, resolution of inflammation depends on the severity of the tissue damage caused by these events and the patient's (co)-morbidities. We augment a portion of a previously published mathematical model of acute inflammation with the inflammatory effects of T cells in the absence of antigenic allograft mismatch (but with DAMP release proportional to the degree of graft damage prior to transplant). Finally, we include the antigenic mismatch of the graft, which leads to the stimulation of potent memory T cell responses, leading to further DAMP release from the graft and concomitant increase in allograft damage. Regulatory mechanisms are also included at the final stage. Our simulations suggest that surgical injury and I/R-induced graft damage can be well-tolerated by the recipient when each is present alone, but that their combination (along with antigenic mismatch) may lead to acute rejection, as seen clinically in a subset of patients. An emergent phenomenon from our simulations is that low-level DAMP release can tolerize the recipient to a mismatched allograft, whereas different restimulation regimens resulted in an exaggerated rejection response, in agreement with published studies. We suggest that mechanistic mathematical models might serve as an adjunct for patient- or sub-group-specific predictions, simulated clinical studies, and rational design of immunosuppression. link: http://identifiers.org/doi/10.3389/fimmu.2015.00484

deBack2012 - Lineage Specification in Pancreas Development: BIOMD0000000435v0.0.1

deBack2012 - Lineage Specification in Pancreas DevelopmentThis model of two neighbouring pancreas precursor cells, descr…

Details

The cell fate decision of multi-potent pancreatic progenitor cells between the exocrine and endocrine lineages is regulated by Notch signalling, mediated by cell-cell interactions. However, canonical models of Notch-mediated lateral inhibition cannot explain the scattered spatial distribution of endocrine cells and the cell-type ratio in the developing pancreas. Based on evidence from acinar-to-islet cell transdifferentiation in vitro, we propose that lateral stabilization, i.e. positive feedback between adjacent progenitor cells, acts in parallel with lateral inhibition to regulate pattern formation in the pancreas. A simple mathematical model of transcriptional regulation and cell-cell interaction reveals the existence of multi-stability of spatial patterns whose simultaneous occurrence causes scattering of endocrine cells in the presence of noise. The scattering pattern allows for control of the endocrine-to-exocrine cell-type ratio by modulation of lateral stabilization strength. These theoretical results suggest a previously unrecognized role for lateral stabilization in lineage specification, spatial patterning and cell-type ratio control in organ development. link: http://identifiers.org/pubmed/23193107

Parameters:

NameDescription
k1=1.0Reaction: species_4 => ; species_4, Rate Law: compartment_1*k1*species_4
b=21.0; theta=1.0E-4; c=1.0; n=4.0Reaction: => species_2; species_2, species_4, species_1, species_2, species_4, species_1, Rate Law: compartment_1*(theta+b*(species_2*species_4)^n)/(theta+c*species_1^n+b*(species_2*species_4)^n)
a=1.0; theta=1.0E-4; n=4.0Reaction: => species_3; species_1, species_1, Rate Law: compartment_1*theta/(theta+a*species_1^n)

States:

NameDescription
species 2[Pancreas transcription factor 1 subunit alpha]
species 3[Neurogenin-3]
species 1[Neurogenin-3]
species 4[Pancreas transcription factor 1 subunit alpha]

DeCaluwe2016 - Circadian Clock: BIOMD0000000631v0.0.1

DeCaluwé2016 - Circadian ClockThis model is described in the article: [A Compact Model for the Complex Plant Circadian…

Details

The circadian clock is an endogenous timekeeper that allows organisms to anticipate and adapt to the daily variations of their environment. The plant clock is an intricate network of interlocked feedback loops, in which transcription factors regulate each other to generate oscillations with expression peaks at specific times of the day. Over the last decade, mathematical modeling approaches have been used to understand the inner workings of the clock in the model plant Arabidopsis thaliana. Those efforts have produced a number of models of ever increasing complexity. Here, we present an alternative model that combines a low number of equations and parameters, similar to the very earliest models, with the complex network structure found in more recent ones. This simple model describes the temporal evolution of the abundance of eight clock gene mRNA/protein and captures key features of the clock on a qualitative level, namely the entrained and free-running behaviors of the wild type clock, as well as the defects found in knockout mutants (such as altered free-running periods, lack of entrainment, or changes in the expression of other clock genes). Additionally, our model produces complex responses to various light cues, such as extreme photoperiods and non-24 h environmental cycles, and can describe the control of hypocotyl growth by the clock. Our model constitutes a useful tool to probe dynamical properties of the core clock as well as clock-dependent processes. link: http://identifiers.org/pubmed/26904049

Parameters:

NameDescription
K12 = 0.56; g2 = 0.12Reaction: => hypocotyl; PIF_p, Rate Law: g2*PIF_p^2/(K12^2+PIF_p^2)
k3 = 0.56Reaction: P51_m =>, Rate Law: k3*P51_m
K5 = 0.3; K4 = 0.23; v2A = 1.27Reaction: => P97_m; P51_p, EL_p, Rate Law: v2A/(1+(P51_p/K4)^2+(EL_p/K5)^2)
d3D = 0.48; D = 0.0Reaction: P51_p =>, Rate Law: d3D*D*P51_p
p5 = 0.62Reaction: => PIF_p; PIF_m, Rate Law: p5*PIF_m
p1 = 0.76Reaction: => CL_p; CL_m, Rate Law: p1*CL_m
k1D = 0.21; D = 0.0Reaction: CL_m =>, Rate Law: k1D*D*CL_m
p3 = 0.64Reaction: => P51_p; P51_m, Rate Law: p3*P51_m
K2 = 1.18; v1 = 4.58; K1 = 0.16Reaction: => CL_m; P51_p, P97_p, Rate Law: v1/(1+(P97_p/K1)^2+(P51_p/K2)^2)
d5D = 0.52; D = 0.0Reaction: PIF_p =>, Rate Law: d5D*D*PIF_p
d2D = 0.5; D = 0.0Reaction: P97_p =>, Rate Law: d2D*D*P97_p
k4 = 0.57Reaction: EL_m =>, Rate Law: k4*EL_m
L = 1.0; d2L = 0.29Reaction: P97_p =>, Rate Law: d2L*L*P97_p
L = 1.0; d5L = 4.0Reaction: PIF_p =>, Rate Law: d5L*L*PIF_p
K2 = 1.18; L = 1.0; v1L = 3.0; K1 = 0.16Reaction: => CL_m; P51_p, P97_p, P, Rate Law: v1L*L*P/(1+(P97_p/K1)^2+(P51_p/K2)^2)
K8 = 0.36; L = 1.0; v4 = 1.47; K10 = 1.9; K9 = 1.9Reaction: => EL_m; P51_p, CL_p, EL_p, Rate Law: L*v4/(1+(CL_p/K8)^2+(P51_p/K9)^2+(EL_p/K10)^2)
v3 = 1.0; K7 = 2.0; K6 = 0.46Reaction: => P51_m; P51_p, CL_p, Rate Law: v3/(1+(CL_p/K6)^2+(P51_p/K7)^2)
d4D = 1.21; D = 0.0Reaction: EL_p =>, Rate Law: d4D*D*EL_p
d4L = 0.38; L = 1.0Reaction: EL_p =>, Rate Law: d4L*L*EL_p
p2 = 1.01Reaction: => P97_p; P97_m, Rate Law: p2*P97_m
D = 0.0Reaction: => P, Rate Law: 0.3*(1-P)*D
L = 1.0; p1L = 0.42Reaction: => CL_p; CL_m, Rate Law: p1L*L*CL_m
p4 = 1.01Reaction: => EL_p; EL_m, Rate Law: p4*EL_m
L = 1.0Reaction: P =>, Rate Law: P*L
v5 = 0.1; K11 = 0.21Reaction: => PIF_m; EL_p, Rate Law: v5/(1+(EL_p/K11)^2)
d3L = 0.78; L = 1.0Reaction: P51_p =>, Rate Law: d3L*L*P51_p
v2L = 5.0; L = 1.0; K5 = 0.3; K4 = 0.23Reaction: => P97_m; P51_p, EL_p, P, Rate Law: v2L*L*P/(1+(P51_p/K4)^2+(EL_p/K5)^2)
d1 = 0.68Reaction: CL_p =>, Rate Law: d1*CL_p
g1 = 0.01Reaction: => hypocotyl, Rate Law: g1
k2 = 0.35Reaction: P97_m =>, Rate Law: k2*P97_m
v2B = 1.48; K5 = 0.3; K3 = 0.24; K4 = 0.23Reaction: => P97_m; P51_p, CL_p, EL_p, Rate Law: v2B*CL_p^2/(K3^2+CL_p^2)/(1+(P51_p/K4)^2+(EL_p/K5)^2)
k5 = 0.14Reaction: PIF_m =>, Rate Law: k5*PIF_m
L = 1.0; k1L = 0.53Reaction: CL_m =>, Rate Law: k1L*L*CL_m

States:

NameDescription
CL p[Protein LHY; Protein CCA1]
P97 p[Two-component response regulator-like APRR9; Two-component response regulator-like APRR7]
EL p[Transcription factor LUX; Protein EARLY FLOWERING 4]
EL m[823817; 818596]
P[Transcription factor PIF3; Transcription factor PIL1]
PIF m[825075; 818903]
PIF p[Transcription factor PIF5; Transcription factor PIF4]
P51 m[836259; 832518]
hypocotyl[hypocotyl]
P51 p[Two-component response regulator-like APRR1; Two-component response regulator-like APRR5]
P97 m[831793; 819292]
CL m[839341; 819296]

Decroly1982_Enzymatic_Oscillator: BIOMD0000000319v0.0.1

This is the scaled model described in the article: **Birhythmicity, chaos, and other patterns of temporal self-organiza…

Details

We analyze on a model biochemical system the effect of a coupling between two instability-generating mechanisms. The system considered is that of two allosteric enzymes coupled in series and activated by their respective products. In addition to simple periodic oscillations, the system can exhibit a variety of new modes of dynamic behavior; coexistence between two stable periodic regimes (birhythmicity), random oscillations (chaos), and coexistence of a stable periodic regime with a stable steady state (hard excitation) or with chaos. The relationship between these patterns of temporal self-organization is analyzed as a function of the control parameters of the model. Chaos and birhythmicity appear to be rare events in comparison with simple periodic behavior. We discuss the relevance of these results with respect to the regularity of most biological rhythms. link: http://identifiers.org/pubmed/6960354

Parameters:

NameDescription
L1=5.0E8 dimensionless; sigma1=10.0 per secReaction: alpha => beta, Rate Law: sigma1*alpha*(1+alpha)*(1+beta)^2/(L1+(1+alpha)^2*(1+beta)^2)
v_Km1=0.45 per secReaction: => alpha, Rate Law: v_Km1
ks=1.99 per secReaction: gamma =>, Rate Law: ks*gamma
d=0.0 dimensionless; sigma2=10.0 per sec; L2=100.0 dimensionlessReaction: beta => gamma, Rate Law: sigma2*beta*(1+d*beta)*(1+gamma)^2/(L2+(1+d*beta)^2*(1+gamma)^2)

States:

NameDescription
alphaalpha
gammagamma
betabeta

Deineko2003_CellCycle: BIOMD0000000208v0.0.1

The model reproduces Fig 3 of the paper corresponding to the transition to S phase. Units have not been defined for this…

Details

The study of the molecular mechanisms determining cellular programs of proliferation, differentiation, and apoptosis is currently attracting much attention. Recent studies have demonstrated that the system of cell-cycle control based on the transcriptional regulation of the expression of specific genes is responsible for the transition between programs. These groups of functionally connected genes from so-called gene networks characterized by numerous feedbacks and a complex behavioral dynamics. Computer simulation methods have been applied to studying the dynamics of gene networks regulating the cell cycle of vertebrates. The data on the regulation of the key genes obtained from the CYCLE-TRRD database have been used as a basis to construct gene networks of different degrees of complexity controlling the G1/S transition, one of the most important stages of the cell cycle. The behavior dynamics of the model constructed has been analyzed. Two qualitatively different functional modes of the system has been obtained. It has also been shown that the transition between these modes depends on the duration of the proliferation signal. It has also been demonstrated that the additional feedback from factor E2F to genes c-fos and c-jun, which was predicted earlier based on the computer analysis of promoters, plays an important role in the transition of the cell to the S phase. link: http://identifiers.org/pubmed/14582399

Parameters:

NameDescription
phi6 = 0.1Reaction: y6 =>, Rate Law: phi6*y6
phi1 = 0.1Reaction: y1 =>, Rate Law: phi1*y1
k4i = 1.0Reaction: y4 => y5, Rate Law: k4i*y4*y5
emax = 2.0; k1 = 1.0; k1_prime = 1.0; k1_double_prime = 10.0Reaction: => y1; y2, Rate Law: emax*k1*y1/(k1*y1+(k1_prime+k1_double_prime*y1)*y2)
k4 = 0.09Reaction: => y4; y1, Rate Law: k4*y1
k4_double_prime = 0.1Reaction: => y4; y6, Rate Law: k4_double_prime*y6
k4a = 2.0Reaction: y5 => y4, Rate Law: k4a*y5
phi2 = 0.01Reaction: y2 =>, Rate Law: phi2*y2
F6 = 0.044Reaction: => y6, Rate Law: F6
k6 = 0.0Reaction: => y6; y1, Rate Law: k6*y1
phi4a = 0.01Reaction: y5 =>, Rate Law: phi4a*y5
k2 = 1.0Reaction: => y2; y1, Rate Law: k2*y1
k3 = 0.4Reaction: y2 => y3; y5, Rate Law: k3*y2*y5
phi4i = 0.01Reaction: y4 =>, Rate Law: phi4i*y4
phi3 = 0.1Reaction: y3 =>, Rate Law: phi3*y3

States:

NameDescription
y3[Retinoblastoma-associated protein]
y1[Transcription factor E2F1]
y4[Cyclin-dependent kinase 2; G1/S-specific cyclin-E1]
y2[Retinoblastoma-associated protein]
y6[Transcription factor AP-1]
y5[Cyclin-dependent kinase 2; G1/S-specific cyclin-E1]

Del_Conte_Zerial2008_Rab5_Rab7_cut_out_switch: BIOMD0000000174v0.0.1

Key cellular functions and developmental processes rely on cascades of GTPases. GTPases of the Rab family provide a mole…

Details

Cut-out switch model

Membrane identity and GTPase cascades regulated by toggle and cut-out switches

Perla Del Conte-Zerial, Lutz Brusch, Jochen C Rink, Claudio Collinet, Yannis Kalaidzidis, Marino Zerial, and Andreas Deutsch: Molecular Systems Biology, 4:206, 15 July 2008, doi:10.1038/msb.2008.45

This is the cut-out switch model for the Rab5 - Rab7 transition, also referred to as model 2 in the original publication.

This model is not completely described in all details in the publication. Thanks go to Barbara Szomolay and Lutz Brusch for finding and clarifying this. According to Dr. Brusch this model represents the mechanism identified by the qualitative analysis in the article in the scenario deemed most useful by the authors. For the time-course simulations it was necessary to add a time dependency to one of the parameters, which is only verbally described in the article.

As argued in the publication the switch between early and late endosomes can be triggered by a parameter change. While with fixed parameter values each switch just converges to one steady state from its initial conditions and stays there, endosomes should switch between two different states. These changes would in reality of course depend on many different factors, such as cargo composition and amount in the specific endosome, its location and some additional cellular control mechanisms and encompass many different parameters. To keep the model simple the authors chose to add a time dependency to only one reaction - ke in the activation of RAB5 is multiplied with a term monotonously increasing over time from 0 to 1. They also hard coded a time dependence in this term, 100 minutes, to make the switch occur after several hundred minutes. As long as this modulating term remains monotonic all resulting time courses should look similar, with the switching behavior depending on the initial conditions and whether the term is increasing or decreasing. Monotonic increase is a reasonable assumption for the described mechanism of cargo accumulation.

Not explicitly described in the article:

activation of Rab5 (time): $r*ke*time/(100+time)/(1+e_{(kg-R)*kf})$ instead of $r*ke/(1+e_{(kg-R)*kf})$

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

For more information see the terms of use.

To cite BioModels Database, please use Le Novère N., Bornstein B., Broicher A., Courtot M., Donizelli M., Dharuri H., Li L., Sauro H., Schilstra M., Shapiro B., Snoep J.L., Hucka M. (2006) BioModels Database: A Free, Centralized Database of Curated, Published, Quantitative Kinetic Models of Biochemical and Cellular Systems Nucleic Acids Res., 34: D689-D691.

Parameters:

NameDescription
kminus1=1.0 persecReaction: r5 =>, Rate Law: endosome*kminus1*r5
K1=0.483 MpersReaction: => r7, Rate Law: endosome*K1
kh=0.06 persecReaction: R5 => r5, Rate Law: endosome*kh*R5
kg=1.0 M; ke=0.021 persec; kf=3.0 lpermoleReaction: r7 => R7; R5, Rate Law: endosome*ke*r7/(1+exp((kg-R5)*kf))
kf=2.5 lpermole; ke=0.3 persec; kg=0.1 MReaction: r5 => R5; R5, Rate Law: endosome*r5*ke*time/(100+time)/(1+exp((kg-R5)*kf))
ke=0.21 persec; kg=0.1; h=3.0 dimensionlessReaction: r7 => R7; R7, Rate Law: endosome*r7*ke*R7^h/(kg+R7^h)
kg=0.3 M; ke=0.31 persec; kf=3.0 lpermoleReaction: R5 => r5; R7, Rate Law: endosome*ke*R5/(1+exp((kg-R7)*kf))
kh=0.15 persecReaction: R7 => r7, Rate Law: endosome*kh*R7
kminus1=0.483 persecReaction: r7 =>, Rate Law: endosome*kminus1*r7
K1=1.0 MpersReaction: => r5, Rate Law: endosome*K1

States:

NameDescription
r7[GDP; Ras-related protein Rab-7a; anchored component of membrane]
r5[GDP; Ras-related protein Rab-5A; anchored component of membrane]
R5[GTP; Ras-related protein Rab-5A; anchored component of membrane]
R7[GTP; Ras-related protein Rab-7a; anchored component of membrane]

Delattre2020 - SARS-CoV-2-infected human lung cell: MODEL2010280002v0.0.1

Modified Recon2.2 where the host biomass objective function has been modified to reflect the average composition of the…

Details

Viruses rely on their host for reproduction. Here, we made use of genomic and structural information to create a biomass function capturing the amino and nucleic acid requirements of SARSCoV- 2. Incorporating this biomass function into a stoichiometric metabolic model of the human lung cell and applying metabolic flux balance analysis, we identified host-based metabolic perturbations inhibiting SARS-CoV-2 reproduction. Our results highlight reactions in the central metabolism, as well as amino acid and nucleotide biosynthesis pathways. By incorporating host cellular maintenance into the model based on available protein expression data from human lung cells, we find thaViruses rely on their host for reproduction. Here, we made use of genomic and structural information to create a biomass function capturing the amino and nucleic acid requirements of SARSCoV- 2. Incorporating this biomass function into a stoichiometric metabolic model of the human lung cell and applying metabolic flux balance analysis, we identified host-based metabolic perturbations inhibiting SARS-CoV-2 reproduction. Our results highlight reactions in the central metabolism, as well as amino acid and nucleotide biosynthesis pathways. By incorporating host cellular maintenance into the model based on available protein expression data from human lung cells, we find that only few of these metabolic perturbations are able to selectively inhibit virus reproduction. Some of the catalysing enzymes of such reactions have demonstrated interactions with existing drugs, which can be used for experimental testing of the presented predictions using gene knockouts and RNA interference techniques. In summary, the developed computational approach offers a platform for rapid, experimentally testable generation of drug predictions against existing and emerging viruses based on their biomass requirements.t only few of these metabolic perturbations are able to selectively inhibit virus reproduction. Some of the catalysing enzymes of such reactions have demonstrated interactions with existing drugs, which can be used for experimental testing of the presented predictions using gene knockouts and RNA interference techniques. In summary, the developed computational approach offers a platform for rapid, experimentally testable generation of drug predictions against existing and emerging viruses based on their biomass requirements. link: http://identifiers.org/doi/10.26508/LSA.202000869

DellOrco2009_phototransduction: BIOMD0000000326v0.0.1

This a model from the article: Network-level analysis of light adaptation in rod cells under normal and altered cond…

Details

Photoreceptor cells finely adjust their sensitivity and electrical response according to changes in light stimuli as a direct consequence of the feedback and regulation mechanisms in the phototransduction cascade. In this study, we employed a systems biology approach to develop a dynamic model of vertebrate rod phototransduction that accounts for the details of the underlying biochemistry. Following a bottom-up strategy, we first reproduced the results of a robust model developed by Hamer et al. (Vis. Neurosci., 2005, 22(4), 417), and then added a number of additional cascade reactions including: (a) explicit reactions to simulate the interaction between the activated effector and the regulator of G-protein signalling (RGS); (b) a reaction for the reformation of the G-protein from separate subunits; (c) a reaction for rhodopsin (R) reconstitution from the association of the opsin apoprotein with the 11-cis-retinal chromophore; (d) reactions for the slow activation of the cascade by opsin. The extended network structure successfully reproduced a number of experimental conditions that were inaccessible to prior models. With a single set of parameters the model was able to predict qualitative and quantitative features of rod photoresponses to light stimuli ranging over five orders of magnitude, in normal and altered conditions, including genetic manipulations of the cascade components. In particular, the model reproduced the salient dynamic features of the rod from Rpe65(-/-) animals, a well established model for Leber congenital amaurosis and vitamin A deficiency. The results of this study suggest that a systems-level approach can help to unravel the adaptation mechanisms in normal and in disease-associated conditions on a molecular basis. link: http://identifiers.org/pubmed/19756313

Parameters:

NameDescription
kGshutoff = 0.05Reaction: Ga_GTP => Ga_GDP, Rate Law: kGshutoff*Ga_GTP
kGrecyc = 2.0Reaction: Ga_GDP + Gbg => Gt, Rate Law: kGrecyc*Gbg*Ga_GDP
kRec3 = 9.68777; Rec_wCa2 = 0.0; kRec4 = 0.610084Reaction: RK => Rec_wCa2_RK; Ca2_free, Rate Law: kRec3*Rec_wCa2*RK-kRec4*Rec_wCa2_RK
kP1 = 0.0549715; kP1_rev = 0.0Reaction: Ga_GTP + PDE => PDE_Ga_GTP, Rate Law: kP1*PDE*Ga_GTP-kP1_rev*PDE_Ga_GTP
kG7 = 200.0Reaction: G_GTP => Ga_GTP + Gbg, Rate Law: kG7*G_GTP
kRK2 = 250.0; kRK1_5 = 0.0Reaction: R5 + RK => R5_RKpre, Rate Law: kRK1_5*RK*R5-kRK2*R5_RKpre
kP3 = 1.49834E-9Reaction: Ga_GTP + PDE_a_Ga_GTP => Ga_GTP_PDE_a_Ga_GTP, Rate Law: kP3*PDE_a_Ga_GTP*Ga_GTP
kRGS1 = 1.57E-7Reaction: Ga_GTP_a_PDE_a_Ga_GTP + RGS => RGS_Ga_GTP_a_PDE_a_Ga_GTP, Rate Law: kRGS1*RGS*Ga_GTP_a_PDE_a_Ga_GTP
kRGS2 = 256.07Reaction: RGS_Ga_GTP_a_PDE_a_Ga_GTP => Ga_GDP + PDE_a_Ga_GTP + RGS, Rate Law: kRGS2*RGS_Ga_GTP_a_PDE_a_Ga_GTP
kA2 = 0.00323198; kA1_2 = 0.0Reaction: Arr + R2 => R2_Arr, Rate Law: kA1_2*Arr*R2-kA2*R2_Arr
kP4 = 21.0881Reaction: Ga_GTP_PDE_a_Ga_GTP => Ga_GTP_a_PDE_a_Ga_GTP, Rate Law: kP4*Ga_GTP_PDE_a_Ga_GTP
kRK2 = 250.0; kRK1_0 = 0.0076429599557114Reaction: R0 + RK => R0_RKpre, Rate Law: kRK1_0*RK*R0-kRK2*R0_RKpre
kRK3_ATP = 400.0Reaction: R0_RKpre => R1_RKpost, Rate Law: kRK3_ATP*R0_RKpre
kRK4 = 20.0Reaction: R2_RKpost => R2 + RK, Rate Law: kRK4*R2_RKpost
kA3 = 0.0445091Reaction: R1_Arr => Arr + Ops, Rate Law: kA3*R1_Arr
kG1_5 = 0.0; kG2 = 2250.34Reaction: Gt + R5 => R5_Gt, Rate Law: kG1_5*Gt*R5-kG2*R5_Gt
kG6 = 2000.0Reaction: Ops_G_GTP => G_GTP + Ops, Rate Law: kG6*Ops_G_GTP
kOps = 6.1172E-13; kG2 = 2250.34Reaction: Gt + Ops => Ops_Gt, Rate Law: kOps*Ops*Gt-kG2*Ops_Gt
kRK2 = 250.0; kRK1_4 = 0.0Reaction: R4 + RK => R4_RKpre, Rate Law: kRK1_4*RK*R4-kRK2*R4_RKpre
k2 = 1.9094; k1 = 0.381529; eT = 400.0Reaction: Ca2_free => Ca2_buff, Rate Law: k1*(eT-Ca2_buff)*Ca2_free-k2*Ca2_buff
kG2 = 2250.34; kG1_4 = 0.0Reaction: Gt + R4 => R4_Gt, Rate Law: kG1_4*Gt*R4-kG2*R4_Gt
kRK1_3 = 0.0; kRK2 = 250.0Reaction: R3 + RK => R3_RKpre, Rate Law: kRK1_3*RK*R3-kRK2*R3_RKpre
kG2 = 2250.34; kG1_2 = 0.0Reaction: Gt + R2 => R2_Gt, Rate Law: kG1_2*Gt*R2-kG2*R2_Gt
kG2 = 2250.34; kG1_6 = 0.0Reaction: Gt + R6 => R6_Gt, Rate Law: kG1_6*Gt*R6-kG2*R6_Gt
kG4_GDP = 600.0; kG3 = 2000.0Reaction: Ops_Gt => Ops_G, Rate Law: kG3*Ops_Gt-kG4_GDP*Ops_G
kG5_GTP = 750.0Reaction: Ops_G => Ops_G_GTP, Rate Law: kG5_GTP*Ops_G
betasub = 4.3E-4; E = 0.0; betadark = 1.2Reaction: cGMP => ; Ga_GTP_a_PDE_a_Ga_GTP, PDE_a_Ga_GTP, Rate Law: (betadark+betasub*E)*cGMP
ktherm = 0.0238Reaction: R0 => Ops, Rate Law: ktherm*R0
kRK2 = 250.0; kRK1_1 = 0.0Reaction: R1 + RK => R1_RKpre, Rate Law: kRK1_1*RK*R1-kRK2*R1_RKpre
kG2 = 2250.34; kG1_3 = 0.0Reaction: Gt + R3 => R3_Gt, Rate Law: kG1_3*Gt*R3-kG2*R3_Gt
Kc = 0.17; alfamax = 0.0; m = 2.5Reaction: => cGMP; Ca2_free, Rate Law: alfamax/(1+(Ca2_free/Kc)^m)
kRK2 = 250.0; kRK1_6 = 0.0Reaction: R6 + RK => R6_RKpre, Rate Law: kRK1_6*RK*R6-kRK2*R6_RKpre
kPDEshutoff = 0.033Reaction: Ga_GTP_a_PDE_a_Ga_GTP => Ga_GDP + PDE_a_Ga_GTP, Rate Law: kPDEshutoff*Ga_GTP_a_PDE_a_Ga_GTP
gammaCa = 47.554; Ca2_0 = 0.01Reaction: Ca2_free =>, Rate Law: gammaCa*(Ca2_free-Ca2_0)
kRK2 = 250.0; kRK1_2 = 0.0Reaction: R2 + RK => R2_RKpre, Rate Law: kRK1_2*RK*R2-kRK2*R2_RKpre
fCa = 0.2; Jdark = 29.7778; Vcyto = 1.0; cGMPdark = 4.0; ncg = 3.0; F = 96485.3415Reaction: => Ca2_free; cGMP, Rate Law: 1E6*fCa*Jdark/((2+fCa)*F*Vcyto)*(cGMP/cGMPdark)^ncg
kG1_0 = 3.0586111111E-5; kG2 = 2250.34Reaction: Gt + R0 => R0_Gt, Rate Law: kG1_0*Gt*R0-kG2*R0_Gt
kA1_6 = 0.0; kA2 = 0.00323198Reaction: Arr + R6 => R6_Arr, Rate Law: kA1_6*Arr*R6-kA2*R6_Arr
kG1_1 = 0.0; kG2 = 2250.34Reaction: Gt + R1 => R1_Gt, Rate Law: kG1_1*Gt*R1-kG2*R1_Gt
kA1_1 = 0.0; kA2 = 0.00323198Reaction: Arr + R1 => R1_Arr, Rate Law: kA1_1*Arr*R1-kA2*R1_Arr

States:

NameDescription
RGS Ga GTP a PDE a Ga GTP[GTP; Retinal rod rhodopsin-sensitive cGMP 3',5'-cyclic phosphodiesterase subunit gamma; Guanine nucleotide-binding protein subunit alpha-12; Regulator of G-protein signaling 9]
Ga GDP[GDP; Guanine nucleotide-binding protein subunit alpha-12]
G GTP[GTP; G-protein coupled receptor 183]
R0 Gt[Rhodopsin; Transducin beta-like protein 2]
RGS[Regulator of G-protein signaling 9]
R0 RKpre[Rhodopsin kinase; Rhodopsin]
Ga GTP a PDE a Ga GTP[GTP; Guanine nucleotide-binding protein subunit alpha-12; Retinal rod rhodopsin-sensitive cGMP 3',5'-cyclic phosphodiesterase subunit gamma]
cGMP[3',5'-cyclic GMP]
Ca2 buff[calcium(2+); Calcium cation]
Ops G[G-protein coupled receptor 183; Medium-wave-sensitive opsin 1]
R6 G[G-protein coupled receptor 183; Phosphorhodopsin]
Gt[Transducin beta-like protein 2]
R6 Arr[S-arrestin; Phosphorhodopsin]
Ops G GTP[GTP; Medium-wave-sensitive opsin 1; G-protein coupled receptor 183]
R6 Gt[Transducin beta-like protein 2; Phosphorhodopsin]
R6 RKpost[Rhodopsin kinase; Phosphorhodopsin]
R0 G GTP[GTP; Rhodopsin; G-protein coupled receptor 183]
Ops[Medium-wave-sensitive opsin 1]
R0 G[Rhodopsin; G-protein coupled receptor 183]
RK[Rhodopsin kinase]
R1[Phosphorhodopsin]
R6 G GTP[GTP; G-protein coupled receptor 183; Phosphorhodopsin]
Ga GTP[GTP; Guanine nucleotide-binding protein subunit alpha-12]
R6 RKpre[Rhodopsin kinase; Phosphorhodopsin]
Ga GTP PDE a Ga GTP[GTP; Retinal rod rhodopsin-sensitive cGMP 3',5'-cyclic phosphodiesterase subunit gamma; Guanine nucleotide-binding protein subunit alpha-12]
Arr[S-arrestin]
RGS PDE a Ga GTP[GTP; Regulator of G-protein signaling 9; Guanine nucleotide-binding protein subunit alpha-12; Retinal rod rhodopsin-sensitive cGMP 3',5'-cyclic phosphodiesterase subunit gamma]
Gbg[G-protein beta/gamma-subunit complex]
Rec wCa2 RK[calcium(2+); Recoverin; Rhodopsin kinase]
Ca2 free[calcium(2+); Calcium cation]

Demin2013 - PKPD behaviour - 5-Lipoxygenase inhibitors: BIOMD0000000490v0.0.1

Demin2013 - PKPD behaviour - 5-Lipoxygenase inhibitorsThis model is described in the article: [Systems pharmacology mod…

Details

Zileuton, a 5-lipoxygenase (5LO) inhibitor, displays complex pharmaokinetic (PK)-pharmacodynamic (PD) behavior. Available clinical data indicate a lack of dose-bronchodilatory response during initial treatment, with a dose response developing after ~1-2 weeks. We developed a quantitative systems pharmacology (QSP) model to understand the mechanism behind this phenomenon. The model described the release, maturation, and trafficking of eosinophils into the airways, leukotriene synthesis by the 5LO enzyme, leukotriene signaling and bronchodilation, and the PK of zileuton. The model provided a plausible explanation for the two-phase bronchodilatory effect of zileuton-the short-term bronchodilation was due to leukotriene inhibition and the long-term bronchodilation was due to inflammatory cell infiltration blockade. The model also indicated that the theoretical maximum bronchodilation of both 5LO inhibition and leukotriene receptor blockade is likely similar. QSP modeling provided interesting insights into the effects of leukotriene modulation.CPT: Pharmacometrics & Systems Pharmacology (2013) 2, e74; doi:10.1038/psp.2013.49; advance online publication 11 September 2013. link: http://identifiers.org/pubmed/24026253

Parameters:

NameDescription
FLO3_b = 0.0; r1 = 0.0; FLO2_b = 0.0; Ke_ox = 99.99979Reaction: HPETE_b => HETE_b; HPETE_b, HETE_b, Rate Law: Default*r1*(HPETE_b*FLO2_b-HETE_b*FLO3_b/Ke_ox)
k_Hn_p = 1.8E10Reaction: => Hn_aw; EO_a_aw, EO_i_aw, EO_aw, EO_a_aw, EO_i_aw, EO_aw, Rate Law: V_AW*k_Hn_p*(EO_a_aw+EO_i_aw+EO_aw)
k_lo = 4642.68; FLO3t_aw = 0.0; K_AA = 10.74959Reaction: AA_aw => ; AA_aw, Rate Law: Default*k_lo*AA_aw*FLO3t_aw/K_AA
FLO5HP_aw = 0.0; Ki_AA = 551.8748; k_3 = 263640.0; FLO3t_aw = 0.0; k3 = 34.0Reaction: => HPETE_aw; AA_aw, HPETE_aw, AA_aw, Rate Law: Default*(k_3*FLO5HP_aw-k3*FLO3t_aw*HPETE_aw)*(1.0+AA_aw/Ki_AA)
R_Hn_B = 141.0; R_Hn_AW = 5130.0; Kp_Hn_AW = 3950.0; Q_AW_blf = 5.23Reaction: Hn_aw => Hn_b; Hn_aw, Hn_b, Rate Law: Q_AW_blf*R_Hn_B*(Hn_aw*R_Hn_AW/Kp_Hn_AW-Hn_b)
Kp_LTE_AW = 0.22; Q_AW_blf = 5.23; R_LTE_B = 0.538; R_LTE_AW = 1.4Reaction: LTE4_aw => LTE4_b; LTE4_aw, LTE4_b, Rate Law: Q_AW_blf*R_LTE_B*(LTE4_aw*R_LTE_AW/Kp_LTE_AW-LTE4_b)
ca = 10.0; kia = 0.001Reaction: EO_a_aw => EO_aw; EO_a_aw, Rate Law: ca*V_AW*kia*EO_a_aw
k_lo = 4642.68; FLO3t_b = 0.0; K_AA = 10.74959Reaction: AA_b => ; AA_b, Rate Law: Default*k_lo*AA_b*FLO3t_b/K_AA
fup_Hn = 0.77; k_Hn_d = 0.033Reaction: Hn_b => ; Hn_b, Rate Law: Vd_Hn*k_Hn_d*fup_Hn*Hn_b
EC50_migr = 0.115; h_migr = 3.0; k_EO_t_baw = 0.04; Rec_occup_migr = 0.0Reaction: EO_b => EO_aw; EO_b, Rate Law: V_B*k_EO_t_baw*EO_b*Rec_occup_migr^h_migr/(EC50_migr^h_migr+Rec_occup_migr^h_migr)
R_LTD_AW = 1.4; Q_AW_blf = 5.23; R_LTD_B = 0.538; Kp_LTD_AW = 0.22Reaction: LTD4_aw => LTD4_b; LTD4_aw, LTD4_b, Rate Law: Q_AW_blf*R_LTD_B*(LTD4_aw*R_LTD_AW/Kp_LTD_AW-LTD4_b)
B_aw = 0.0; A_aw = 0.0; GPx = 1.6Reaction: HPETE_aw => HETE_aw, Rate Law: Default*GPx*B_aw/A_aw
R_ZF_B = 0.533; Q_AW_blf = 5.23; R_ZF_AW = 2.96; Kp_ZF_AW = 0.204Reaction: ZF_blood => ZF_airways; ZF_blood, ZF_airways, Rate Law: Q_AW_blf*R_ZF_B*(ZF_blood-ZF_airways*R_ZF_AW/Kp_ZF_AW)
k1_min = 1.6E-7; h_matur = 1.0; k1 = 1.0E-6; Km_1 = 2.0Reaction: => EO_bm; IL_bm, IL_bm, Rate Law: V_BM*(k1*IL_bm^h_matur/(Km_1^h_matur+IL_bm^h_matur)+k1_min)
k_lte_el = 0.04; k_acet = 0.002703885Reaction: LTE4_aw => ; LTE4_aw, Rate Law: Vd_AW_LTE*(k_lte_el+k_acet)*LTE4_aw
k_IL_t_bbm = 0.001; J_BM_lymfl = 4.9E-4Reaction: IL_b => IL_bm; IL_b, IL_bm, Rate Law: k_IL_t_bbm*(IL_b-IL_bm)-J_BM_lymfl*IL_bm
Kd50 = 0.43; V_LTC_CB = 0.0Reaction: => LTC4_b_out; LTC4_b, LTC4_b, Rate Law: Default*Kd50*LTC4_b*V_LTC_CB*1E1^6.0
A_hedh_aw = 0.0; HEDH5 = 0.5; B_hedh_aw = 0.0Reaction: HETE_aw =>, Rate Law: Default*HEDH5*B_hedh_aw/A_hedh_aw
den_LTCs_b = 0.0; nom_LTCs_b = 0.0Reaction: LTA4_b => LTC4_b, Rate Law: Default*nom_LTCs_b/den_LTCs_b
Kd12 = 0.007Reaction: LTA4_aw => ; LTA4_aw, Rate Law: Default*Kd12*LTA4_aw
k_EO_m = 10.0; ca = 10.0Reaction: EO_i_aw => EO_a_aw; EO_i_aw, Rate Law: ca*V_AW*k_EO_m*EO_i_aw
k_EO_a_d = 1.5E-4Reaction: EO_a_aw => ; EO_a_aw, Rate Law: V_AW*k_EO_a_d*EO_a_aw
ka = 500.0; ca = 10.0; h_act = 3.0; EC50_act = 0.75; OL_b = 0.0Reaction: EO_b => EO_i_b; EO_b, Rate Law: ca*V_B*ka*EO_b*OL_b^h_act/(EC50_act^h_act+OL_b^h_act)
fup_LT = 0.16; k_lte_el = 0.04; k_acet = 0.002703885Reaction: LTE4_b => ; LTE4_b, Rate Law: Vd_LTE*(k_lte_el+k_acet)*fup_LT*LTE4_b
k_IL_d = 0.0046Reaction: IL_aw => ; IL_aw, Rate Law: V_AW*k_IL_d*IL_aw
k_lta_syn = 54420.0; FLO5HP_aw = 0.0Reaction: => LTA4_aw, Rate Law: Default*k_lta_syn*FLO5HP_aw
k_ggt = 0.1Reaction: LTC4_aw_out => LTD4_aw; LTC4_aw_out, Rate Law: Vd_AW_LTC*k_ggt*LTC4_aw_out
Q_AW_blf = 5.23; R_LTC_B = 0.538; R_LTC_AW = 1.4; Kp_LTC_AW = 0.22Reaction: LTC4_aw_out => LTC4_b_out; LTC4_aw_out, LTC4_b_out, Rate Law: Q_AW_blf*R_LTC_B*(LTC4_aw_out*R_LTC_AW/Kp_LTC_AW-LTC4_b_out)
k_IL_p = 16.0Reaction: => IL_aw; EO_a_aw, EO_a_aw, Rate Law: V_AW*k_IL_p*EO_a_aw
Kd50 = 0.43Reaction: LTC4_aw => ; LTC4_aw, Rate Law: Default*Kd50*LTC4_aw
k_EO_d = 3.0E-4Reaction: EO_aw => ; EO_aw, Rate Law: V_AW*k_EO_d*EO_aw
k_ltc_ltd_el = 0.1Reaction: LTC4_aw_out => ; LTC4_aw_out, Rate Law: Vd_AW_LTC*k_ltc_ltd_el*LTC4_aw_out
FLO3_aw = 0.0; r1 = 0.0; FLO2_aw = 0.0; Ke_ox = 99.99979Reaction: HPETE_aw => HETE_aw; HPETE_aw, HETE_aw, Rate Law: Default*r1*(HPETE_aw*FLO2_aw-HETE_aw*FLO3_aw/Ke_ox)
PLA2_Ca = 0.0; PL = 110.0; Km_PLA2_APC = 20.0; Km_CoA_AA = 0.005; V_CoA = 350.0; Vmax_PLA2 = 450.0Reaction: => AA_b; AA_b, Rate Law: Default*(Vmax_PLA2*PLA2_Ca*PL/(Km_PLA2_APC+PL)-V_CoA*AA_b/(Km_CoA_AA+AA_b))
Kd50 = 0.43; V_LTC_CAW = 0.0Reaction: => LTC4_aw_out; LTC4_aw, LTC4_aw, Rate Law: Default*Kd50*LTC4_aw*V_LTC_CAW*1E1^6.0
HEDH5 = 0.5; B_hedh_b = 0.0; A_hedh_b = 0.0Reaction: HETE_b =>, Rate Law: Default*HEDH5*B_hedh_b/A_hedh_b
k_lta_syn = 54420.0; FLO5HP_b = 0.0Reaction: => LTA4_b, Rate Law: Default*k_lta_syn*FLO5HP_b
ka = 500.0; OL_aw = 0.0; ca = 10.0; h_act = 3.0; EC50_act = 0.75Reaction: EO_aw => EO_i_aw; EO_aw, Rate Law: ca*V_AW*ka*EO_aw*OL_aw^h_act/(EC50_act^h_act+OL_aw^h_act)
k_ltc_ltd_el = 0.1; fup_LT = 0.16Reaction: LTD4_b => ; LTD4_b, Rate Law: Vd_LTD*k_ltc_ltd_el*fup_LT*LTD4_b
k_elim_zf = 0.004Reaction: ZF_blood => ; ZF_blood, Rate Law: Vd_ZF*k_elim_zf*ZF_blood
fup_LT = 0.16; k_dp = 0.067Reaction: LTD4_b => LTE4_b; LTD4_b, Rate Law: Vd_LTD*k_dp*fup_LT*LTD4_b
Ki_AA = 551.8748; k_3 = 263640.0; FLO3t_b = 0.0; FLO5HP_b = 0.0; k3 = 34.0Reaction: => HPETE_b; AA_b, HPETE_b, AA_b, Rate Law: Default*(k_3*FLO5HP_b-k3*FLO3t_b*HPETE_b)*(1.0+AA_b/Ki_AA)
k_Hn_d = 0.033Reaction: Hn_aw => ; Hn_aw, Rate Law: Vd_AW_Hn*k_Hn_d*Hn_aw
ft_zf = 0.0; F_zf = 0.082; a = 1.0; oral = 1.0; M_ZF = 236.0; DOSE_zf = 0.0; k_abs_zf = 0.018Reaction: ZF_intes => ZF_blood; ZF_intes, Rate Law: Default*k_abs_zf*(ZF_intes+oral*F_zf*(a*ft_zf+(1.0-a))*DOSE_zf*1E3/M_ZF)
B_b = 0.0; A_b = 0.0; GPx = 1.6Reaction: HPETE_b => HETE_b, Rate Law: Default*GPx*B_b/A_b
J_AW_lymfl = 0.00115; k_IL_t_awb = 0.05Reaction: IL_aw => IL_b; IL_aw, IL_b, Rate Law: k_IL_t_awb*(IL_aw-IL_b)+J_AW_lymfl*IL_aw
k_dp = 0.067Reaction: LTD4_aw => LTE4_aw; LTD4_aw, Rate Law: Vd_AW_LTD*k_dp*LTD4_aw
k_EO_t_bmb = 0.02; ca = 10.0Reaction: EO_bm => EO_b; EO_bm, Rate Law: ca*V_BM*k_EO_t_bmb*EO_bm
den_LTCs_aw = 0.0; nom_LTCs_aw = 0.0Reaction: LTA4_aw => LTC4_aw, Rate Law: Default*nom_LTCs_aw/den_LTCs_aw
k_elim_ml = 0.00225Reaction: ML_blood => ; ML_blood, Rate Law: Vd_ML*k_elim_ml*ML_blood
fup_LT = 0.16; k_ggt = 0.1Reaction: LTC4_b_out => LTD4_b; LTC4_b_out, Rate Law: Vd_LTC*k_ggt*fup_LT*LTC4_b_out
a = 1.0; oral = 1.0; F_ml = 0.660688; k_abs_ml = 0.012; M_ML = 586.18; ft_ml = 0.0; DOSE_ml = 0.0Reaction: ML_intes => ML_blood; ML_intes, Rate Law: Default*k_abs_ml*(ML_intes+oral*F_ml*(a*ft_ml+(1.0-a))*DOSE_ml*1E9/M_ML)

States:

NameDescription
LTA4 b[5280383; blood plasma]
HETE aw[5280733; respiratory smooth muscle]
EO a aw[respiratory smooth muscle; eosinophil]
ML intes[montelukast; intestine]
IL b[Interleukin-5; blood plasma]
LTD4 aw[respiratory smooth muscle; 6435286]
EO b[blood plasma; eosinophil]
ZF airways[zileuton; respiratory smooth muscle]
LTE4 b[5280879]
EO i aw[respiratory smooth muscle; eosinophil]
LTC4 aw[5280493; respiratory smooth muscle]
AA aw[arachidonic acid; respiratory smooth muscle]
ZF intes[zileuton; intestine]
HETE b[5280733; blood plasma]
ML blood[montelukast; blood plasma]
EO bm[eosinophil; bone marrow]
HPETE aw[5280778; respiratory smooth muscle]
EO i b[blood plasma; eosinophil]
LTE4 aw[5280879; respiratory smooth muscle]
IL aw[Interleukin-5; respiratory smooth muscle]
AA b[arachidonic acid; blood plasma]
LTC4 b[5280493; blood plasma]
LTC4 b out[5280493]
HPETE b[5280778; blood plasma]
EO a b[blood plasma; eosinophil]
Hn aw[respiratory smooth muscle; histamine]
EO aw[respiratory smooth muscle; eosinophil]
LTA4 aw[5280383; respiratory smooth muscle]
Hn b[histamine; blood plasma]
LTC4 aw out[5280493]
LTD4 b[6435286]
ZF blood[zileuton; respiratory smooth muscle]
IL bm[Interleukin-5; bone marrow]

Demir1994_SinoatrialNode: MODEL0912887467v0.0.1

This a model from the article: A mathematical model of a rabbit sinoatrial node cell. Demir SS, Clark JW, Murphey CR…

Details

A mathematical model for the electrophysiological responses of a rabbit sinoatrial node cell that is based on whole cell recordings from enzymatically isolated single pacemaker cells at 37 degrees C has been developed. The ion channels, Na(+)-K+ and Ca2+ pumps, and Na(+)-Ca2+ exchanger in the surface membrane (sarcolemma) are described using equations for these known currents in mammalian pacemaker cells. The extracellular environment is treated as a diffusion-limited space, and the myoplasm contains Ca(2+)-binding proteins (calmodulin and troponin). Original features of this model include 1) new equations for the hyperpolarization-activated inward current, 2) assessment of the role of the transient-type Ca2+ current during pacemaker depolarization, 3) inclusion of an Na+ current based on recent experimental data, and 4) demonstration of the possible influence of pump and exchanger currents and background currents on the pacemaker rate. This model provides acceptable fits to voltage-clamp and action potential data and can be used to seek biophysically based explanations of the electrophysiological activity in the rabbit sinoatrial node cell. link: http://identifiers.org/pubmed/8166247

Demir1999_SinoatrialNodeActivity_Heart: MODEL0912940495v0.0.1

This a model from the article: Parasympathetic modulation of sinoatrial node pacemaker activity in rabbit heart: a uni…

Details

We have extended our compartmental model [Am. J. Physiol. 266 (Cell Physiol. 35): C832-C852, 1994] of the single rabbit sinoatrial node (SAN) cell so that it can simulate cellular responses to bath applications of ACh and isoprenaline as well as the effects of neuronally released ACh. The model employs three different types of muscarinic receptors to explain the variety of responses observed in mammalian cardiac pacemaking cells subjected to vagal stimulation. The response of greatest interest is the ACh-sensitive change in cycle length that is not accompanied by a change in action potential duration or repolarization or hyperpolarization of the maximum diastolic potential. In this case, an ACh-sensitive K+ current is not involved. Membrane hyperpolarization occurs in response to much higher levels of vagal stimulation, and this response is also mimicked by the model. Here, an ACh-sensitive K+ current is involved. The well-known phase-resetting response of the SAN cell to single and periodically applied vagal bursts of impulses is also simulated in the presence and absence of the beta-agonist isoprenaline. Finally, the responses of the SAN cell to longer continuous trains of periodic vagal stimulation are simulated, and this can result in the complete cessation of pacemaking. Therefore, this model is 1) applicable over the full range of intensity and pattern of vagal input and 2) can offer biophysically based explanations for many of the phenomena associated with the autonomic control of cardiac pacemaking. link: http://identifiers.org/pubmed/10362707

Dempsher1984_ACTH_CortisolSecretion: MODEL0912835813v0.0.1

This a model from the article: A mechanistic model of ACTH-stimulated cortisol secretion. Dempsher DP, Gann DS, Phai…

Details

Adrenal secretory rates of cortisol and arterial concentrations of adrenocorticotropin (ACTH) were measured in conscious trained dogs subjected to intravenous infusion of ACTH. To investigate the causal relation of ACTH to the secretion of cortisol, a mechanistic mathematical model based on current hypotheses of adrenocortical function was constructed and tested. It is widely believed that ACTH stimulates cortisol secretion through adenosine 3',5'-cyclic monophosphate (cAMP), which provides substrate cholesterol by activating cholesterol ester hydrolase and facilitating transport of cholesterol to the side-chain cleavage enzyme. In addition, cholesterol modulates its own synthesis by inhibiting beta-hydroxy-beta-methylglutaryl (HMG)-CoA reductase in the adrenocortical cell. These and other steps in the biosynthetic reaction sequence were described using differential equations subject to the additional constraints imposed by available measurements of intracellular quantities. The resulting model is consistent with many of the known characteristics of the canine adrenal response to ACTH. In this model, steady-state nonlinearities arise from cooperative binding of cAMP to its receptor protein and saturation of mitochondrial pregnenolone transport. The transient response is dominated by a depletable pool of intracellular free cholesterol. Other inferences based on the model are presented, and a quantifiable cellular basis for increased adrenal sensitivity to ACTH is proposed. link: http://identifiers.org/pubmed/6326602

den Breems2015 - macrophage in cancer: BIOMD0000000759v0.0.1

The paper describes a model of re-polarisation of M2 and M1 macrophages and its role on cancer outcomes. Created by COP…

Details

The anti-tumour and pro-tumour roles of Th1/Th2 immune cells and M1/M2 macrophages have been documented by numerous experimental studies. However, it is still unknown how these immune cells interact with each other to control tumour dynamics. Here, we use a mathematical model for the interactions between mouse melanoma cells, Th2/Th1 cells and M2/M1 macrophages, to investigate the unknown role of the re-polarisation between M1 and M2 macrophages on tumour growth. The results show that tumour growth is associated with a type-II immune response described by large numbers of Th2 and M2 cells. Moreover, we show that (i) the ratio k of the transition rates k12 (for the re-polarisation M1→M2) and k21 (for the re-polarisation M2→M1) is important in reducing tumour population, and (ii) the particular values of these transition rates control the delay in tumour growth and the final tumour size. We also perform a sensitivity analysis to investigate the effect of various model parameters on changes in the tumour cell population, and confirm that the ratio k alone and the ratio of M2 and M1 macrophage populations at earlier times (e.g., day 7) cannot always predict the final tumour size. link: http://identifiers.org/pubmed/26551154

Parameters:

NameDescription
rh2 = 9.0E-6 1/d; bth = 1.0E8 1Reaction: => Th2; M2, Th1, Rate Law: tumor_microenvironment*rh2*M2*Th2*(1-(Th2+Th1)/bth)
ah2 = 0.008 1/dReaction: => Th2; M2, Rate Law: tumor_microenvironment*ah2*M2
dh1 = 0.05 1/dReaction: Th1 =>, Rate Law: tumor_microenvironment*dh1*Th1
dm2 = 0.2 1/dReaction: M2 =>, Rate Law: tumor_microenvironment*dm2*M2
ah1 = 0.008 1/dReaction: => Th1; M1, Rate Law: tumor_microenvironment*ah1*M1
as = 1.0E-6 1/d; am1 = 5.0E-8 1/d; bm = 100000.0 1Reaction: => M1; Ts, Th1, M2, Rate Law: tumor_microenvironment*(as*Ts+am1*Th1)*M1*(1-(M1+M2)/bm)
rh1 = 9.0E-6 1/d; bth = 1.0E8 1Reaction: => Th1; M1, Th2, Rate Law: tumor_microenvironment*rh1*M1*Th1*(1-(Th1+Th2)/bth)
k21 = 4.0E-5 1/dReaction: M2 => M1, Rate Law: tumor_microenvironment*k21*M2*M1
dmn = 2.0E-6 1/dReaction: Tn => ; M1, Rate Law: tumor_microenvironment*dmn*M1*Tn
r = 0.565 1/d; bt = 2.0E9 1Reaction: => Tn; Ts, Rate Law: tumor_microenvironment*r*Tn*(1-(Tn+Ts)/bt)
rmn = 1.0E-7 1/dReaction: => Tn; M2, Rate Law: tumor_microenvironment*rmn*Tn*M2
dh2 = 0.05 1/dReaction: Th2 =>, Rate Law: tumor_microenvironment*dh2*Th2
dm1 = 0.2 1/dReaction: M1 =>, Rate Law: tumor_microenvironment*dm1*M1
an = 5.0E-8 1/d; bm = 100000.0 1; am2 = 5.0E-8 1/dReaction: => M2; Tn, Th2, M1, Rate Law: tumor_microenvironment*(an*Tn+am2*Th2)*M2*(1-(M2+M1)/bm)
dms = 2.0E-6 1/dReaction: Ts => ; M1, Rate Law: tumor_microenvironment*dms*M1*Ts
dts = 5.3E-8 1/dReaction: Ts => ; Th1, Rate Law: tumor_microenvironment*dts*Th1*Ts
k12 = 5.0E-5 1/dReaction: M1 => M2, Rate Law: tumor_microenvironment*k12*M1*M2
ksn = 0.1 1/dReaction: Ts => Tn, Rate Law: tumor_microenvironment*ksn*Ts

States:

NameDescription
M2[M2 Macrophage]
M1[M1 Macrophage]
Ts[malignant cell]
Tn[malignant cell]
Th2[T-helper 2 cell]
Th1[T-helper 1 cell]

deOliveiraDalMolin2010 - Genome-scale metabolic network of Arabidopsis thaliana (AraGEM): MODEL1507180028v0.0.1

deOliveiraDalMolin2010 - Genome-scale metabolic network of Arabidopsis thaliana (AraGEM)This model is described in the a…

Details

Genome-scale metabolic network models have been successfully used to describe metabolism in a variety of microbial organisms as well as specific mammalian cell types and organelles. This systems-based framework enables the exploration of global phenotypic effects of gene knockouts, gene insertion, and up-regulation of gene expression. We have developed a genome-scale metabolic network model (AraGEM) covering primary metabolism for a compartmentalized plant cell based on the Arabidopsis (Arabidopsis thaliana) genome. AraGEM is a comprehensive literature-based, genome-scale metabolic reconstruction that accounts for the functions of 1,419 unique open reading frames, 1,748 metabolites, 5,253 gene-enzyme reaction-association entries, and 1,567 unique reactions compartmentalized into the cytoplasm, mitochondrion, plastid, peroxisome, and vacuole. The curation process identified 75 essential reactions with respective enzyme associations not assigned to any particular gene in the Kyoto Encyclopedia of Genes and Genomes or AraCyc. With the addition of these reactions, AraGEM describes a functional primary metabolism of Arabidopsis. The reconstructed network was transformed into an in silico metabolic flux model of plant metabolism and validated through the simulation of plant metabolic functions inferred from the literature. Using efficient resource utilization as the optimality criterion, AraGEM predicted the classical photorespiratory cycle as well as known key differences between redox metabolism in photosynthetic and nonphotosynthetic plant cells. AraGEM is a viable framework for in silico functional analysis and can be used to derive new, nontrivial hypotheses for exploring plant metabolism. link: http://identifiers.org/pubmed/20044452

DePaor1986_CirculatoryAutoregulation: MODEL1172940336v0.0.1

This a model from the article: A feedback oscillator model for circulatory autoregulation Annraoi M. De Paor, Patric…

Details

Circulatory autregulation is the phenomenon whereby an isolated organ can maintain a constant or almost-constant blood flow rate over a range of perfusion pressures. A mathematical model is developed, based on work reported in the physiological literature, and tuned to show that autoregulation can be accomplished by pressure-induced oscillations in arteriolar radius. Various features known lo be exhibited by skeletal muscle and by stretch receptors are incorporated in the model ofsmooth muscle surrounding the arterioles. link: http://identifiers.org/doi/10.1080/00207178608933494

dePillis2003 - The dynamics of an optimally controlled tumor model: A case study: BIOMD0000000909v0.0.1

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

Details

Abstract

We present a phase-space analysis of a mathematical model of tumor growth with an immune response and chemotherapy. We prove that all orbits are bounded and must converge to one of several possible equilibrium points. Therefore, the long-term behavior of an orbit is classified according to the basin of attraction in which it starts. The addition of a drug term to the system can move the solution trajectory into a desirable basin of attraction. We show that the solutions of the model with a time-varying drug term approach the solutions of the system without the drug once traatment has stopped. We present numerical experiments in which optimal control therapy is able to drive the system into a desirable basin of attraction, whereas traditional pulsed chemotherapy is not.

Volume 37, Issue 11, June 2003, Pages 1221-1244 link: http://identifiers.org/doi/10.1016/S0895-7177(03)00133-X

Parameters:

NameDescription
d2 = 1.0Reaction: u =>, Rate Law: compartment*d2*u
v = 0.0Reaction: => u, Rate Law: compartment*v
alpha = 0.3; s = 0.33; p = 0.01Reaction: => I; T, Rate Law: compartment*(s+p*I*T/(alpha+T))
b1 = 1.0; r1 = 1.5Reaction: => T, Rate Law: compartment*r1*T*(1-b1*T)
c2 = 0.5; c3 = 1.0; a2 = 0.3Reaction: T => ; I, N, u, Rate Law: compartment*(c2*I*T+c3*T*N+a2*(1-exp(-u))*T)
d1 = 0.2; c1 = 1.0; a1 = 0.2Reaction: I => ; T, u, Rate Law: compartment*(c1*I*T+d1*I+a1*(1-exp(-u))*I)
b2 = 1.0; r2 = 1.0Reaction: => N, Rate Law: compartment*r2*N*(1-b2*N)
a3 = 0.1; c4 = 1.0Reaction: N => ; T, u, Rate Law: compartment*(c4*T*N+a3*(1-exp(-u))*N)

States:

NameDescription
I[C12735]
T[Neoplastic Cell]
NN
u[C2252]

dePillis2005 - A validated mathematical model of cell-mediated immune response to tumor growth: MODEL1907260001v0.0.1

This model describes interactions between a tumour and the immune system, with specific emphasis on the role of natural…

Details

Mathematical models of tumor-immune interactions provide an analytic framework in which to address specific questions about tumor-immune dynamics. We present a new mathematical model that describes tumor-immune interactions, focusing on the role of natural killer (NK) and CD8+ T cells in tumor surveillance, with the goal of understanding the dynamics of immune-mediated tumor rejection. The model describes tumor-immune cell interactions using a system of differential equations. The functions describing tumor-immune growth, response, and interaction rates, as well as associated variables, are developed using a least-squares method combined with a numerical differential equations solver. Parameter estimates and model validations use data from published mouse and human studies. Specifically, CD8+ T-tumor and NK-tumor lysis data from chromium release assays as well as in vivo tumor growth data are used. A variable sensitivity analysis is done on the model. The new functional forms developed show that there is a clear distinction between the dynamics of NK and CD8+ T cells. Simulations of tumor growth using different levels of immune stimulating ligands, effector cells, and tumor challenge are able to reproduce data from the published studies. A sensitivity analysis reveals that the variable to which the model is most sensitive is patient specific, and can be measured with a chromium release assay. The variable sensitivity analysis suggests that the model can predict which patients may positively respond to treatment. Computer simulations highlight the importance of CD8+ T-cell activation in cancer therapy. link: http://identifiers.org/pubmed/16140967

dePillis2007 - Chemotherapy for tumors An analysis of the dynamics and a study of quadratic and linear optimal controls: MODEL2001160001v0.0.1

Abstract We investigate a mathematical model of tumor–immune interactions with chemotherapy, and strategies for optima…

Details

We investigate a mathematical model of tumor-immune interactions with chemotherapy, and strategies for optimally administering treatment. In this paper we analyze the dynamics of this model, characterize the optimal controls related to drug therapy, and discuss numerical results of the optimal strategies. The form of the model allows us to test and compare various optimal control strategies, including a quadratic control, a linear control, and a state-constraint. We establish the existence of the optimal control, and solve for the control in both the quadratic and linear case. In the linear control case, we show that we cannot rule out the possibility of a singular control. An interesting aspect of this paper is that we provide a graphical representation of regions on which the singular control is optimal. link: http://identifiers.org/pubmed/17306310

Parameters:

NameDescription
a = 0.002; b = 1.02E-9Reaction: => T, Rate Law: compartment*a*T*(1-b*T)
K_L = 0.6; q = 3.42E-10; u = 3.0; m = 0.02Reaction: L => ; T, M, Rate Law: compartment*(m*L+q*L*T+u*L*L+K_L*M*L)
gamma = 0.9Reaction: M =>, Rate Law: compartment*gamma*M
alpha_1 = 13000.0; h = 600.0; g = 0.025; eta = 1.0Reaction: => N; T, Rate Law: compartment*(alpha_1+g*T^eta/(h+T^eta)*N)
mu_I = 10.0Reaction: I =>, Rate Law: compartment*mu_I*I
p = 1.0E-7; f = 0.0412; K_N = 0.6Reaction: N => ; T, M, Rate Law: compartment*(f*N+p*N*T+K_N*M*N)
V_M=0.0Reaction: => M, Rate Law: compartment*V_M
alpha_2 = 5.0E8Reaction: => C, Rate Law: compartment*alpha_2
beta = 0.012; K_C = 0.6Reaction: C => ; M, Rate Law: compartment*(beta*C+K_C*M*C)
r2 = 3.0E-11; p_I = 0.125; V_L=0.0; g_I = 2.0E7Reaction: => L; C, T, I, Rate Law: compartment*(r2*C*T+p_I*L*I/(g_I+I)+V_L)
V_I=0.0; w = 2.0E-4; g_T = 100000.0; p_T = 0.6Reaction: => I; T, L, Rate Law: compartment*(p_T*T/(g_T+T)*L+w*L*I+V_I)
D = 6.6666657777779E-7; K_T = 0.8; c1 = 3.23E-7Reaction: T => ; N, M, Rate Law: compartment*(c1*N*T+D*T+K_T*M*T)

States:

NameDescription
I[Interleukin-2]
T[neoplasm]
M[Combination Chemotherapy]
N[Immune Cell]
C[C120462]
L[cytotoxic T-lymphocyte]

dePillis2007 - Seeking Bang Bang Solutions of Mixed ImmunoChemotherapy of Tumors: MODEL2003060001v0.0.1

SEEKING BANG-BANG SOLUTIONS OF MIXED IMMUNO-CHEMOTHERAPY OF TUMORS LISETTE G. DE PILLIS, K. RENEE FISTER, WEIQING GU, CR…

Details

It is known that a beneficial cancer treatment approach for a single patient often involves the administration of more than one type of therapy. The question of how best to combine multiple cancer therapies, however, is still open. In this study, we investigate the theoretical interaction of three treatment types (two biological therapies and one chemotherapy) with a growing cancer, and present an analysis of an optimal control strategy for administering all three therapies in combination. In the situations with controls introduced linearly, we find that there are conditions on which the controls exist singularly. Although bang-bang controls (on-off) reflect the drug treatment approach that is often implemented clinically, we have demonstrated, in the context of our mathematical model, that there can exist regions on which this may not be the best strategy for minimizing a tumor burden. We characterize the controls in singular regions by taking time derivatives of the switching functions. We will examine these representations and the conditions necessary for the controls to be minimizing in the singular region. We begin by assuming only one of the controls is singular on a given interval. Then we analyze the conditions on which a pair and then all three controls are singular. link: https://scholarship.claremont.edu/hmcfacpub/439/

dePillis2008 - Optimal control of mixed immunotherapy and chemotherapy of tumors: BIOMD0000000913v0.0.1

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

Details

We investigate a mathematical population model of tumor-immune interactions. Thepopulations involved are tumor cells, specific and non-specific immune cells, and con-centrations of therapeutic treatments. We establish the existence of an optimal con-trol for this model and provide necessary conditions for the optimal control triple forsimultaneous application of chemotherapy, tumor infiltrating lymphocyte (TIL) ther-apy, and interleukin-2 (IL-2) treatment. We discuss numerical results for the combina-tion of the chemo-immunotherapy regimens. We find that the qualitative nature of ourresults indicates that chemotherapy is the dominant intervention with TIL interactingin a complementary fashion with the chemotherapy. However, within the optimal con-trol context, the interleukin-2 treatment does not become activated for the estimatedparameter ranges. link: http://identifiers.org/doi/10.1142/S0218339008002435

Parameters:

NameDescription
a = 0.002; b = 1.02E-9Reaction: => T, Rate Law: compartment*a*T*(1-b*T)
K_L = 0.6; q = 3.42E-10; u = 3.0; m = 0.02Reaction: L => ; T, M, Rate Law: compartment*(m*L+q*L*T+u*L*L+K_L*M*L)
gamma = 0.9Reaction: M =>, Rate Law: compartment*gamma*M
alpha_1 = 13000.0; h = 600.0; g = 0.025; eta = 1.0Reaction: => N; T, Rate Law: compartment*(alpha_1+g*T^eta/(h+T^eta)*N)
mu_I = 10.0Reaction: I =>, Rate Law: compartment*mu_I*I
p = 1.0E-7; f = 0.0412; K_N = 0.6Reaction: N => ; T, M, Rate Law: compartment*(f*N+p*N*T+K_N*M*N)
V_M=0.0Reaction: => M, Rate Law: compartment*V_M
alpha_2 = 5.0E8Reaction: => C, Rate Law: compartment*alpha_2
beta = 0.012; K_C = 0.6Reaction: C => ; M, Rate Law: compartment*(beta*C+K_C*M*C)
r2 = 3.0E-11; p_I = 0.125; V_L=0.0; g_I = 2.0E7Reaction: => L; C, T, I, Rate Law: compartment*(r2*C*T+p_I*L*I/(g_I+I)+V_L)
V_I=0.0; w = 2.0E-4; g_T = 100000.0; p_T = 0.6Reaction: => I; T, L, Rate Law: compartment*(p_T*T/(g_T+T)*L+w*L*I+V_I)
D = 6.6666657777779E-7; K_T = 0.8; c1 = 3.23E-7Reaction: T => ; N, M, Rate Law: compartment*(c1*N*T+D*T+K_T*M*T)

States:

NameDescription
I[Interleukin-2]
T[neoplasm]
M[Combination Chemotherapy]
N[Immune Cell]
C[C120462]
L[cytotoxic T-lymphocyte]

dePillis2009 - Mathematical model creation for cancer chemo-immunotherapy: BIOMD0000000779v0.0.1

This is an updated version of a previous model that described the dynamics of cancer treatment, with descriptions of tum…

Details

One of the most challenging tasks in constructing a mathematical model of cancer treatment is the calculation of biological parameters from empirical data. This task becomes increasingly difficult if a model involves several cell populations and treatment modalities. A sophisticated model constructed by de Pillis et al., Mixed immunotherapy and chemotherapy of tumours: Modelling, applications and biological interpretations, J. Theor. Biol. 238 (2006), pp. 841-862; involves tumour cells, specific and non-specific immune cells (natural killer (NK) cells, CD8+T cells and other lymphocytes) and employs chemotherapy and two types of immunotherapy (IL-2 supplementation and CD8+T-cell infusion) as treatment modalities. Despite the overall success of the aforementioned model, the problem of illustrating the effects of IL-2 on a growing tumour remains open. In this paper, we update the model of de Pillis et al. and then carefully identify appropriate values for the parameters of the new model according to recent empirical data. We determine new NK and tumour antigen-activated CD8+T-cell count equilibrium values; we complete IL-2 dynamics; and we modify the model in de Pillis et al. to allow for endogenous IL-2 production, IL-2-stimulated NK cell proliferation and IL-2-dependent CD8+T-cell self-regulations. Finally, we show that the potential patient-specific efficacy of immunotherapy may be dependent on experimentally determinable parameters. link: http://identifiers.org/doi/10.1080/17486700802216301

Parameters:

NameDescription
phi = 2.38405E-7Reaction: => I_IL_2; C_Lymphocytes, Rate Law: compartment*phi*C_Lymphocytes
omega = 0.07874; zeta = 2503.6Reaction: => I_IL_2; L_CD8_T_Cells, Rate Law: compartment*omega*L_CD8_T_Cells*I_IL_2/(zeta+I_IL_2)
alphabeta = 2.25E9; beta = 0.0063Reaction: => C_Lymphocytes; C_Lymphocytes, Rate Law: compartment*beta*(alphabeta-C_Lymphocytes)
a = 0.431; b = 1.02E-9Reaction: => T_Tumour_Cells, Rate Law: compartment*a*T_Tumour_Cells*(1-b*T_Tumour_Cells)
j = 0.01245; k = 2.019E7Reaction: => L_CD8_T_Cells; T_Tumour_Cells, Rate Law: compartment*j*T_Tumour_Cells*L_CD8_T_Cells/(k+T_Tumour_Cells)
delta_C = 1.8328; K_C = 0.034Reaction: C_Lymphocytes => ; M_Chemotherapy_Drug, Rate Law: compartment*K_C*(1-exp((-1)*delta_C*M_Chemotherapy_Drug))*C_Lymphocytes
gamma = 0.5199Reaction: M_Chemotherapy_Drug =>, Rate Law: compartment*gamma*M_Chemotherapy_Drug
r_2 = 5.8467E-13Reaction: => L_CD8_T_Cells; C_Lymphocytes, T_Tumour_Cells, Rate Law: compartment*r_2*C_Lymphocytes*T_Tumour_Cells
v_I = 0.0Reaction: => I_IL_2, Rate Law: compartment*v_I
p_I = 2.971; g_I = 2503.6Reaction: => L_CD8_T_Cells; I_IL_2, Rate Law: compartment*p_I*L_CD8_T_Cells*I_IL_2/(g_I+I_IL_2)
g_N = 250360.0; p_N = 0.068Reaction: => N_Natural_Killer_Cells; I_IL_2, Rate Law: compartment*p_N*N_Natural_Killer_Cells*I_IL_2/(g_N+I_IL_2)
r_1 = 2.9077E-11Reaction: => L_CD8_T_Cells; N_Natural_Killer_Cells, T_Tumour_Cells, Rate Law: compartment*r_1*N_Natural_Killer_Cells*T_Tumour_Cells
D = 0.0Reaction: T_Tumour_Cells =>, Rate Law: compartment*D*T_Tumour_Cells
q = 3.422E-10Reaction: L_CD8_T_Cells => ; T_Tumour_Cells, Rate Law: compartment*q*L_CD8_T_Cells*T_Tumour_Cells
K_L = 0.0486; delta_L = 1.8328Reaction: L_CD8_T_Cells => ; M_Chemotherapy_Drug, Rate Law: compartment*K_L*(1-exp((-1)*delta_L*M_Chemotherapy_Drug))*L_CD8_T_Cells
phi = 2.38405E-7; m = 0.009Reaction: L_CD8_T_Cells => ; I_IL_2, Rate Law: compartment*phi*m*L_CD8_T_Cells/(phi+I_IL_2)
v_M = 0.0Reaction: => M_Chemotherapy_Drug, Rate Law: compartment*v_M
mu_I = 11.7427Reaction: I_IL_2 =>, Rate Law: compartment*mu_I*I_IL_2
ef = 0.111; f = 0.0125Reaction: => N_Natural_Killer_Cells; C_Lymphocytes, N_Natural_Killer_Cells, Rate Law: compartment*f*(ef*C_Lymphocytes-N_Natural_Killer_Cells)
c = 2.9077E-13Reaction: T_Tumour_Cells => ; N_Natural_Killer_Cells, Rate Law: compartment*c*N_Natural_Killer_Cells*T_Tumour_Cells
K_T = 0.9; delta_T = 1.8328Reaction: T_Tumour_Cells => ; M_Chemotherapy_Drug, Rate Law: compartment*K_T*(1-exp((-1)*delta_T*M_Chemotherapy_Drug))*T_Tumour_Cells
v_L = 0.0Reaction: => L_CD8_T_Cells, Rate Law: compartment*v_L
K_N = 0.0675; delta_N = 1.8328Reaction: N_Natural_Killer_Cells => ; M_Chemotherapy_Drug, Rate Law: compartment*K_N*(1-exp((-1)*delta_N*M_Chemotherapy_Drug))*N_Natural_Killer_Cells
u = 4.417E-14; kappa = 2503.6Reaction: L_CD8_T_Cells => ; C_Lymphocytes, I_IL_2, Rate Law: compartment*u*L_CD8_T_Cells^2*C_Lymphocytes*I_IL_2/(kappa+I_IL_2)
p = 2.794E-13Reaction: N_Natural_Killer_Cells => ; T_Tumour_Cells, Rate Law: compartment*p*N_Natural_Killer_Cells*T_Tumour_Cells

States:

NameDescription
I IL 2[Interleukin-2]
M Chemotherapy Drug[doxorubicin]
C Lymphocytes[lymphocyte]
N Natural Killer Cells[natural killer cell]
L CD8 T Cells[CD8-Positive T-Lymphocyte]
T Tumour Cells[Neoplastic Cell]

dePillis2013 - Mathematical modeling of regulatory T cell effects on renal cell carcinoma treatment: BIOMD0000000908v0.0.1

Mathematical modeling of regulatory T cell effects on renal cell carcinoma treatment Lisette dePillis 1, , Trevor Caldw…

Details

Abstract. We present a mathematical model to study the effects of the regu-latory T cells (T reg ) on Renal Cell Carcinoma (RCC) treatment with sunitinib.The drug sunitinib inhibits the natural self-regulation of the immune system,allowing the effector components of the immune system to function for longerperiods of time. This mathematical model builds upon our non-linear ODEmodel by de Pillis et al. (2009) [13] to incorporate sunitinib treatment, regula-tory T cell dynamics, and RCC-specific parameters. The model also elucidatesthe roles of certain RCC-specific parameters in determining key differences be-tween in silico patients whose immune profiles allowed them to respond wellto sunitinib treatment, and those whose profiles did not.Simulations from our model are able to produce results that reflect clinicaloutcomes to sunitinib treatment such as: (1) sunitinib treatments followingstandard protocols led to improved tumor control (over no treatment) in about40% of patients; (2) sunitinib treatments at double the standard dose led to agreater response rate in about 15% the patient population; (3) simulations ofpatient response indicated improved responses to sunitinib treatment when thepatient’s immune strength scaling and the immune system strength coefficientsparameters were low, allowing for a slightly stronger natural immune response link: http://identifiers.org/doi/10.3934/dcdsb.2013.18.915

Parameters:

NameDescription
p = 6.682E-14Reaction: N => ; T, Rate Law: compartment*p*N*T
p_N = 0.0668; f = 0.0125; g_N = 250360.0; e_f = 0.1168Reaction: => N; C, I, Rate Law: compartment*(f*(e_f*C-N)+p_N*N*I/(g_N+I))
delta_R = 50.02; H_R = 0.227Reaction: R => ; S, Rate Law: compartment*H_R*(1-exp((-delta_R)*S))*R
j = 0.1245; r_2 = 1.0E-15; g_I = 2503.6; r_1 = 6.682E-12; p_I = 1.111; k = 2.019E7Reaction: => L; N, C, T, I, Rate Law: compartment*((r_1*N+r_2*C)*T+p_I*L*I/(g_I+I)+j*T/(k+T)*L)
zeta = 2503.6; phi = 3.594E-7; w = 0.05314Reaction: => I; C, L, Rate Law: compartment*(phi*C+w*L*I/(zeta+I))
n = 0.277Reaction: S =>, Rate Law: compartment*(-n)*S
mu_I = 11.7427Reaction: I =>, Rate Law: compartment*mu_I*I
alpha_beta = 2.14E9; beta = 0.0063Reaction: => C, Rate Law: compartment*beta*(alpha_beta-C)
delta_T = 1.59E-9; c = 8.68E-10; D = 9.47552761007735E-6Reaction: T => ; R, N, Rate Law: compartment*(c*exp((-delta_T)*R)*N*T+D*T)
g_R = 11.027; w_u = 0.0122; p_R = 0.03598; u = 0.03851Reaction: => R; C, I, Rate Law: compartment*(u*(w_u*C-R)+p_R*R*I/(g_R+I))
kappa = 2503.6; q = 3.422E-10; z = 2.3085E-13; m = 0.009Reaction: L => ; T, R, I, Rate Law: compartment*(m*L+q*L*T+z*L*L*R*I/(kappa+I))
vs = 0.0Reaction: => S, Rate Law: compartment*vs
a = 0.2065; b = 2.145E-10Reaction: => T, Rate Law: compartment*a*T*(1-b*T)

States:

NameDescription
I[Interleukin-2]
S[C71622]
T[Neoplastic Cell]
C[Neoplastic Cell]
N[natural killer cell]
L[C12543]
R[CD4+ CD25+ Regulatory T Cells]

DeVries2000_PancreaticBetaCells_InsulinSecretion: BIOMD0000000371v0.0.1

This a model from the article: Channel sharing in pancreatic beta -cells revisited: enhancement of emergent bursting…

Details

Secretion of insulin by electrically coupled populations of pancreatic beta -cells is governed by bursting electrical activity. Isolated beta -cells, however, exhibit atypical bursting or continuous spike activity. We study bursting as an emergent property of the population, focussing on interactions among the subclass of spiking cells. These are modelled by equipping the fast subsystem with a saddle-node-loop bifurcation, which makes it monostable. Such cells can only spike tonically or remain silent when isolated, but can be induced to burst with weak diffusive coupling. With stronger coupling, the cells revert to tonic spiking. We demonstrate that the addition of noise dramatically increases, via a phenomenon like stochastic resonance, the coupling range over which bursting is seen. link: http://identifiers.org/pubmed/11093836

Parameters:

NameDescription
tau_potassium_current_n_gate = 20.0; n_infinity = 1.89405943825186E-4; lamda = 0.8Reaction: n = lamda*(n_infinity-n)/tau_potassium_current_n_gate, Rate Law: lamda*(n_infinity-n)/tau_potassium_current_n_gate
tau_s = 20000.0; s_infinity = 0.00460957217937421Reaction: s = (s_infinity-s)/tau_s, Rate Law: (s_infinity-s)/tau_s
tau_membrane = 20.0; i_Ca = -7.4446678508483; i_K = 5.0; i_K_ATP = 6.0; i_s = 1.0Reaction: V_membrane = (-(i_Ca+i_K+i_K_ATP+i_s))/tau_membrane, Rate Law: (-(i_Ca+i_K+i_K_ATP+i_s))/tau_membrane

States:

NameDescription
V membrane[membrane potential]
s[variant]
n[delayed rectifier potassium channel activity]

Dhawan2019 - Endogenous miRNA sponges mediate the generation of oscillatory dynamics for a non-coding RNA network: BIOMD0000000841v0.0.1

This is a delay differential equation model showing how non-coding RNA, acting as microRNA (miRNA) sponges in a conserve…

Details

Oscillations are crucial to the normal function of living organisms, across a wide variety of biological processes. In eukaryotes, oscillatory dynamics are thought to arise from interactions at the protein and RNA levels; however, the role of non-coding RNA in regulating these dynamics remains understudied. In this work, we show how non-coding RNA acting as microRNA (miRNA) sponges in a conserved miRNA - transcription factor feedback motif, can give rise to oscillatory behaviour, and how to test for this experimentally. Control of these non-coding RNA can dynamically create oscillations or stability, and we show how this behaviour predisposes to oscillations in the stochastic limit. These results, supported by emerging evidence for the role of miRNA sponges in development, point towards key roles of different species of miRNA sponges, such as circular RNA, potentially in the maintenance of yet unexplained oscillatory behaviour. These results help to provide a paradigm for understanding functional differences between the many redundant, but distinct RNA species thought to act as miRNA sponges in nature, such as long non-coding RNA, pseudogenes, competing mRNA, circular RNA, and3' UTRs. link: http://identifiers.org/pubmed/30385313

Parameters:

NameDescription
gamma_FM = 100.0; n = 8.0; tau1 = 0.5; beta_FM = 200.0Reaction: => M; P, Rate Law: compartment*beta_FM/((gamma_FM/delay(P, tau1))^n+1)
k_CM = 10.0Reaction: M + C =>, Rate Law: compartment*k_CM*M*C
alpha_M = 1.0Reaction: => M, Rate Law: compartment*alpha_M
delta_F = 0.1Reaction: F =>, Rate Law: compartment*delta_F*F
delta_M = 1.0Reaction: M =>, Rate Law: compartment*delta_M*M
alpha_F = 1.0Reaction: => F, Rate Law: compartment*alpha_F
alpha_C = 1.0Reaction: => C, Rate Law: compartment*alpha_C
delta_P = 0.1Reaction: P =>, Rate Law: compartment*delta_P*P
delta_C = 0.01Reaction: C =>, Rate Law: compartment*delta_C*C
tau2 = 0.5; k_P = 10.0Reaction: => P; F, Rate Law: compartment*k_P*delay(F, tau2)
k_MF = 0.1Reaction: M + F =>, Rate Law: compartment*k_MF*M*F

States:

NameDescription
M[C25966]
C[C25966]
P[1,4-beta-D-Mannooligosaccharide]
F[Messenger RNA]

diCenzo2020 - Sinorhizobium meliloti 1021 genome-scale metabolic model: MODEL2003240001v0.0.1

An updated representation of S. meliloti metabolism that was manually-curated and encompasses information from 240 liter…

Details

The mutualistic association between leguminous plants and endosymbiotic rhizobial bacteria is a paradigmatic example of a symbiosis driven by metabolic exchanges. Here, we report the reconstruction and modelling of a genome-scale metabolic network of Medicago truncatula (plant) nodulated by Sinorhizobium meliloti (bacterium). The reconstructed nodule tissue contains five spatially distinct developmental zones and encompasses the metabolism of both the plant and the bacterium. Flux balance analysis (FBA) suggests that the metabolic costs associated with symbiotic nitrogen fixation are primarily related to supporting nitrogenase activity, and increasing N2-fixation efficiency is associated with diminishing returns in terms of plant growth. Our analyses support that differentiating bacteroids have access to sugars as major carbon sources, ammonium is the main nitrogen export product of N2-fixing bacteria, and N2 fixation depends on proton transfer from the plant cytoplasm to the bacteria through acidification of the peribacteroid space. We expect that our model, called 'Virtual Nodule Environment' (ViNE), will contribute to a better understanding of the functioning of legume nodules, and may guide experimental studies and engineering of symbiotic nitrogen fixation. link: http://identifiers.org/pubmed/32444627

Diedrichs2018 - A data-entrained computational model for testing the regulatory logic of the vertebrate unfolded protein response: BIOMD0000000703v0.0.1

A data-entrained computational model for testing the regulatory logic of the vertebrate unfolded protein responseThis mo…

Details

The vertebrate unfolded protein response (UPR) is characterized by multiple interacting nodes among its three pathways, yet the logic underlying this regulatory complexity is unclear. To begin to address this issue, we created a computational model of the vertebrate UPR that was entrained upon and then validated against experimental data. As part of this validation, the model successfully predicted the phenotypes of cells with lesions in UPR signaling, including a surprising and previously unreported differential role for the eIF2α phosphatase GADD34 in exacerbating severe stress but ameliorating mild stress. We then used the model to test the functional importance of a feedforward circuit within the PERK/CHOP axis and of cross-regulatory control of BiP and CHOP expression. We found that the wiring structure of the UPR appears to balance the ability of the response to remain sensitive to endoplasmic reticulum stress and to be deactivated rapidly by improved protein-folding conditions. This model should serve as a valuable resource for further exploring the regulatory logic of the UPR. link: http://identifiers.org/pubmed/29668363

Parameters:

NameDescription
A6_star = 1.0 1; B = 0.444444444444444 1; kcl=4.0; A6tot_norm = 15.0 1; kdA6 = 0.00384 1/(16.6667*s); KBA6 = 1.6E-5 1; U_star = 1.0 1Reaction: => A6; U, A6, Rate Law: ER*(kdA6*A6_star+kcl*(U-U_star)*(A6tot_norm-A6)/(1+B/KBA6))
KA4g=0.75; C_star = 1.0 1; etaC=0.012; Kth4g=0.1; kdg = 0.003468 1/(16.6667*s); g_star = 1.0 1; KC=5.0; A4_star = 1.0 1Reaction: => g; A4, C, Rate Law: ER*(kdg*g_star+etaC*((A4-A4_star)+KA4g*(A4-A4_star)*(C-C_star))/((A4-A4_star)+Kth4g*(A4-A4_star)*(C-C_star)+KC))
C_star = 1.0 1; ktC=1.0E-4; kdC = 0.005478 1/(16.6667*s); c_star = 1.0 1; Ep_star = 1.0 1Reaction: => C; Ep, c, Rate Law: ER*(kdC*C_star/c_star+ktC*(Ep-Ep_star))*c
kdx = 0.006546 1/(16.6667*s)Reaction: x => ; A6, Rate Law: ER*kdx*x
Ip_star = 1.0 1; KII=0.01; B = 0.444444444444444 1; delta=1.5; Ip = 1.0 1Reaction: U => ; x, Rate Law: ER*delta*U/(1+KII*(Ip-Ip_star))*B
kdb = 0.001284 1/(16.6667*s); alphaI = 0.2 1; Ip_star = 1.0 1; betaI = 0.1 1; Ip = 1.0 1Reaction: b => ; A4, A6, Rate Law: ER*kdb*(1+alphaI*(Ip-Ip_star))/(1+betaI*(Ip-Ip_star))*b
ksp=0.00785; Kx=3.0; xtot_norm = 16.0 1; Ip = 1.0 1Reaction: => x, Rate Law: ER*ksp*Ip*(xtot_norm-x)/((Kx+xtot_norm)-x)
gamma=0.001; A4_star = 1.0 1; U_star = 1.0 1; kdA4 = 0.00384 1/(16.6667*s)Reaction: => A4; U, Ep, Rate Law: ER*(kdA4*A4_star+gamma*(U-U_star)*Ep)
kdA4 = 0.00384 1/(16.6667*s)Reaction: A4 =>, Rate Law: ER*kdA4*A4
kdC = 0.005478 1/(16.6667*s)Reaction: C =>, Rate Law: ER*kdC*C
kdg = 0.003468 1/(16.6667*s)Reaction: g => ; C, Rate Law: ER*kdg*g
KA4c=2.0; Kth4c=0.25; c_star = 1.0 1; muA4=0.1; A4_star = 1.0 1; Kc4=0.56; n=2.0; kdc = 0.00393 1/(16.6667*s)Reaction: => c; A6, A4, C, Rate Law: ER*(kdc*c_star+muA4*(1+Kc4*A6)*(A4-A4_star)^n/((A4-A4_star)^n+KA4c^n*(1+Kth4c*A6)^n))
kdG = 0.003852 1/(16.6667*s)Reaction: G =>, Rate Law: ER*kdG*G
kdB = 2.514E-4 1/(16.6667*s); b_star = 1.0 1; Btot_star = 1.0 1Reaction: => Btot; b, Rate Law: ER*kdB*Btot_star/b_star*b
kdc = 0.00393 1/(16.6667*s)Reaction: c => ; C, Rate Law: ER*kdc*c
A6_star = 1.0 1; nA4=2.0; nA=7.0; KA6=3.0; Kth6=1.0E-5; x_star = 1.0 1; Ip = 1.0 1; Kb4=0.5; alphaX=0.002; alphaA6=0.012; kdb = 0.001284 1/(16.6667*s); Ip_star = 1.0 1; betaI = 0.1 1; KA4=3.0; alphaA4=0.007; Kb6=0.56; alphaI = 0.2 1; nA6=2.0; b_star = 1.0 1; KX=8.0; Kth4=0.167; A4_star = 1.0 1Reaction: => b; A4, A6, x, Rate Law: ER*(kdb*(1+alphaI*(Ip-Ip_star))/(1+betaI*(Ip-Ip_star))*b_star+alphaA6*(1+Kb6*A4)*(A6-A6_star)^nA6/((A6-A6_star)^nA6+KA6^nA6*(1+Kth6*A4^nA))+alphaA4*(1+Kb4*A6)*(A4-A4_star)^nA4/((A4-A4_star)^nA4+KA4^nA4*(1+Kth4*A6)^nA4)+alphaX*(x-x_star)/((x-x_star)+KX))
Etot_norm = 20.0 1; Kph=14.0; kph=0.00651; Pp = 1.0 1Reaction: => Ep, Rate Law: ER*kph*(Etot_norm-Ep)*Pp/(Kph+(Etot_norm-Ep))
kdeph1=0.03; kdeph2=0.08; G_star = 1.0 1; Kdeph=7.0Reaction: Ep => ; G, Rate Law: ER*(kdeph1+kdeph2*(G-G_star))*Ep/(Kdeph+Ep)
kdB = 2.514E-4 1/(16.6667*s)Reaction: Btot =>, Rate Law: ER*kdB*Btot
kdA6 = 0.00384 1/(16.6667*s)Reaction: A6 =>, Rate Law: ER*kdA6*A6
KUI=0.01; Ip_star = 1.0 1; KUI = 2.17848410757946 1; Stress = 2.0 1/(16.6667*s); KE=3.0; ksU=0.89; n=4.0; Ip = 1.0 1; KUU=6.0Reaction: => U; Ep, U, Rate Law: ER*(ksU/(1+KUI*(Ip-Ip_star))+Stress)/(1+Ep/KE+(U/KUU)^n)
g_star = 1.0 1; G_star = 1.0 1; ktG=0.0015; kdG = 0.003852 1/(16.6667*s); Ep_star = 1.0 1Reaction: => G; Ep, g, Rate Law: ER*(kdG*G_star/g_star+ktG*(Ep-Ep_star))*g

States:

NameDescription
g[Protein phosphatase 1 regulatory subunit 15A; mRNA]
c[DNA damage-inducible transcript 3 protein; mRNA]
C[DNA damage-inducible transcript 3 protein]
b[Immunoglobulin Binding Protein; mRNA]
A4[Cyclic AMP-dependent transcription factor ATF-4]
x[X-box-binding protein 1]
G[Protein phosphatase 1 regulatory subunit 15A]
Ep[Eukaryotic translation initiation factor 2 subunit 1; phosphorylation]
U[unfolded protein [endoplasmic reticulum lumen]]
Btot[Immunoglobulin Binding Protein]
A6[Cyclic AMP-dependent transcription factor ATF-6 alpha; Cyclic AMP-dependent transcription factor ATF-6 beta]

Dikicioglu2018 - Genome-scale metabolic model of Saccharomyces cerevisiae including comprehensive iron metabolism network: MODEL1709260000v0.0.1

This SBML representation of the yeast metabolic network is made available under the Creative Commons Attribution-Share A…

Details

Metabolic networks adapt to changes in their environment by modulating the activity of their enzymes and transporters; often by changing their abundance. Understanding such quantitative changes can shed light onto how metabolic adaptation works, or how it can fail and lead to a metabolically dysfunctional state. We propose a strategy to quantify metabolic protein requirements for cofactor-utilising enzymes and transporters through constraint-based modelling. The first eukaryotic genome-scale metabolic model to comprehensively represent iron metabolism was constructed, extending the most recent community model of the Saccharomyces cerevisiae metabolic network. Partial functional impairment of the genes involved in the maturation of iron-sulphur (Fe-S) proteins was investigated employing the model and the in silico analysis revealed extensive rewiring of the fluxes in response to this functional impairment, despite its marginal phenotypic effect. The optimal turnover rate of enzymes bearing ion cofactors can be determined via this novel approach; yeast metabolism, at steady state, was determined to employ a constant turnover of its iron-recruiting enzyme at a rate of 3.02 × 10 -11  mmol·(g biomass) -1 ·h  -1 . link: http://identifiers.org/pubmed/30578666

DiVentura2011_Min_System_E_coli: MODEL1101180000v0.0.1

This is an SBML version with MesoRD annotations of the model described in: **Self-organized partitioning of dynamicall…

Details

How cells manage to get equal distribution of their structures and molecules at cell division is a crucial issue in biology. In principle, a feedback mechanism could always ensure equality by measuring and correcting the distribution in the progeny. However, an elegant alternative could be a mechanism relying on self-organization, with the interplay between system properties and cell geometry leading to the emergence of equal partitioning. The problem is exemplified by the bacterial Min system that defines the division site by oscillating from pole to pole. Unequal partitioning of Min proteins at division could negatively impact system performance and cell growth because of loss of Min oscillations and imprecise mid-cell determination. In this study, we combine live cell and computational analyses to show that known properties of the Min system together with the gradual reduction of protein exchange through the constricting septum are sufficient to explain the observed highly precise spontaneous protein partitioning. Our findings reveal a novel and effective mechanism of protein partitioning in dividing cells and emphasize the importance of self-organization in basic cellular processes. link: http://identifiers.org/pubmed/21206490

Dokos1996_SinoatrialNode: MODEL0912503622v0.0.1

This a model from the article: Ion currents underlying sinoatrial node pacemaker activity: a new single cell mathemati…

Details

The ionic currents underlying autorhythmicity of the mammalian sinoatrial node and their wider contribution to each phase of the action potential have been investigated in this study using a new single cell mathematical model. The new model provides a review and update of existing formulations of sinoatrial node membrane currents, derived from a wide range of electrophysiological data available in the literature. Simulations of spontaneous activity suggest that the dominant mechanism underlying pacemaker depolarisation is the inward background Na+ current, ib,Na. In contrast to previous models, the decay of the delayed rectifying K+ current, iK, was insignificant during this phase. Despite the presence of a pseudo-outward background current throughout the pacemaker range of potentials (Na-K pump+leak currents), the hyperpolarisation-activated current i(f) was not essential to pacemaker activity. A closer inspection of the current-voltage characteristics of the model revealed that the "instantaneous" time-independent current was inward for holding potentials in the pacemaker range, which rapidly became outward within 2 ms due to the inactivation of the L-type Ca2+ current, iCa,L. This suggests that reports in the literature in which the net background current is outward throughout the pacemaker range of potentials may be exaggerated. The magnitudes of the action potential overshoot and the maximum diastolic potential were determined largely by the reversal potentials of iCa,L and iK respectively. The action potential was sustained by the incomplete deactivation of iCa,L and the Na-Ca exchanger, iNaCa. Despite the incorporation of "square-root" activation by [K]o of all K+ currents, the model was unable to correctly simulate the response to elevated [K]o. link: http://identifiers.org/pubmed/8869126

DoldánMartelli2013 - A Mathematical Model for the Rational Design of Chimeric Ligands in Selective Drug Therapies: MODEL1907310001v0.0.1

This model attempts to provide a mathematical framework for describing the dynamics of receptor-drug complex formation o…

Details

Chimeric drugs with selective potential toward specific cell types constitute one of the most promising forefronts of modern Pharmacology. We present a mathematical model to test and optimize these synthetic constructs, as an alternative to conventional empirical design. We take as a case study a chimeric construct composed of epidermal growth factor (EGF) linked to different mutants of interferon (IFN). Our model quantitatively reproduces all the experimental results, illustrating how chimeras using mutants of IFN with reduced affinity exhibit enhanced selectivity against cell overexpressing EGF receptor. We also investigate how chimeric selectivity can be improved based on the balance between affinity rates, receptor abundance, activity of ligand subunits, and linker length between subunits. The simplicity and generality of the model facilitate a straightforward application to other chimeric constructs, providing a quantitative systematic design and optimization of these selective drugs against certain cell-based diseases, such as Alzheimer's and cancer.CPT: Pharmacometrics & Systems Pharmacology (2013) 2, e26; doi:10.1038/psp.2013.2; advance online publication 13 February 2013. link: http://identifiers.org/pubmed/23887616

Dong2014 - Mathematical modeling on helper t cells in a tumor immune system: BIOMD0000000783v0.0.1

This model gives a mathematical description of the interactions between tumor cells, cytotoxic T lymphocytes and helper…

Details

Activation of CD8+ cytotoxic T lymphocytes (CTLs) is naturally regarded as a major antitumor mechanism of the immune system. In contrast, CD4+ T cells are commonly classified as helper T cells (HTCs) on the basis of their roles in providing help to the generation and maintenance of effective CD8+ cytotoxic and memory T cells. In order to get a better insight on the role of HTCs in a tumor immune system, we incorporate the third population of HTCs into a previous two dimensional ordinary differential equations (ODEs) model. Further we introduce the adoptive cellular immunotherapy (ACI) as the treatment to boost the immune system to fight against tumors. Compared tumor cells (TCs) and effector cells (ECs), the recruitment of HTCs changes the dynamics of the system substantially, by the effects through particular parameters, i.e., the activation rate of ECs by HTCs, π (scaled as π), and the HTCs stimulation rate by the presence of identified tumor antigens, k2 (scaled as υ2). We describe the stability regions of the interior equilibria É (no treatment case) and E+ (treatment case) in the scaled (π,υ2) parameter space respectively. Both π and υ2 can destabilize É and E+ and cause Hopf bifurcations. Our results show that HTCs might play a crucial role in the long term periodic oscillation behaviors of tumor immune system interactions. They also show that TCs may be eradicated from the patient's body under the ACI treatment. link: http://identifiers.org/doi/10.3934/dcdsb.2014.19.55

Parameters:

NameDescription
beta = 0.002; alpha = 1.636Reaction: => x_Tumor_Cells, Rate Law: compartment*alpha*x_Tumor_Cells*(1-beta*x_Tumor_Cells)
rho = 0.01Reaction: => y_Effector_Cells; z_Helper_T_Cells, Rate Law: compartment*rho*y_Effector_Cells*z_Helper_T_Cells
delta_2 = 0.055Reaction: z_Helper_T_Cells =>, Rate Law: compartment*delta_2*z_Helper_T_Cells
delta_1 = 0.3743Reaction: y_Effector_Cells =>, Rate Law: compartment*delta_1*y_Effector_Cells
omega_2 = 0.002Reaction: => z_Helper_T_Cells; x_Tumor_Cells, Rate Law: compartment*omega_2*x_Tumor_Cells*z_Helper_T_Cells
omega_1 = 0.04Reaction: => y_Effector_Cells; x_Tumor_Cells, Rate Law: compartment*omega_1*x_Tumor_Cells*y_Effector_Cells
sigma_2 = 0.38Reaction: => z_Helper_T_Cells, Rate Law: compartment*sigma_2

States:

NameDescription
x Tumor Cells[neoplastic cell]
y Effector Cells[Effector Immune Cell]
z Helper T Cells[helper T cell]

Dong2018 - E2F dynamics in G1 phase: MODEL1811050001v0.0.1

The length of the G1 phase in the cell cycle shows significant variability in different cell types and tissue types. To…

Details

The length of the G1 phase in the cell cycle shows significant variability in different cell types and tissue types. To gain insights into the control of G1 length, we generated an E2F activity reporter that captures free E2F activity after dissociation from Rb sequestration and followed its kinetics of activation at the single-cell level, in real time. Our results demonstrate that its activity is precisely coordinated with S phase progression. Quantitative analysis indicates that there is a pre-S phase delay between E2F transcriptional dynamic and activity dynamics. This delay is variable among different cell types and is strongly modulated by the cyclin D/CDK4/6 complex activity through Rb phosphorylation. Our findings suggest that the main function of this complex is to regulate the appropriate timing of G1 length. link: http://identifiers.org/pubmed/29309421

Dorvash2019 - Dynamic modeling of signal transduction by mTOR complexes in cancer: BIOMD0000000822v0.0.1

This model is based on: Dynamic modeling of signal transduction by mTOR complexes in cancer Author: Mohammadreza Do…

Details

Signal integration has a crucial role in the cell fate decision and dysregulation of the cellular signaling pathways is a primary characteristic of cancer. As a signal integrator, mTOR shows a complex dynamical behavior which determines the cell fate at different cellular processes levels, including cell cycle progression, cell survival, cell death, metabolic reprogramming, and aging. The dynamics of the complex responses to rapamycin in cancer cells have been attributed to its differential time-dependent inhibitory effects on mTORC1 and mTORC2, the two main complexes of mTOR. Two explanations were previously provided for this phenomenon: 1-Rapamycin does not inhibit mTORC2 directly, whereas it prevents mTORC2 formation by sequestering free mTOR protein (Le Chatelier's principle). 2-Components like Phosphatidic Acid (PA) further stabilize mTORC2 compared with mTORC1. To understand the mechanism by which rapamycin differentially inhibits the mTOR complexes in the cancer cells, we present a mathematical model of rapamycin mode of action based on the first explanation, i.e., Le Chatelier's principle. Translating the interactions among components of mTORC1 and mTORC2 into a mathematical model revealed the dynamics of rapamycin action in different doses and time-intervals of rapamycin treatment. This model shows that rapamycin has stronger effects on mTORC1 compared with mTORC2, simply due to its direct interaction with free mTOR and mTORC1, but not mTORC2, without the need to consider other components that might further stabilize mTORC2. Based on our results, even when mTORC2 is less stable compared with mTORC1, it can be less inhibited by rapamycin. link: http://identifiers.org/pubmed/31493485

Parameters:

NameDescription
K_el_Rapam = 0.0718632 1/hReaction: Cytosolic_Rapamycin =>, Rate Law: compartment*K_el_Rapam*Cytosolic_Rapamycin
K_syn_mTOR = 1.6E-27 mol/sReaction: => mTOR, Rate Law: compartment*K_syn_mTOR
K_syn_Rictor = 5.9E-28 mol/sReaction: => Rictor, Rate Law: compartment*K_syn_Rictor
Rapamycin_Dose = 0.0; K_abs_Rapam = 2.77 1/hReaction: => Cytosolic_Rapamycin, Rate Law: compartment*K_abs_Rapam*Rapamycin_Dose
k_forward_Raptor_release = 0.01 1/s; k_reverse_Raptor_release = 1.0E-5 1/(mol*s)Reaction: mTORC1_Rapamycin => mTOR_Rapamycin + Raptor, Rate Law: compartment*(k_forward_Raptor_release*mTORC1_Rapamycin-k_reverse_Raptor_release*mTOR_Rapamycin*Raptor)
k_form_mTOR_Rapam = 1920000.0 1/(mol*s); k_diss_mTOR_Rapam = 0.022 1/sReaction: mTOR + Cytosolic_Rapamycin => mTOR_Rapamycin, Rate Law: compartment*(k_form_mTOR_Rapam*mTOR*Cytosolic_Rapamycin-k_diss_mTOR_Rapam*mTOR_Rapamycin)
K_syn_Raptor = 2.15E-27 mol/sReaction: => Raptor, Rate Law: compartment*K_syn_Raptor
K_deg_Raptor = 1.0E-8 1/sReaction: Raptor =>, Rate Law: compartment*K_deg_Raptor*Raptor
K_deg_mTOR = 1.0E-8 1/sReaction: mTOR =>, Rate Law: compartment*K_deg_mTOR*mTOR
k_form_C2 = 1.6666666E7 1/(mol*s); k_diss_C2 = 0.08333 1/sReaction: mTOR + Rictor => mTORC2, Rate Law: compartment*(k_form_C2*mTOR*Rictor-k_diss_C2*mTORC2)
k_form_C1 = 1.6666666E7 1/(mol*s); k_diss_C1 = 0.08333 1/sReaction: mTOR + Raptor => mTORC1, Rate Law: compartment*(k_form_C1*mTOR*Raptor-k_diss_C1*mTORC1)
k_diss_C1_Rapam = 0.022 1/s; k_form_C1_Rapam = 1920000.0 1/(mol*s)Reaction: mTORC1 + Cytosolic_Rapamycin => mTORC1_Rapamycin, Rate Law: compartment*(k_form_C1_Rapam*mTORC1*Cytosolic_Rapamycin-k_diss_C1_Rapam*mTORC1_Rapamycin)
K_deg_Rictor = 1.0E-8 1/sReaction: Rictor =>, Rate Law: compartment*K_deg_Rictor*Rictor

States:

NameDescription
Raptor[Regulatory-Associated Protein of mTOR]
mTOR[431220; mTOR Inhibitor]
mTORC2[mTORC2]
Rictor[Rapamycin-Insensitive Companion of mTOR]
mTOR Rapamycin[sirolimus; mTOR Inhibitor; 431220]
Cytosolic Rapamycin[sirolimus; Cytosol]
mTORC1[mTORC1]
mTORC1 Rapamycin[sirolimus; mTORC1]

Draghi2019 - Parameter identification of a model for prostate cancer treated by intermittent therapy: MODEL1911100005v0.0.1

This is a mathematical describing the interaction between the prostate adenocarcinoma tumor environment, the prostate sp…

Details

Adenocarcinoma is the most frequent cancer affecting the prostate walnut-size gland in the male reproductive system. Such cancer may have a very slow progression or may be associated with a "dark prognosis" when tumor cells are spreading very quickly. Prostate cancers have the particular properties to be marked by the level of prostate specific antigen (PSA) in blood which allows to follow its evolution. At least in its first phase, prostate adenocarcinoma is most often hormone-dependent and, consequently, hormone therapy is a possible treatment. Since few years, hormone therapy started to be provided intermittently for improving patient's quality of life. Today, durations of on- and off-treatment periods are still chosen empirically, most likely explaining why there is no clear benefit from the survival point of view. We therefore developed a model for describing the interaction between the tumor environment, the PSA produced by hormone-dependent and hormone-independent tumor cells, respectively, and the level of androgens. Model parameters were identified using a genetic algorithm applied to the PSA time series measured in a few patients who initially received prostatectomy and were then treated by intermittent hormone therapy (LHRH analogs and anti-androgen). The measured PSA time series is quite correctly reproduced by free runs over the whole follow-up. Model parameter values allow for distinguishing different types of patient (age and Gleason score) meaning that the model can be individualized. We thus showed that the long-term evolution of the cancer can be affected by durations of on- and off-treatment periods. link: http://identifiers.org/pubmed/30292801

Dreyfuss2013 - Genome-Scale Metabolic Model - N.crassa iJDZ836: MODEL1212060001v0.0.1

Dreyfuss2013 - Genome-Scale Metabolic Model - N.crassa iJDZ836Genome-scale metabolic model of the filamentous fungus Neu…

Details

The filamentous fungus Neurospora crassa played a central role in the development of twentieth-century genetics, biochemistry and molecular biology, and continues to serve as a model organism for eukaryotic biology. Here, we have reconstructed a genome-scale model of its metabolism. This model consists of 836 metabolic genes, 257 pathways, 6 cellular compartments, and is supported by extensive manual curation of 491 literature citations. To aid our reconstruction, we developed three optimization-based algorithms, which together comprise Fast Automated Reconstruction of Metabolism (FARM). These algorithms are: LInear MEtabolite Dilution Flux Balance Analysis (limed-FBA), which predicts flux while linearly accounting for metabolite dilution; One-step functional Pruning (OnePrune), which removes blocked reactions with a single compact linear program; and Consistent Reproduction Of growth/no-growth Phenotype (CROP), which reconciles differences between in silico and experimental gene essentiality faster than previous approaches. Against an independent test set of more than 300 essential/non-essential genes that were not used to train the model, the model displays 93% sensitivity and specificity. We also used the model to simulate the biochemical genetics experiments originally performed on Neurospora by comprehensively predicting nutrient rescue of essential genes and synthetic lethal interactions, and we provide detailed pathway-based mechanistic explanations of our predictions. Our model provides a reliable computational framework for the integration and interpretation of ongoing experimental efforts in Neurospora, and we anticipate that our methods will substantially reduce the manual effort required to develop high-quality genome-scale metabolic models for other organisms. link: http://identifiers.org/pubmed/23935467

Dritschel2018 - A mathematical model of cytotoxic and helper T cell interactions in a tumour microenvironment: BIOMD0000000763v0.0.1

This model examines the role of helper and cytotoxic T cells in an anti-tumour response, with implicit inclusions of imm…

Details

We develop a mathematical model to examine the role of helper and cytotoxic T cells in an anti-tumour immune response. The model comprises three ordinary differential equations describing the dynamics of the tumour cells, the helper and the cytotoxic T cells, and implicitly accounts for immunosuppressive effects. The aim is to investigate how the anti-tumour immune response varies with the level of infiltrating helper and cytotoxic T cells. Through a combination of analytical studies and numerical simulations, our model exemplifies the three Es of immunoediting: elimination, equilibrium and escape. Specifically, it reveals that the three Es of immunoediting depend highly on the infiltration rates of the helper and cytotoxic T cells. The model’s results indicate that both the helper and cytotoxic T cells play a key role in tumour elimination. They also show that combination therapies that boost the immune system and block tumour-induced immunosuppression may have a synergistic effect in reducing tumour growth. link: http://identifiers.org/doi/10.1080/23737867.2018.1465863

Parameters:

NameDescription
gamma = 10.0Reaction: => N_Tumour; N_Tumour, Rate Law: compartment*gamma*(1-N_Tumour)*N_Tumour
sigma_H = 0.5Reaction: => T_H, Rate Law: compartment*sigma_H
Ntilde = 0.04; alpha = 0.19Reaction: => T_H; T_H, N_Tumour, Rate Law: compartment*alpha*N_Tumour*T_H/(Ntilde^2+N_Tumour^2)
sigma_C = 2.0Reaction: => T_C, Rate Law: compartment*sigma_C
delta_H = 1.0Reaction: T_H =>, Rate Law: compartment*delta_H*T_H
p = 0.5; k = 4.15Reaction: T_C => ; N_Tumour, Rate Law: compartment*(1-p)*k*T_C*N_Tumour

States:

NameDescription
T C[cytotoxic T cell]
N Tumour[Neoplastic Cell]
T H[helper T cell]

Duarte2004 - Genome-scale metabolic network of Saccharomyces cerevisiae (iND750): MODEL1507180019v0.0.1

Duarte2004 - Genome-scale metabolic network of Saccharomyces cerevisiae (iND750)This model is described in the article:…

Details

A fully compartmentalized genome-scale metabolic model of Saccharomyces cerevisiae that accounts for 750 genes and their associated transcripts, proteins, and reactions has been reconstructed and validated. All of the 1149 reactions included in this in silico model are both elementally and charge balanced and have been assigned to one of eight cellular locations (extracellular space, cytosol, mitochondrion, peroxisome, nucleus, endoplasmic reticulum, Golgi apparatus, or vacuole). When in silico predictions of 4154 growth phenotypes were compared to two published large-scale gene deletion studies, an 83% agreement was found between iND750's predictions and the experimental studies. Analysis of the failure modes showed that false predictions were primarily caused by iND750's limited inclusion of cellular processes outside of metabolism. This study systematically identified inconsistencies in our knowledge of yeast metabolism that require specific further experimental investigation. link: http://identifiers.org/pubmed/15197165

Duarte2007_Homo_sapiens_Metabol_Recon_1: MODEL6399676120v0.0.1

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

Details

Metabolism is a vital cellular process, and its malfunction is a major contributor to human disease. Metabolic networks are complex and highly interconnected, and thus systems-level computational approaches are required to elucidate and understand metabolic genotype-phenotype relationships. We have manually reconstructed the global human metabolic network based on Build 35 of the genome annotation and a comprehensive evaluation of >50 years of legacy data (i.e., bibliomic data). Herein we describe the reconstruction process and demonstrate how the resulting genome-scale (or global) network can be used (i) for the discovery of missing information, (ii) for the formulation of an in silico model, and (iii) as a structured context for analyzing high-throughput biological data sets. Our comprehensive evaluation of the literature revealed many gaps in the current understanding of human metabolism that require future experimental investigation. Mathematical analysis of network structure elucidated the implications of intracellular compartmentalization and the potential use of correlated reaction sets for alternative drug target identification. Integrated analysis of high-throughput data sets within the context of the reconstruction enabled a global assessment of functional metabolic states. These results highlight some of the applications enabled by the reconstructed human metabolic network. The establishment of this network represents an important step toward genome-scale human systems biology. link: http://identifiers.org/pubmed/17267599

Dubey2007 - A mathematical model for the effect of toxicant on the immune system (with toxicant effect) Model2: BIOMD0000000905v0.0.1

In this paper, a nonlinear mathematical model is proposed and analyzed to study the effect of environmental toxicant on…

Details

In this paper, a nonlinear mathematical model is proposed and analyzed to study the effect of environmental toxicant on the immune response of the body. Criteria for local stability, instability and global stability are obtained. It is shown that the immune response of the body decreases as the concentration of environmental toxicant increases, and certain criteria are obtained under which it settles down at its equilibrium level. In the absence of toxicant, an oscillatory behavior of immune system and pathogenic growth is observed. However, in the presence of toxicant, oscillatory behavior is not observed. These studies show that the toxicant may have a grave effect on our body's defense mechanism. link: http://identifiers.org/doi/10.1142/S0218339007002301

Parameters:

NameDescription
Q0 = 5.0Reaction: => T, Rate Law: compartment*Q0
beta = 0.5Reaction: => P, Rate Law: compartment*beta*P
delta_0 = 0.4Reaction: T =>, Rate Law: compartment*delta_0*T
alpha0 = 0.1Reaction: M =>, Rate Law: compartment*alpha0*M
a = 0.8; gamma = 0.05; n = 0.1; k1 = 0.6; n1 = 0.5Reaction: I => ; P, U, Rate Law: compartment*(a*I+n*gamma*I*P+n1*k1*U*I)
theta_0 = 1.2Reaction: => U; T, Rate Law: compartment*theta_0*T
mu = 0.04; b = 0.3Reaction: => I; P, Rate Law: compartment*(mu+b*I*P)
beta0 = 0.2; gamma = 0.05Reaction: P => ; I, Rate Law: compartment*(gamma*I*P+beta0*P*P)
delta_1 = 0.02; k1 = 0.6Reaction: U => ; I, Rate Law: compartment*(delta_1*U+k1*U*I)
alpha = 2.4Reaction: => M; P, Rate Law: compartment*alpha*P

States:

NameDescription
I[BTO:0005810]
UU
MM
P[Microorganism]
T[C894]

Dubey2007 - A mathematical model for the effect of toxicant on the immune system (without toxicant effect) Model1: BIOMD0000000906v0.0.1

In this paper, a nonlinear mathematical model is proposed and analyzed to study the effect of environmental toxicant on…

Details

In this paper, a nonlinear mathematical model is proposed and analyzed to study the effect of environmental toxicant on the immune response of the body. Criteria for local stability, instability and global stability are obtained. It is shown that the immune response of the body decreases as the concentration of environmental toxicant increases, and certain criteria are obtained under which it settles down at its equilibrium level. In the absence of toxicant, an oscillatory behavior of immune system and pathogenic growth is observed. However, in the presence of toxicant, oscillatory behavior is not observed. These studies show that the toxicant may have a grave effect on our body's defense mechanism. link: http://identifiers.org/doi/10.1142/S0218339007002301

Parameters:

NameDescription
beta = 0.9Reaction: => P, Rate Law: compartment*beta*P
alpha0 = 0.1Reaction: M =>, Rate Law: compartment*alpha0*M
mu = 0.04; b = 0.3Reaction: => I; P, Rate Law: compartment*(mu+b*I*P)
beta0 = 0.2; gamma = 0.05Reaction: P => ; I, Rate Law: compartment*(gamma*I*P+beta0*P*P)
a = 0.8; gamma = 0.05; n = 0.1Reaction: I => ; P, Rate Law: compartment*(a*I+n*gamma*I*P)
alpha = 2.4Reaction: => M; P, Rate Law: compartment*alpha*P

States:

NameDescription
I[BTO:0005810]
MM
P[Microorganism]

Dubey2008 - Modeling the interaction between avascular cancerous cells and acquired immune response: BIOMD0000000886v0.0.1

MODELING THE INTERACTION BETWEEN AVASCULAR CANCEROUS CELLS AND ACQUIRED IMMUNE RESPONSE B. DUBEY, UMA S. DUBEY and SAND…

Details

This paper deals with the interaction between dispersed cancer cells and the major populations of the immune system, namely, the T helper cells, T Cytotoxic cells, B cells, and antibodies produced. The system is described by a set of five ordinary differential equations. Both local and global stability of the system has been investigated. It has been observed that under appropriate conditions this interaction is capable of controlling the growth of these cancer cells. The analytical findings are supported by numerical and computational analytical methods. link: http://identifiers.org/doi/10.1142/S0218339008002605

Parameters:

NameDescription
mu_10 = 0.2Reaction: Th =>, Rate Law: compartment*mu_10*Th
alpha = 0.18Reaction: => T, Rate Law: compartment*alpha*T
gamma_2 = 0.3; mu_3 = 0.45; gamma_1 = 0.4Reaction: => B; T, Th, Rate Law: compartment*(mu_3*T+gamma_1*T*B+gamma_2*Th*B)
mu_2 = 1.4; beta_1 = 0.3; beta_2 = 0.05Reaction: => Tc; T, Th, Rate Law: compartment*(mu_2*T+beta_1*T*Tc+beta_2*Th*Tc)
mu_30 = 0.03Reaction: B =>, Rate Law: compartment*mu_30*B
mu_1 = 1.5; mu_11 = 0.3Reaction: => Th; T, Rate Law: compartment*(mu_1*T+mu_11*T*Th)
alpha_0 = 4.6; alpha_1 = 0.101; delta_2 = 0.008Reaction: T => ; Tc, A, Rate Law: compartment*(alpha_0*T*T+alpha_1*T*Tc+delta_2*T*A)
mu_20 = 0.0412Reaction: Tc =>, Rate Law: compartment*mu_20*Tc
mu_40 = 0.3; delta_1 = 0.5Reaction: A => ; T, Rate Law: compartment*(mu_40*A+delta_1*T*A)
mu_4 = 0.35Reaction: => A; B, Rate Law: compartment*mu_4*B

States:

NameDescription
B[C12474]
A[Antibody]
Th[helper T-lymphocyte]
T[C12476]
TcTc

Ducrot2009 - Malaria transmission in two host types: MODEL1808280013v0.0.1

The main purpose of this article is to formulate a deterministic mathematical model for the transmission of malaria that…

Details

The main purpose of this article is to formulate a deterministic mathematical model for the transmission of malaria that considers two host types in the human population. The first type is called "non-immune" comprising all humans who have never acquired immunity against malaria and the second type is called "semi-immune". Non-immune are divided into susceptible, exposed and infectious and semi-immune are divided into susceptible, exposed, infectious and immune. We obtain an explicit formula for the reproductive number, R(0) which is a function of the weight of the transmission semi-immune-mosquito-semi-immune, R(0a), and the weight of the transmission non-immune-mosquito-non-immune, R(0e). Then, we study the existence of endemic equilibria by using bifurcation analysis. We give a simple criterion when R(0) crosses one for forward and backward bifurcation. We explore the possibility of a control for malaria through a specific sub-group such as non-immune or semi-immune or mosquitoes. link: http://identifiers.org/pubmed/22880962

Dudziuk2019 - Biologically sound formal model of Hsp70 heat induction: BIOMD0000000843v0.0.1

This is a mathematical model of Hsp70 induction. To model heat shock effects, the model incorporates temperature depende…

Details

A proper response to rapid environmental changes is essential for cell survival and requires efficient modifications in the pattern of gene expression. In this respect, a prominent example is Hsp70, a chaperone protein whose synthesis is dynamically regulated in stress conditions. In this paper, we expand a formal model of Hsp70 heat induction originally proposed in previous articles. To accurately capture various modes of heat shock effects, we not only introduce temperature dependencies in transcription to Hsp70 mRNA and in dissociation of transcriptional complexes, but we also derive a new formal expression for the temperature dependence in protein denaturation. We calibrate our model using comprehensive sets of both previously published experimental data and also biologically justified constraints. Interestingly, we obtain a biologically plausible temperature dependence of the transcriptional complex dissociation, despite the lack of biological constraints imposed in the calibration process. Finally, based on a sensitivity analysis of the model carried out in both deterministic and stochastic settings, we suggest that the regulation of the binding of transcriptional complexes plays a key role in Hsp70 induction upon heat shock. In conclusion, we provide a model that is able to capture the essential dynamics of the Hsp70 heat induction whilst being biologically sound in terms of temperature dependencies, description of protein denaturation and imposed calibration constraints. link: http://identifiers.org/pubmed/31181241

Parameters:

NameDescription
I_7_T = 0.6628Reaction: HSE_HSF_3 => HSE + HSF_3, Rate Law: compartment*I_7_T*HSE_HSF_3
k2 = 0.218Reaction: HSF + HSP => HSP_HSF, Rate Law: compartment*k2*HSF*HSP
k4 = 18.85Reaction: => HSP; mRNA, Rate Law: compartment*k4*mRNA
I1 = 0.003028Reaction: HSP_S => HSP + S, Rate Law: compartment*I1*HSP_S
I3 = 2.392Reaction: HSP + HSF_3 => HSF + HSP_HSF, Rate Law: compartment*I3*HSP*HSF_3
k_8_T = 96.0699331208704Reaction: => mRNA; HSE_HSF_3, Rate Law: compartment*k_8_T*HSE_HSF_3
k_11_T = 0.00191435208585195Reaction: P => S, Rate Law: compartment*k_11_T*P
I2 = 1.162Reaction: HSP_HSF => HSP + HSF, Rate Law: compartment*I2*HSP_HSF
k1 = 12.6Reaction: HSP + S => HSP_S, Rate Law: compartment*k1*HSP*S
k6 = 0.08899Reaction: => HSP, Rate Law: compartment*k6
k7 = 5892.0Reaction: HSE + HSF_3 => HSE_HSF_3, Rate Law: compartment*k7*HSE*HSF_3
k9 = 0.001888Reaction: HSP =>, Rate Law: compartment*k9*HSP
k3 = 446500.0Reaction: HSF => HSF_3, Rate Law: compartment*k3*HSF^3
k5 = 8.709E-4Reaction: mRNA =>, Rate Law: compartment*k5*mRNA
k10 = 0.09813Reaction: HSP_S => HSP + P, Rate Law: compartment*k10*HSP_S

States:

NameDescription
S[MI:0908; Protein]
HSF[CCO:37068]
HSP HSF[CCO:37068; Benzylpenicilloic acid]
P[Protein]
HSE[SO:0001850]
HSE HSF 3[SO:0001850; CCO:37068]
mRNA[Benzylpenicilloic acid; Messenger RNA]
HSP S[Benzylpenicilloic acid]
HSP[Benzylpenicilloic acid]
HSF 3[CCO:37068]

Dunster2014 - WBC Interactions (Model1): BIOMD0000000616v0.0.1

Dunster2014 - WBC Interactions (Model1)This is a sub-model of a three-step inflammatory response modelling study. The mo…

Details

There is growing interest in inflammation due to its involvement in many diverse medical conditions, including Alzheimer's disease, cancer, arthritis and asthma. The traditional view that resolution of inflammation is a passive process is now being superceded by an alternative hypothesis whereby its resolution is an active, anti-inflammatory process that can be manipulated therapeutically. This shift in mindset has stimulated a resurgence of interest in the biological mechanisms by which inflammation resolves. The anti-inflammatory processes central to the resolution of inflammation revolve around macrophages and are closely related to pro-inflammatory processes mediated by neutrophils and their ability to damage healthy tissue. We develop a spatially averaged model of inflammation centring on its resolution, accounting for populations of neutrophils and macrophages and incorporating both pro- and anti-inflammatory processes. Our ordinary differential equation model exhibits two outcomes that we relate to healthy and unhealthy states. We use bifurcation analysis to investigate how variation in the system parameters affects its outcome. We find that therapeutic manipulation of the rate of macrophage phagocytosis can aid in resolving inflammation but success is critically dependent on the rate of neutrophil apoptosis. Indeed our model predicts that an effective treatment protocol would take a dual approach, targeting macrophage phagocytosis alongside neutrophil apoptosis. link: http://identifiers.org/pubmed/25053556

Parameters:

NameDescription
Bat = 0.1; Gat = 1.0Reaction: => c; a, Rate Law: default_compartment*Gat*a^2/(Bat^2+a^2)/default_compartment
vt = 0.1Reaction: n =>, Rate Law: default_compartment*vt*n/default_compartment
t1 = 10.0; A = 1.0; alt = 0.05Reaction: => c, Rate Law: default_compartment*alt*piecewise(sin(time)^2, time < (A*pi), 0)*piecewise(1, time < t1, 0)/default_compartment
Tt = 0.001Reaction: a => ; m, Rate Law: default_compartment*Tt*m*a/default_compartment
Gmt = 0.01Reaction: m =>, Rate Law: default_compartment*Gmt*m/default_compartment
Gat = 1.0Reaction: a =>, Rate Law: default_compartment*Gat*a/default_compartment

States:

NameDescription
c[Interleukin-8]
m[macrophage]
a[neutrophil]
n[neutrophil]

Dunster2016 - Nondimensional Coagulation Model: BIOMD0000000925v0.0.1

We undertake a mathematical investigation of a model for the generation of thrombin, an enzyme central to haemostatic bl…

Details

We undertake a mathematical investigation of a model for the generation of thrombin, an enzyme central to haemostatic blood coagulation, as well as to thrombotic disorders, that is the end product of a complicated protein cascade with multiple feedbacks that ensures its production in the right place at the right time. In a laboratory setting, its central role is reflected in thrombin evolution over time being used as a measure of the ability of a patient's blood to clot. Here, we present a model for the generation of thrombin (based on earlier work) and analyse it using the method of matched asymptotic expansions to derive a sequence of simplified models that characterize the roles of distinct interactions over various timescales. In particular, we are able through the asymptotic analysis to provide simplified models that are an excellent substitute for the full model (capturing the explosive growth and decay of thrombin) and approximations for the key experimental measurements used to describe thrombin's characteristic evolution over time. The asymptotic results are validated against numerical simulations. link: http://identifiers.org/doi/10.1093/imamat/hxw007

Parameters:

NameDescription
k_tilde_3a = 150.0; q_tilde_3a = 1.0; k_tilde_3b = 0.038; k_tilde_1b = 0.19Reaction: Xa_ATIII = k_tilde_1b*Xa+k_tilde_3a*k_tilde_3b/q_tilde_3a*Va_Xa, Rate Law: k_tilde_1b*Xa+k_tilde_3a*k_tilde_3b/q_tilde_3a*Va_Xa
q_tilde_3a = 1.0; k_tilde_3b = 0.038; k_tilde_3c = 1.0Reaction: Va_Xa = (q_tilde_3a*Xa*Va-k_tilde_3b*Va_Xa)-k_tilde_3c*q_tilde_3a*APC*Va_Xa/(Va_Xa+1), Rate Law: (q_tilde_3a*Xa*Va-k_tilde_3b*Va_Xa)-k_tilde_3c*q_tilde_3a*APC*Va_Xa/(Va_Xa+1)
gamma_tilde_1a = 0.77; k_tilde_3a = 150.0; k_tilde_1a = 150.0; k_tilde_3c = 1.0; k_tilde_1b = 0.19Reaction: Xa = ((k_tilde_1a*gamma_tilde_1a*exp((-gamma_tilde_1a)*time)+k_tilde_3c*k_tilde_3a*APC*Va_Xa/(Va_Xa+1))-k_tilde_1b*Xa)-k_tilde_3a*Xa*Va, Rate Law: ((k_tilde_1a*gamma_tilde_1a*exp((-gamma_tilde_1a)*time)+k_tilde_3c*k_tilde_3a*APC*Va_Xa/(Va_Xa+1))-k_tilde_1b*Xa)-k_tilde_3a*Xa*Va
k_tilde_5a = 0.0011Reaction: PC = (-k_tilde_5a)*PC, Rate Law: (-k_tilde_5a)*PC
k_tilde_2am = 7.2; k_tilde_2b = 0.013; k_tilde_2a = 2.0Reaction: V = (-k_tilde_2a)*IIa*V/(V+k_tilde_2am*(1+Fibrinogen))-k_tilde_2a*k_tilde_2b*Xa*V/(V+1+II), Rate Law: (-k_tilde_2a)*IIa*V/(V+k_tilde_2am*(1+Fibrinogen))-k_tilde_2a*k_tilde_2b*Xa*V/(V+1+II)
k_tilde_4b = 530.0; k_tilde_4a = 0.12; q_tilde_4a = 0.004; k_tilde_4bm = 3.6Reaction: IIa = (k_tilde_4a*Xa_L*II/(V+1+II)+k_tilde_4a*k_tilde_4b*Va_Xa_L*II/(q_tilde_4a*(II+k_tilde_4bm)))-IIa, Rate Law: (k_tilde_4a*Xa_L*II/(V+1+II)+k_tilde_4a*k_tilde_4b*Va_Xa_L*II/(q_tilde_4a*(II+k_tilde_4bm)))-IIa
k_tilde_6 = 1500.0Reaction: Fibrin = k_tilde_6*Fibrinogen, Rate Law: k_tilde_6*Fibrinogen
k_tilde_4b = 530.0; q_tilde_4a = 0.004; k_tilde_4bm = 3.6Reaction: II = (-q_tilde_4a)*Xa_L*II/(V+1+II)-k_tilde_4b*Va_Xa_L*II/(II+k_tilde_4bm), Rate Law: (-q_tilde_4a)*Xa_L*II/(V+1+II)-k_tilde_4b*Va_Xa_L*II/(II+k_tilde_4bm)
k_tilde_b = 5.0E-4; l_tilde_b = 0.05Reaction: Va_Xa_L = 0.5*((k_tilde_b+l_tilde_b+Va_Xa)-((k_tilde_b+l_tilde_b+Va_Xa)^2-4*l_tilde_b*Va_Xa)^(0.5)), Rate Law: missing
k_tilde_x = 385.0; l_tilde_x = 7.69Reaction: Xa_L = 0.5*((k_tilde_x+l_tilde_x+Xa)-((k_tilde_x+l_tilde_x+Xa)^2-4*l_tilde_x*Xa)^(0.5)), Rate Law: missing
k_tilde_3c = 1.0Reaction: Va_inactive = APC*Va/(Va+1)+k_tilde_3c*APC*Va_Xa/(Va_Xa+1), Rate Law: APC*Va/(Va+1)+k_tilde_3c*APC*Va_Xa/(Va_Xa+1)
q_tilde_3a = 1.0; k_tilde_2am = 7.2; k_tilde_2b = 0.013; k_tilde_3b = 0.038Reaction: Va = ((IIa*V/(V+k_tilde_2am*(1+Fibrinogen))+k_tilde_2b*Xa*V/(V+1+II)+k_tilde_3b/q_tilde_3a*Va_Xa)-APC*Va/(Va+1))-Xa*Va, Rate Law: ((IIa*V/(V+k_tilde_2am*(1+Fibrinogen))+k_tilde_2b*Xa*V/(V+1+II)+k_tilde_3b/q_tilde_3a*Va_Xa)-APC*Va/(Va+1))-Xa*Va
k_tilde_5a = 0.0011; k_tilde_5b = 0.27Reaction: APC = k_tilde_5a*PC-k_tilde_5b*APC, Rate Law: k_tilde_5a*PC-k_tilde_5b*APC
k_tilde_5b = 0.27Reaction: APC_inactive = k_tilde_5b*APC, Rate Law: k_tilde_5b*APC

States:

NameDescription
Va Xa LVa:Xa:L
IIa ATIIIIIa:ATIII
Xa ATIIIXa:ATIII
FibrinogenFibrinogen
FibrinFibrin
APC inactiveAPC_inactive
VV
XaXa
VaVa
IIaIIa
Xa LXa:L
Va XaVa:Xa
APCAPC
Va inactiveVa_inactive
PCPC
IIII

Dupeux2011_ABAreceptor_Dimer: MODEL1202030000v0.0.1

This model is from the article: A thermodynamic switch modulates abscisic acid receptor sensitivity. Dupeux F, Santi…

Details

Abscisic acid (ABA) is a key hormone regulating plant growth, development and the response to biotic and abiotic stress. ABA binding to pyrabactin resistance (PYR)/PYR1-like (PYL)/Regulatory Component of Abscisic acid Receptor (RCAR) intracellular receptors promotes the formation of stable complexes with certain protein phosphatases type 2C (PP2Cs), leading to the activation of ABA signalling. The PYR/PYL/RCAR family contains 14 genes in Arabidopsis and is currently the largest plant hormone receptor family known; however, it is unclear what functional differentiation exists among receptors. Here, we identify two distinct classes of receptors, dimeric and monomeric, with different intrinsic affinities for ABA and whose differential properties are determined by the oligomeric state of their apo forms. Moreover, we find a residue in PYR1, H60, that is variable between family members and plays a key role in determining oligomeric state. In silico modelling of the ABA activation pathway reveals that monomeric receptors have a competitive advantage for binding to ABA and PP2Cs. This work illustrates how receptor oligomerization can modulate hormonal responses and more generally, the sensitivity of a ligand-dependent signalling system. link: http://identifiers.org/pubmed/21847091

Dupeux2011_ABAreceptor_Monomer: MODEL1202030001v0.0.1

This model is from the article: A thermodynamic switch modulates abscisic acid receptor sensitivit y. Dupeux F, Sant…

Details

Abscisic acid (ABA) is a key hormone regulating plant growth, development and the response to biotic and abiotic stress. ABA binding to pyrabactin resistance (PYR)/PYR1-like (PYL)/Regulatory Component of Abscisic acid Receptor (RCAR) intracellular receptors promotes the formation of stable complexes with certain protein phosphatases type 2C (PP2Cs), leading to the activation of ABA signalling. The PYR/PYL/RCAR family contains 14 genes in Arabidopsis and is currently the largest plant hormone receptor family known; however, it is unclear what functional differentiation exists among receptors. Here, we identify two distinct classes of receptors, dimeric and monomeric, with different intrinsic affinities for ABA and whose differential properties are determined by the oligomeric state of their apo forms. Moreover, we find a residue in PYR1, H60, that is variable between family members and plays a key role in determining oligomeric state. In silico modelling of the ABA activation pathway reveals that monomeric receptors have a competitive advantage for binding to ABA and PP2Cs. This work illustrates how receptor oligomerization can modulate hormonal responses and more generally, the sensitivity of a ligand-dependent signalling system. link: http://identifiers.org/pubmed/21847091

Dupont1991_CaOscillation: BIOMD0000000117v0.0.1

This model is according to the paper *Signal-induced Ca2+ oscillations: Properties of a model based on Ca2+-induced Ca2+…

Details

We consider a simple, minimal model for signal-induced Ca2+ oscillations based on Ca(2+)-induced Ca2+ release. The model takes into account the existence of two pools of intracellular Ca2+, namely, one sensitive to inositol 1,4,5 trisphosphate (InsP3) whose synthesis is elicited by the stimulus, and one insensitive to InsP3. The discharge of the latter pool into the cytosol is activated by cytosolic Ca2+. Oscillations in cytosolic Ca2+ arise in this model either spontaneously or in an appropriate range of external stimulation; these oscillations do not require the concomitant, periodic variation of InsP3. The following properties of the model are reviewed and compared with experimental observations: (a) Control of the frequency of Ca2+ oscillations by the external stimulus or extracellular Ca2+; (b) correlation of latency with period of Ca2+ oscillations obtained at different levels of stimulation; (c) effect of a transient increase in InsP3; (d) phase shift and transient suppression of Ca2+ oscillations by Ca2+ pulses, and (e) propagation of Ca2+ waves. It is shown that on all these counts the model provides a simple, unified explanation for a number of experimental observations in a variety of cell types. The model based on Ca(2+)-induced Ca2+ release can be extended to incorporate variations in the level of InsP3 as well as desensitization of the InsP3 receptor; besides accounting for the phenomena described by the minimal model, the extended model might also account for the occurrence of complex Ca2+ oscillations. link: http://identifiers.org/pubmed/1647878

Parameters:

NameDescription
v0 = 1.0Reaction: => z, Rate Law: v0*Cytosol
K2 = 1.0; n = 2.0; VM2 = 65.0Reaction: z => y, Rate Law: intracellular_Ca_storepool*VM2*z^n/(K2^n+z^n)
kf = 1.0Reaction: y => z, Rate Law: kf*y*Cytosol
m = 2.0; KR = 2.0; VM3 = 500.0; p = 4.0; KA = 0.9Reaction: y => z, Rate Law: Cytosol*VM3*y^m/(KR^m+y^m)*z^p/(KA^p+z^p)
v1 = 7.3; beta = 0.0Reaction: => z, Rate Law: v1*beta*Cytosol
k = 10.0Reaction: z =>, Rate Law: k*z*extracellular

States:

NameDescription
z[calcium(2+); Calcium cation]
y[calcium(2+); Calcium cation]

Dupont1992_Ca_dpt_protein_phospho: BIOMD0000000113v0.0.1

Model reproduces Fig 4 of the paper. For fraction of phosphorylated protein, W_star, the model reproduces panel b in the…

Details

Given the ubiquitous nature of signal-induced Ca2+ oscillations, the question arises as to how cellular responses are affected by repetitive Ca2+ spikes. Among these responses, we focus on those involving protein phosphorylation. We examine, by numerical simulations of a theoretical model, the situation where a protein is phosphorylated by a Ca(2+)-activated kinase and dephosphorylated by a phosphatase. This reversible phosphorylation system is coupled to a mechanism generating cytosolic Ca2+ oscillations; for definiteness, this oscillatory mechanism is based on the process of Ca(2+)-induced Ca2+ release. The analysis shows that the average fraction of phosphorylated protein increases with the frequency of repetitive Ca2+ spikes; the latter frequency generally rises with the extent of external stimulation. Protein phosphorylation therefore provides a mechanism for the encoding of the external stimulation in terms of the frequency of signal-induced Ca2+ oscillations. Such a frequency encoding requires precise kinetic conditions on the Michaelis-Menten constants of the kinase and phosphatase, their maximal rates, and the degree of cooperativity in kinase activation by Ca2+. In particular, the most efficient encoding of Ca2+ oscillations based on protein phosphorylation occurs in conditions of zero-order ultrasensitivity, when the kinase and phosphatase are saturated by their protein substrate. The kinetic analysis uncovers a wide variety of temporal patterns of phosphorylation that could be driven by signal-induced Ca2+ oscillations. link: http://identifiers.org/pubmed/1316185

Parameters:

NameDescription
v0 = 1.0Reaction: => Z, Rate Law: cytosol*v0
K_A = 0.9; m = 2.0; Vm3 = 500.0; p = 4.0; Kr = 2.0Reaction: Y => Z, Rate Law: store*Vm3*Y^m*Z^p/((Kr^m+Y^m)*(K_A^p+Z^p))
K1 = 0.01; K2 = 0.01; vk = NaN; vp = 2.5Reaction: => W_star; Wt, Rate Law: cytosol*vp/Wt*(vk/vp*(1-W_star)/((K1+1)-W_star)-W_star/(K2+W_star))
kf = 1.0Reaction: Y => Z, Rate Law: store*kf*Y
Vm2 = 65.0; n = 2.0; Kp = 1.0Reaction: Z => Y, Rate Law: cytosol*Vm2*Z^n/(Kp^n+Z^n)
k = 10.0Reaction: Z =>, Rate Law: cytosol*k*Z
v1_beta = 2.7Reaction: => Z, Rate Law: cytosol*v1_beta

States:

NameDescription
Z[calcium(2+); Calcium cation]
Y[calcium(2+); Calcium cation]
W starPhosphorylated protein

Durot2008_A.Baylyi_ADP1Metabolism: MODEL1949107276v0.0.1

This is the constraint based model from: **Iterative reconstruction of a global metabolic model of Acinetobacter bayly…

Details

BACKGROUND: Genome-scale metabolic models are powerful tools to study global properties of metabolic networks. They provide a way to integrate various types of biological information in a single framework, providing a structured representation of available knowledge on the metabolism of the respective species. RESULTS: We reconstructed a constraint-based metabolic model of Acinetobacter baylyi ADP1, a soil bacterium of interest for environmental and biotechnological applications with large-spectrum biodegradation capabilities. Following initial reconstruction from genome annotation and the literature, we iteratively refined the model by comparing its predictions with the results of large-scale experiments: (1) high-throughput growth phenotypes of the wild-type strain on 190 distinct environments, (2) genome-wide gene essentialities from a knockout mutant library, and (3) large-scale growth phenotypes of all mutant strains on 8 minimal media. Out of 1412 predictions, 1262 were initially consistent with our experimental observations. Inconsistencies were systematically examined, leading in 65 cases to model corrections. The predictions of the final version of the model, which included three rounds of refinements, are consistent with the experimental results for (1) 91% of the wild-type growth phenotypes, (2) 94% of the gene essentiality results, and (3) 94% of the mutant growth phenotypes. To facilitate the exploitation of the metabolic model, we provide a web interface allowing online predictions and visualization of results on metabolic maps. CONCLUSION: The iterative reconstruction procedure led to significant model improvements, showing that genome-wide mutant phenotypes on several media can significantly facilitate the transition from genome annotation to a high-quality model. link: http://identifiers.org/pubmed/18840283

Dutta-Roy2015 - Opening of the multiple AMPA receptor conductance states: BIOMD0000000569v0.0.1

Dutta-Roy2015 - Opening of the multiple AMPA receptor conductance statesThis model is described in the article: [Ligand…

Details

Modulation of the properties of AMPA receptors at the post-synaptic membrane is one of the main suggested mechanisms underlying fast synaptic transmission in the central nervous system of vertebrates. Electrophysiological recordings of single channels stimulated with agonists showed that both recombinant and native AMPA receptors visit multiple conductance states in an agonist concentration dependent manner. We propose an allosteric model of the multiple conductance states based on concerted conformational transitions of the four subunits, as an iris diaphragm. Our model predicts that the thermodynamic behaviour of the conductance states upon full and partial agonist stimulations can be described with increased affinity of receptors as they progress to higher conductance states. The model also predicts the existence of AMPA receptors in non-liganded conductive substates. However, the probability of spontaneous openings decreases with increasing conductances. Finally, we predict that the large conductance states are stabilized within the rise phase of a whole-cell EPSC in glutamatergic hippocampal neurons. Our model provides a mechanistic link between ligand concentration and conductance states that can explain thermodynamic and kinetic features of AMPA receptor gating. link: http://identifiers.org/pubmed/25629405

Parameters:

NameDescription
LMk3 = 2630.59204225087Reaction: L3 => M3; L3, Rate Law: synapse*LMk3*L3
MSk0 = 38212.5990892225Reaction: M0 => S0; M0, Rate Law: synapse*MSk0*M0
SBk4 = 860.0Reaction: S4 => B4; S4, Rate Law: synapse*SBk4*S4
MLk1 = 906.240862022895Reaction: M1 => L1; M1, Rate Law: synapse*MLk1*M1
Lkoff = 1.149Reaction: L1 => L0 + glu; L1, Rate Law: synapse*Lkoff*L1
Skon = 5000000.0Reaction: S3 + glu => S4; S3, glu, Rate Law: synapse*Skon*S3*glu
SMk3 = 4963.86945839634Reaction: S3 => M3; S3, Rate Law: synapse*SMk3*S3
SMk2 = 1232.0Reaction: S2 => M2; S2, Rate Law: synapse*SMk2*S2
MSk4 = 145.0Reaction: M4 => S4; M4, Rate Law: synapse*MSk4*M4
MLk0 = 238.05Reaction: M0 => L0; M0, Rate Law: synapse*MLk0*M0
LMk2 = 10014.4927536232Reaction: L2 => M2; L2, Rate Law: synapse*LMk2*L2
Mkon = 5000000.0Reaction: M1 + glu => M2; M1, glu, Rate Law: synapse*3*Mkon*M1*glu
MLk2 = 3450.0Reaction: M2 => L2; M2, Rate Law: synapse*MLk2*M2
MLk3 = 13133.9255365637Reaction: M3 => L3; M3, Rate Law: synapse*MLk3*M3
MSk3 = 584.221648918401Reaction: M3 => S3; M3, Rate Law: synapse*MSk3*M3
Bkoff = 4495.0Reaction: B4 => B3 + glu; B4, Rate Law: synapse*4*Bkoff*B4
LMk0 = 145137.576139466Reaction: L0 => M0; L0, Rate Law: synapse*LMk0*L0
BSk4 = 300000.0Reaction: B4 => S4; B4, Rate Law: synapse*BSk4*B4
SMk4 = 20000.0Reaction: S4 => M4; S4, Rate Law: synapse*SMk4*S4
LMk4 = 691.0Reaction: L4 => M4; L4, Rate Law: synapse*LMk4*L4
SMk0 = 75.8912Reaction: S0 => M0; S0, Rate Law: synapse*SMk0*S0
Mkoff = 16.65Reaction: M1 => M0 + glu; M1, Rate Law: synapse*Mkoff*M1
Skoff = 270.4Reaction: S4 => S3 + glu; S4, Rate Law: synapse*4*Skoff*S4
LMk1 = 38124.5223514619Reaction: L1 => M1; L1, Rate Law: synapse*LMk1*L1
MSk2 = 2353.8961038961Reaction: M2 => S2; M2, Rate Law: synapse*MSk2*M2
SMk1 = 305.774358637215Reaction: S1 => M1; S1, Rate Law: synapse*SMk1*S1
Lkon = 5000000.0Reaction: L0 + glu => L1; L0, glu, Rate Law: synapse*4*Lkon*L0*glu
MSk1 = 9484.11767724676Reaction: M1 => S1; M1, Rate Law: synapse*MSk1*M1
MLk4 = 50000.0Reaction: M4 => L4; M4, Rate Law: synapse*MLk4*M4
Bkon = 5000000.0Reaction: B0 + glu => B1; B0, glu, Rate Law: synapse*4*Bkon*B0*glu

States:

NameDescription
L1L1
M0M0
M3M3
S3S3
M2M2
L4L4
B4B4
L2L2
S4S4
L3L3
M1M1
M4M4
glu[glutamic acid]
L0L0

Dwivedi2014 - Crohns IL6 Disease model - Anti-IL6 Antibody: BIOMD0000000535v0.0.1

Dwivedi2014 - Crohns IL6 Disease model - Anti-IL6 AntibodyThis model is comprised of four models: [[BIOMD0000000534]](h…

Details

In this study, we have developed a multiscale systems model of interleukin (IL)-6-mediated immune regulation in Crohn's disease, by integrating intracellular signaling with organ-level dynamics of pharmacological markers underlying the disease. This model was linked to a general pharmacokinetic model for therapeutic monoclonal antibodies and used to comparatively study various biotherapeutic strategies targeting IL-6-mediated signaling in Crohn's disease. Our work illustrates techniques to develop mechanistic models of disease biology to study drug-system interaction. Despite a sparse training data set, predictions of the model were qualitatively validated by clinical biomarker data from a pilot trial with tocilizumab. Model-based analysis suggests that strategies targeting IL-6, IL-6Rα, or the IL-6/sIL-6Rα complex are less effective at suppressing pharmacological markers of Crohn's than dual targeting the IL-6/sIL-6Rα complex in addition to IL-6 or IL-6Rα. The potential value of multiscale system pharmacology modeling in drug discovery and development is also discussed.CPT: Pharmacometrics & Systems Pharmacology (2014) 3, e89; doi:10.1038/psp.2013.64; advance online publication 8 January 2014. link: http://identifiers.org/pubmed/24402116

Parameters:

NameDescription
mw862f1480_c60c_4863_a565_b2c1c77e238e = 0.5Reaction: mwd5313618_89eb_4c8c_bc82_66f10f966349 => mw36ea78c1_ed71_4def_96d3_857a442d7195; mwd5313618_89eb_4c8c_bc82_66f10f966349, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mw862f1480_c60c_4863_a565_b2c1c77e238e*mwd5313618_89eb_4c8c_bc82_66f10f966349/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRint = 1.96Reaction: mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a => ; mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRint*mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRsynth = 0.0685Reaction: => mw10315fa3_6f13_4618_bda8_a8694bd3c374, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRsynth/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kCRPDecay = 0.36Reaction: mw114aa90f_5f5b_4fe8_9406_361c8489b6a1 => ; mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*kCRPDecay*mw114aa90f_5f5b_4fe8_9406_361c8489b6a1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw9f83bdd3_3aa1_47ff_abd6_54e5ce60704a = 0.0104166666666667; mwa071fdbe_d498_4620_a7a4_940aa31c8161 = 0.0208333333333333Reaction: mwf345ed7a_0622_403c_b816_c8749a2c9ded => mwf7796221_1fea_4274_a93e_c00adbf5778c; mwf345ed7a_0622_403c_b816_c8749a2c9ded, mwf7796221_1fea_4274_a93e_c00adbf5778c, Rate Law: mw9f83bdd3_3aa1_47ff_abd6_54e5ce60704a*mwf345ed7a_0622_403c_b816_c8749a2c9ded-mwa071fdbe_d498_4620_a7a4_940aa31c8161*mwf7796221_1fea_4274_a93e_c00adbf5778c
kIL6Decay = 34.82Reaction: mwf626e95e_543f_41e4_aad4_c6bf60ab345b => ; mwf626e95e_543f_41e4_aad4_c6bf60ab345b, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*kIL6Decay*mwf626e95e_543f_41e4_aad4_c6bf60ab345b/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw08950572_81b0_4570_b2e4_b9c3462c1425 = 10.0; mw92d854a7_8aaf_458e_b5e2_20a63ce9b654 = 330.0Reaction: mw0083d743_836f_4238_a17f_4602193d5bc0 = mw92d854a7_8aaf_458e_b5e2_20a63ce9b654*mw48867e93_f170_44e8_ac7a_185b23e1bf3b/(mw08950572_81b0_4570_b2e4_b9c3462c1425+mw48867e93_f170_44e8_ac7a_185b23e1bf3b), Rate Law: missing
mw06241335_b5f2_47ed_bdcc_ef77b68a2b98 = 1.0Reaction: mw2c9b0499_3325_4394_8af3_bbf653a944a0 => ; mw2c9b0499_3325_4394_8af3_bbf653a944a0, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mw06241335_b5f2_47ed_bdcc_ef77b68a2b98*mw2c9b0499_3325_4394_8af3_bbf653a944a0/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwfd291862_195f_4979_94b5_b4e5ae1b7d52 = 5.34; mwd36b0261_2480_4cab_9222_2cf8fb0e65dc = 0.62Reaction: mw48867e93_f170_44e8_ac7a_185b23e1bf3b => mw2b255f94_8018_4b99_bde8_918eeac45446; mw48867e93_f170_44e8_ac7a_185b23e1bf3b, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwd36b0261_2480_4cab_9222_2cf8fb0e65dc*mw48867e93_f170_44e8_ac7a_185b23e1bf3b/(mwfd291862_195f_4979_94b5_b4e5ae1b7d52+mw48867e93_f170_44e8_ac7a_185b23e1bf3b)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwe8fc1900_f07d_468b_b5c8_15400a583c3d = 219.0; mw9442cd0e_4d7c_4ba6_a695_f84919bdf569 = 145.0Reaction: mw2b255f94_8018_4b99_bde8_918eeac45446 + mw6cce2109_0e32_4dd9_98ec_41173e8ef07d => mw48867e93_f170_44e8_ac7a_185b23e1bf3b + mw6cce2109_0e32_4dd9_98ec_41173e8ef07d; mw2b255f94_8018_4b99_bde8_918eeac45446, mw6cce2109_0e32_4dd9_98ec_41173e8ef07d, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mw9442cd0e_4d7c_4ba6_a695_f84919bdf569*mw6cce2109_0e32_4dd9_98ec_41173e8ef07d*mw2b255f94_8018_4b99_bde8_918eeac45446/(mwe8fc1900_f07d_468b_b5c8_15400a583c3d+mw2b255f94_8018_4b99_bde8_918eeac45446)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwf44f7f27_5bb1_4c7f_8964_560fa5e1743a = 0.01Reaction: mw6cce2109_0e32_4dd9_98ec_41173e8ef07d => ; mw6cce2109_0e32_4dd9_98ec_41173e8ef07d, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwf44f7f27_5bb1_4c7f_8964_560fa5e1743a*mw6cce2109_0e32_4dd9_98ec_41173e8ef07d/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
kIL6Synth = 0.0063Reaction: => mwf626e95e_543f_41e4_aad4_c6bf60ab345b, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*kIL6Synth/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw43ccad8c_cabf_4eaf_90d5_e06ae43be2cb = 0.0208333333333333; mw640ca705_e089_4c64_a5f4_9562317e8c76 = 0.0208333333333333Reaction: mw1da111f2_a036_4392_8512_015005bdcbb7 => mwf405687b_7401_44ec_a0d6_4a2b35c13e8a; mw1da111f2_a036_4392_8512_015005bdcbb7, mwf405687b_7401_44ec_a0d6_4a2b35c13e8a, Rate Law: mw640ca705_e089_4c64_a5f4_9562317e8c76*mw1da111f2_a036_4392_8512_015005bdcbb7-mw43ccad8c_cabf_4eaf_90d5_e06ae43be2cb*mwf405687b_7401_44ec_a0d6_4a2b35c13e8a
kgp130Off = 1.026; kgp130On = 20.52Reaction: mw03db56ac_8dc6_4931_ae82_fef706d2ee3d + mwbbbce920_e8dd_4320_9386_fc94bfb2fc99 => mw810ff751_fa4e_4143_bd50_169b3e325e1e; mw03db56ac_8dc6_4931_ae82_fef706d2ee3d, mw810ff751_fa4e_4143_bd50_169b3e325e1e, mwbbbce920_e8dd_4320_9386_fc94bfb2fc99, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*(kgp130On*mw03db56ac_8dc6_4931_ae82_fef706d2ee3d*mwbbbce920_e8dd_4320_9386_fc94bfb2fc99-kgp130Off*mw810ff751_fa4e_4143_bd50_169b3e325e1e)/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw88a75379_f9a1_4acc_baeb_94c32bb736a5 = 0.3Reaction: mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc => ; mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw88a75379_f9a1_4acc_baeb_94c32bb736a5*mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwce10678d_8197_408c_ad47_1daec8104cd8 = 0.8473; mwc67e1333_079a_4bea_9b4f_0a1b15ddd7bb = 1.2125Reaction: mwf626e95e_543f_41e4_aad4_c6bf60ab345b => mw2c9b0499_3325_4394_8af3_bbf653a944a0; mwf626e95e_543f_41e4_aad4_c6bf60ab345b, mw2c9b0499_3325_4394_8af3_bbf653a944a0, Rate Law: mwc67e1333_079a_4bea_9b4f_0a1b15ddd7bb*mwf626e95e_543f_41e4_aad4_c6bf60ab345b-mwce10678d_8197_408c_ad47_1daec8104cd8*mw2c9b0499_3325_4394_8af3_bbf653a944a0
mw1667a8e0_9d20_4e59_ba51_596148aba787 = 0.525; mwfcf06900_5f2f_4bb3_bb1f_12023612b8a8 = 155.3Reaction: mw0eb6c959_d408_45a0_a450_928b8c5876bb => mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a; mw0eb6c959_d408_45a0_a450_928b8c5876bb, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mw1667a8e0_9d20_4e59_ba51_596148aba787*mw0eb6c959_d408_45a0_a450_928b8c5876bb/(mwfcf06900_5f2f_4bb3_bb1f_12023612b8a8+mw0eb6c959_d408_45a0_a450_928b8c5876bb)/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mwbd1d5bc3_d4b9_4aec_9b86_6f776da20a30 = 0.0018Reaction: mw1da111f2_a036_4392_8512_015005bdcbb7 => ; mw1da111f2_a036_4392_8512_015005bdcbb7, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mwbd1d5bc3_d4b9_4aec_9b86_6f776da20a30*mw1da111f2_a036_4392_8512_015005bdcbb7/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
kRAct = 155.0Reaction: mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a => mw0eb6c959_d408_45a0_a450_928b8c5876bb; mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRAct*mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mwa8d72918_f6c2_4d81_bf3b_fc2b464d5e69 = 0.036Reaction: => mw2c9b0499_3325_4394_8af3_bbf653a944a0, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwa8d72918_f6c2_4d81_bf3b_fc2b464d5e69/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwbcb5a310_9b67_405e_89ec_43d25e8cc93d = 1.0Reaction: mwbbbce920_e8dd_4320_9386_fc94bfb2fc99 => ; mwbbbce920_e8dd_4320_9386_fc94bfb2fc99, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mwbcb5a310_9b67_405e_89ec_43d25e8cc93d*mwbbbce920_e8dd_4320_9386_fc94bfb2fc99/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwc4c58db7_5535_4590_aaa5_bbc8ed53cdab = 0.1Reaction: => mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mwc4c58db7_5535_4590_aaa5_bbc8ed53cdab/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwa09d6284_843e_404e_abbb_052fbb535197 = 1000.0; mw1c4bc9c3_52ad_4ef7_bf7f_97b0e2101ead = 2.5Reaction: mwf626e95e_543f_41e4_aad4_c6bf60ab345b + mwf345ed7a_0622_403c_b816_c8749a2c9ded => mw1da111f2_a036_4392_8512_015005bdcbb7; mw1da111f2_a036_4392_8512_015005bdcbb7, mwf345ed7a_0622_403c_b816_c8749a2c9ded, mwf626e95e_543f_41e4_aad4_c6bf60ab345b, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*(mwa09d6284_843e_404e_abbb_052fbb535197*mwf626e95e_543f_41e4_aad4_c6bf60ab345b*mwf345ed7a_0622_403c_b816_c8749a2c9ded-mw1c4bc9c3_52ad_4ef7_bf7f_97b0e2101ead*mw1da111f2_a036_4392_8512_015005bdcbb7)/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw65c85954_5ca0_4df2_9e22_ff2aa3fbe3f1 = 0.42Reaction: => mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw65c85954_5ca0_4df2_9e22_ff2aa3fbe3f1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwf67caf9d_2f4b_4986_abf2_e6090bbb72ce = 3.47222222222222E-4; mw4aea26f6_8860_414c_97f5_40d325196f2e = 0.00173611111111111Reaction: mwf345ed7a_0622_403c_b816_c8749a2c9ded => mwbc2f5464_81e5_43fd_8b39_f5a2756af72f; mwf345ed7a_0622_403c_b816_c8749a2c9ded, mwbc2f5464_81e5_43fd_8b39_f5a2756af72f, Rate Law: mwf67caf9d_2f4b_4986_abf2_e6090bbb72ce*mwf345ed7a_0622_403c_b816_c8749a2c9ded-mw4aea26f6_8860_414c_97f5_40d325196f2e*mwbc2f5464_81e5_43fd_8b39_f5a2756af72f
kRdeg = 0.1561Reaction: mw80848184_e2dd_47ce_86d7_7a21479342bd => ; mw80848184_e2dd_47ce_86d7_7a21479342bd, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRdeg*mw80848184_e2dd_47ce_86d7_7a21479342bd/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRLOff = 1.92; kRLOn = 0.384Reaction: mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc + mwf626e95e_543f_41e4_aad4_c6bf60ab345b => mw03db56ac_8dc6_4931_ae82_fef706d2ee3d; mw03db56ac_8dc6_4931_ae82_fef706d2ee3d, mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc, mwf626e95e_543f_41e4_aad4_c6bf60ab345b, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*(kRLOn*mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc*mwf626e95e_543f_41e4_aad4_c6bf60ab345b-kRLOff*mw03db56ac_8dc6_4931_ae82_fef706d2ee3d)/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw5832a2dc_ee18_44df_aa59_ccb21cb74df2 = 0.0054Reaction: mw114aa90f_5f5b_4fe8_9406_361c8489b6a1 => mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc + mw114aa90f_5f5b_4fe8_9406_361c8489b6a1; mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw5832a2dc_ee18_44df_aa59_ccb21cb74df2*mw114aa90f_5f5b_4fe8_9406_361c8489b6a1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
Metabolite_3 = 221.06367608557Reaction: CRP_Suppression___ = (mw114aa90f_5f5b_4fe8_9406_361c8489b6a1-Metabolite_3)/(Metabolite_3/(-100)), Rate Law: missing
mw1f41474c_c399_4a60_a53a_9926dd092e8d = 3.9Reaction: => mwbbbce920_e8dd_4320_9386_fc94bfb2fc99, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw1f41474c_c399_4a60_a53a_9926dd092e8d/mw53ffe9e6_beef_45c4_90a5_a79197ed506e

States:

NameDescription
CRP Suppression[C-reactive protein]
mw810ff751 fa4e 4143 bd50 169b3e325e1e[soluble in; Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta]
mw7d86cc23 a1af 44c3 bdb9 71e9b1bb2a83[Interleukin-6 receptor subunit alpha; Interleukin-6]
mwa2d8dd1c bb9a 4552 8738 e24671651c1d[soluble in; Interleukin-6 receptor subunit alpha; Interleukin-6; Immunoglobulin]
mwd2d9d93a 3bd1 4f17 bac1 baba9ef2d55a[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta]
mw03db56ac 8dc6 4931 ae82 fef706d2ee3d[soluble in; Interleukin-6 receptor subunit alpha; Interleukin-6]
mw0083d743 836f 4238 a17f 4602193d5bc0[CCO:U0000003]
mw0adf3eb4 a196 4c48 b10d 4e9e9faaf9e1[Interleukin-6]
mw48867e93 f170 44e8 ac7a 185b23e1bf3b[signal transducer and activator of transcription 3; phosphorylation]
mw6cce2109 0e32 4dd9 98ec 41173e8ef07d[SBO:0000286; active; Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta]
mwd5313618 89eb 4c8c bc82 66f10f966349[C-reactive protein]
mw30ae63db 6cd3 4b6f 93ad 3350cd360bcc[soluble in; Interleukin-6 receptor subunit alpha]
mwf345ed7a 0622 403c b816 c8749a2c9ded[Immunoglobulin; pharmaceutical]
mw147d30ec 478e 4090 b496 128a131d29eb[soluble in; interleukin-6 receptor subunit beta]
mwf7796221 1fea 4274 a93e c00adbf5778c[Immunoglobulin]
mw1da111f2 a036 4392 8512 015005bdcbb7[Interleukin-6; Immunoglobulin]
mwd65b5b39 dc1b 4e77 a999 67277a880e5e[soluble in; interleukin-6 receptor subunit beta]
mw36ea78c1 ed71 4def 96d3 857a442d7195[C-reactive protein]
mwbbbce920 e8dd 4320 9386 fc94bfb2fc99[soluble in; interleukin-6 receptor subunit beta]
mw4638f126 8cb8 4021 ab41 6ae195743ba0[Interleukin-6 receptor subunit alpha; Interleukin-6; soluble in]
mw10315fa3 6f13 4618 bda8 a8694bd3c374[Interleukin-6 receptor subunit alpha]
mw0eb6c959 d408 45a0 a450 928b8c5876bb[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta; SBO:0000286; active]
mwd31f52cc 04e7 40e0 885f c7b2d9e62215[Interleukin-6 receptor subunit alpha; soluble in]
mw80848184 e2dd 47ce 86d7 7a21479342bd[interleukin-6 receptor subunit beta]
mw2e464cf3 a09c 4b7c 9f3c 06720016a48e[soluble in; Interleukin-6 receptor subunit alpha]
mw2b255f94 8018 4b99 bde8 918eeac45446[signal transducer and activator of transcription 3]
mw8c9107e6 f51d 442d b2dc 2bfdbb8482ca[interleukin-6 receptor subunit beta]
mw114aa90f 5f5b 4fe8 9406 361c8489b6a1[C-reactive protein]
mw824bc3d4 1ac3 4912 9b51 8f14ff1c96b9[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta]
mw39c2e431 fdc3 4964 be29 6ca856620b1b[signal transducer and activator of transcription 3; phosphorylation]
mwf626e95e 543f 41e4 aad4 c6bf60ab345b[Interleukin-6]
mw2c9b0499 3325 4394 8af3 bbf653a944a0[Interleukin-6]
mw6335d5d7 c7b0 4bc0 b883 f7ee4915c2c3[soluble in; Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta]

Dwivedi2014 - Crohns IL6 Disease model - Anti-IL6R Antibody: BIOMD0000000537v0.0.1

Dwivedi2014 - Crohns IL6 Disease model - Anti-IL6R AntibodyThis model is comprised of four models: [[BIOMD0000000534]](…

Details

In this study, we have developed a multiscale systems model of interleukin (IL)-6-mediated immune regulation in Crohn's disease, by integrating intracellular signaling with organ-level dynamics of pharmacological markers underlying the disease. This model was linked to a general pharmacokinetic model for therapeutic monoclonal antibodies and used to comparatively study various biotherapeutic strategies targeting IL-6-mediated signaling in Crohn's disease. Our work illustrates techniques to develop mechanistic models of disease biology to study drug-system interaction. Despite a sparse training data set, predictions of the model were qualitatively validated by clinical biomarker data from a pilot trial with tocilizumab. Model-based analysis suggests that strategies targeting IL-6, IL-6Rα, or the IL-6/sIL-6Rα complex are less effective at suppressing pharmacological markers of Crohn's than dual targeting the IL-6/sIL-6Rα complex in addition to IL-6 or IL-6Rα. The potential value of multiscale system pharmacology modeling in drug discovery and development is also discussed.CPT: Pharmacometrics & Systems Pharmacology (2014) 3, e89; doi:10.1038/psp.2013.64; advance online publication 8 January 2014. link: http://identifiers.org/pubmed/24402116

Parameters:

NameDescription
mw862f1480_c60c_4863_a565_b2c1c77e238e = 0.5Reaction: mwd5313618_89eb_4c8c_bc82_66f10f966349 => mw36ea78c1_ed71_4def_96d3_857a442d7195; mwd5313618_89eb_4c8c_bc82_66f10f966349, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mw862f1480_c60c_4863_a565_b2c1c77e238e*mwd5313618_89eb_4c8c_bc82_66f10f966349/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRsynth = 0.0685Reaction: => mw8c9107e6_f51d_442d_b2dc_2bfdbb8482ca, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*kRsynth/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
kRint = 1.96Reaction: mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9 => ; mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*kRint*mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
kCRPDecay = 0.36Reaction: mw114aa90f_5f5b_4fe8_9406_361c8489b6a1 => ; mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*kCRPDecay*mw114aa90f_5f5b_4fe8_9406_361c8489b6a1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw9f83bdd3_3aa1_47ff_abd6_54e5ce60704a = 0.0104166666666667; mwa071fdbe_d498_4620_a7a4_940aa31c8161 = 0.0208333333333333Reaction: mwf345ed7a_0622_403c_b816_c8749a2c9ded => mwf7796221_1fea_4274_a93e_c00adbf5778c; mwf345ed7a_0622_403c_b816_c8749a2c9ded, mwf7796221_1fea_4274_a93e_c00adbf5778c, Rate Law: mw9f83bdd3_3aa1_47ff_abd6_54e5ce60704a*mwf345ed7a_0622_403c_b816_c8749a2c9ded-mwa071fdbe_d498_4620_a7a4_940aa31c8161*mwf7796221_1fea_4274_a93e_c00adbf5778c
mw08950572_81b0_4570_b2e4_b9c3462c1425 = 10.0; mw92d854a7_8aaf_458e_b5e2_20a63ce9b654 = 330.0Reaction: mw0083d743_836f_4238_a17f_4602193d5bc0 = mw92d854a7_8aaf_458e_b5e2_20a63ce9b654*mw48867e93_f170_44e8_ac7a_185b23e1bf3b/(mw08950572_81b0_4570_b2e4_b9c3462c1425+mw48867e93_f170_44e8_ac7a_185b23e1bf3b), Rate Law: missing
mw06241335_b5f2_47ed_bdcc_ef77b68a2b98 = 1.0Reaction: mw2c9b0499_3325_4394_8af3_bbf653a944a0 => ; mw2c9b0499_3325_4394_8af3_bbf653a944a0, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mw06241335_b5f2_47ed_bdcc_ef77b68a2b98*mw2c9b0499_3325_4394_8af3_bbf653a944a0/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
kIL6Decay = 34.82Reaction: mwf626e95e_543f_41e4_aad4_c6bf60ab345b => ; mwf626e95e_543f_41e4_aad4_c6bf60ab345b, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*kIL6Decay*mwf626e95e_543f_41e4_aad4_c6bf60ab345b/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwfd291862_195f_4979_94b5_b4e5ae1b7d52 = 5.34; mwd36b0261_2480_4cab_9222_2cf8fb0e65dc = 0.62Reaction: mw48867e93_f170_44e8_ac7a_185b23e1bf3b => mw2b255f94_8018_4b99_bde8_918eeac45446; mw48867e93_f170_44e8_ac7a_185b23e1bf3b, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwd36b0261_2480_4cab_9222_2cf8fb0e65dc*mw48867e93_f170_44e8_ac7a_185b23e1bf3b/(mwfd291862_195f_4979_94b5_b4e5ae1b7d52+mw48867e93_f170_44e8_ac7a_185b23e1bf3b)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwe8fc1900_f07d_468b_b5c8_15400a583c3d = 219.0; mw9442cd0e_4d7c_4ba6_a695_f84919bdf569 = 145.0Reaction: mw2b255f94_8018_4b99_bde8_918eeac45446 + mw6cce2109_0e32_4dd9_98ec_41173e8ef07d => mw48867e93_f170_44e8_ac7a_185b23e1bf3b + mw6cce2109_0e32_4dd9_98ec_41173e8ef07d; mw2b255f94_8018_4b99_bde8_918eeac45446, mw6cce2109_0e32_4dd9_98ec_41173e8ef07d, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mw9442cd0e_4d7c_4ba6_a695_f84919bdf569*mw6cce2109_0e32_4dd9_98ec_41173e8ef07d*mw2b255f94_8018_4b99_bde8_918eeac45446/(mwe8fc1900_f07d_468b_b5c8_15400a583c3d+mw2b255f94_8018_4b99_bde8_918eeac45446)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwf44f7f27_5bb1_4c7f_8964_560fa5e1743a = 0.01Reaction: mw6cce2109_0e32_4dd9_98ec_41173e8ef07d => ; mw6cce2109_0e32_4dd9_98ec_41173e8ef07d, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwf44f7f27_5bb1_4c7f_8964_560fa5e1743a*mw6cce2109_0e32_4dd9_98ec_41173e8ef07d/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mw43ccad8c_cabf_4eaf_90d5_e06ae43be2cb = 0.0208333333333333; mw640ca705_e089_4c64_a5f4_9562317e8c76 = 0.0208333333333333Reaction: mw1da111f2_a036_4392_8512_015005bdcbb7 => mwf405687b_7401_44ec_a0d6_4a2b35c13e8a; mw1da111f2_a036_4392_8512_015005bdcbb7, mwf405687b_7401_44ec_a0d6_4a2b35c13e8a, Rate Law: mw640ca705_e089_4c64_a5f4_9562317e8c76*mw1da111f2_a036_4392_8512_015005bdcbb7-mw43ccad8c_cabf_4eaf_90d5_e06ae43be2cb*mwf405687b_7401_44ec_a0d6_4a2b35c13e8a
kIL6Synth = 0.0063Reaction: => mwf626e95e_543f_41e4_aad4_c6bf60ab345b, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*kIL6Synth/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
kgp130Off = 1.026; kgp130On = 20.52Reaction: mw7becb5fe_8da8_4285_a821_0d77ad811b62 + mw8c9107e6_f51d_442d_b2dc_2bfdbb8482ca => mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9; mw7becb5fe_8da8_4285_a821_0d77ad811b62, mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9, mw8c9107e6_f51d_442d_b2dc_2bfdbb8482ca, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*(kgp130On*mw7becb5fe_8da8_4285_a821_0d77ad811b62*mw8c9107e6_f51d_442d_b2dc_2bfdbb8482ca-kgp130Off*mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwbd1d5bc3_d4b9_4aec_9b86_6f776da20a30 = 0.0018Reaction: mw772cbf20_3fc1_4800_ae59_77884f1ae333 => ; mw772cbf20_3fc1_4800_ae59_77884f1ae333, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mwbd1d5bc3_d4b9_4aec_9b86_6f776da20a30*mw772cbf20_3fc1_4800_ae59_77884f1ae333/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mwce10678d_8197_408c_ad47_1daec8104cd8 = 0.8473; mwc67e1333_079a_4bea_9b4f_0a1b15ddd7bb = 1.2125Reaction: mw9947742a_0e4b_4636_9a4b_b6eef2a8f6ac => mw2ba2b802_9f07_4f4d_94c6_24c8de1a95cf; mw9947742a_0e4b_4636_9a4b_b6eef2a8f6ac, mw2ba2b802_9f07_4f4d_94c6_24c8de1a95cf, Rate Law: mwc67e1333_079a_4bea_9b4f_0a1b15ddd7bb*mw9947742a_0e4b_4636_9a4b_b6eef2a8f6ac-mwce10678d_8197_408c_ad47_1daec8104cd8*mw2ba2b802_9f07_4f4d_94c6_24c8de1a95cf
kRAct = 155.0Reaction: mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9 => mw6cce2109_0e32_4dd9_98ec_41173e8ef07d; mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*kRAct*mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mw1667a8e0_9d20_4e59_ba51_596148aba787 = 0.525; mwfcf06900_5f2f_4bb3_bb1f_12023612b8a8 = 155.3Reaction: mw6cce2109_0e32_4dd9_98ec_41173e8ef07d => mw824bc3d4_1ac3_4912_9b51_8f14ff1c96b9; mw6cce2109_0e32_4dd9_98ec_41173e8ef07d, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mw1667a8e0_9d20_4e59_ba51_596148aba787*mw6cce2109_0e32_4dd9_98ec_41173e8ef07d/(mwfcf06900_5f2f_4bb3_bb1f_12023612b8a8+mw6cce2109_0e32_4dd9_98ec_41173e8ef07d)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwa8d72918_f6c2_4d81_bf3b_fc2b464d5e69 = 0.036Reaction: => mw2c9b0499_3325_4394_8af3_bbf653a944a0, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwa8d72918_f6c2_4d81_bf3b_fc2b464d5e69/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mw88a75379_f9a1_4acc_baeb_94c32bb736a5 = 0.3Reaction: mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc => ; mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw88a75379_f9a1_4acc_baeb_94c32bb736a5*mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwbcb5a310_9b67_405e_89ec_43d25e8cc93d = 1.0Reaction: mwbbbce920_e8dd_4320_9386_fc94bfb2fc99 => ; mwbbbce920_e8dd_4320_9386_fc94bfb2fc99, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mwbcb5a310_9b67_405e_89ec_43d25e8cc93d*mwbbbce920_e8dd_4320_9386_fc94bfb2fc99/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwa09d6284_843e_404e_abbb_052fbb535197 = 1000.0; mw1c4bc9c3_52ad_4ef7_bf7f_97b0e2101ead = 2.5Reaction: mw3667a5e1_02c9_44a0_acb4_b0431faa822d + mw2e464cf3_a09c_4b7c_9f3c_06720016a48e => mwf405687b_7401_44ec_a0d6_4a2b35c13e8a; mw2e464cf3_a09c_4b7c_9f3c_06720016a48e, mw3667a5e1_02c9_44a0_acb4_b0431faa822d, mwf405687b_7401_44ec_a0d6_4a2b35c13e8a, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*(mwa09d6284_843e_404e_abbb_052fbb535197*mw3667a5e1_02c9_44a0_acb4_b0431faa822d*mw2e464cf3_a09c_4b7c_9f3c_06720016a48e-mw1c4bc9c3_52ad_4ef7_bf7f_97b0e2101ead*mwf405687b_7401_44ec_a0d6_4a2b35c13e8a)/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mwc4c58db7_5535_4590_aaa5_bbc8ed53cdab = 0.1Reaction: => mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mwc4c58db7_5535_4590_aaa5_bbc8ed53cdab/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw65c85954_5ca0_4df2_9e22_ff2aa3fbe3f1 = 0.42Reaction: => mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw65c85954_5ca0_4df2_9e22_ff2aa3fbe3f1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwf67caf9d_2f4b_4986_abf2_e6090bbb72ce = 3.47222222222222E-4; mw4aea26f6_8860_414c_97f5_40d325196f2e = 0.00173611111111111Reaction: mwf345ed7a_0622_403c_b816_c8749a2c9ded => mwbc2f5464_81e5_43fd_8b39_f5a2756af72f; mwf345ed7a_0622_403c_b816_c8749a2c9ded, mwbc2f5464_81e5_43fd_8b39_f5a2756af72f, Rate Law: mwf67caf9d_2f4b_4986_abf2_e6090bbb72ce*mwf345ed7a_0622_403c_b816_c8749a2c9ded-mw4aea26f6_8860_414c_97f5_40d325196f2e*mwbc2f5464_81e5_43fd_8b39_f5a2756af72f
kRdeg = 0.1561Reaction: mw8c9107e6_f51d_442d_b2dc_2bfdbb8482ca => ; mw8c9107e6_f51d_442d_b2dc_2bfdbb8482ca, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*kRdeg*mw8c9107e6_f51d_442d_b2dc_2bfdbb8482ca/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
kRLOff = 1.92; kRLOn = 0.384Reaction: mwd31f52cc_04e7_40e0_885f_c7b2d9e62215 + mw2c9b0499_3325_4394_8af3_bbf653a944a0 => mw7becb5fe_8da8_4285_a821_0d77ad811b62; mw2c9b0499_3325_4394_8af3_bbf653a944a0, mw7becb5fe_8da8_4285_a821_0d77ad811b62, mwd31f52cc_04e7_40e0_885f_c7b2d9e62215, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*(kRLOn*mwd31f52cc_04e7_40e0_885f_c7b2d9e62215*mw2c9b0499_3325_4394_8af3_bbf653a944a0-kRLOff*mw7becb5fe_8da8_4285_a821_0d77ad811b62)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mw5832a2dc_ee18_44df_aa59_ccb21cb74df2 = 0.0054Reaction: mw114aa90f_5f5b_4fe8_9406_361c8489b6a1 => mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc + mw114aa90f_5f5b_4fe8_9406_361c8489b6a1; mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw5832a2dc_ee18_44df_aa59_ccb21cb74df2*mw114aa90f_5f5b_4fe8_9406_361c8489b6a1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw1f41474c_c399_4a60_a53a_9926dd092e8d = 3.9Reaction: => mwbbbce920_e8dd_4320_9386_fc94bfb2fc99, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw1f41474c_c399_4a60_a53a_9926dd092e8d/mw53ffe9e6_beef_45c4_90a5_a79197ed506e

States:

NameDescription
mw810ff751 fa4e 4143 bd50 169b3e325e1e[Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta; soluble in]
mw42054cd7 17af 46da 970c 7f99151906ad[signal transducer and activator of transcription 3]
mw772cbf20 3fc1 4800 ae59 77884f1ae333[Interleukin-6 receptor subunit alpha; Immunoglobulin]
mw7d86cc23 a1af 44c3 bdb9 71e9b1bb2a83[Interleukin-6 receptor subunit alpha; Interleukin-6]
mwbc2f5464 81e5 43fd 8b39 f5a2756af72f[Immunoglobulin]
mw3667a5e1 02c9 44a0 acb4 b0431faa822d[Immunoglobulin]
mwd2d9d93a 3bd1 4f17 bac1 baba9ef2d55a[Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta]
mwedc1bc00 adf7 4144 a1c2 7dc1a9565dc2[soluble in; Interleukin-6 receptor subunit alpha; Interleukin-6; Immunoglobulin]
mw03db56ac 8dc6 4931 ae82 fef706d2ee3d[Interleukin-6 receptor subunit alpha; Interleukin-6; soluble in]
mw0083d743 836f 4238 a17f 4602193d5bc0[CCO:U0000003]
mw48867e93 f170 44e8 ac7a 185b23e1bf3b[signal transducer and activator of transcription 3; phosphorylation]
mw0adf3eb4 a196 4c48 b10d 4e9e9faaf9e1[Interleukin-6]
mw6cce2109 0e32 4dd9 98ec 41173e8ef07d[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta; SBO:0000286; active]
mw5d764bb8 5693 4ac8 9557 f65992cc5eb0[Interleukin-6 receptor subunit alpha; Immunoglobulin; soluble in]
mw30ae63db 6cd3 4b6f 93ad 3350cd360bcc[Interleukin-6 receptor subunit alpha; soluble in]
mwf345ed7a 0622 403c b816 c8749a2c9ded[Immunoglobulin; pharmaceutical]
mwf7796221 1fea 4274 a93e c00adbf5778c[Immunoglobulin]
mw147d30ec 478e 4090 b496 128a131d29eb[interleukin-6 receptor subunit beta; soluble in]
mw1da111f2 a036 4392 8512 015005bdcbb7[Interleukin-6 receptor subunit alpha; Immunoglobulin; soluble in]
mwd65b5b39 dc1b 4e77 a999 67277a880e5e[interleukin-6 receptor subunit beta; soluble in]
mw36ea78c1 ed71 4def 96d3 857a442d7195[C-reactive protein]
mw4638f126 8cb8 4021 ab41 6ae195743ba0[Interleukin-6 receptor subunit alpha; Interleukin-6; soluble in]
mw0eb6c959 d408 45a0 a450 928b8c5876bb[Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta; SBO:0000286; active]
mw10315fa3 6f13 4618 bda8 a8694bd3c374[Interleukin-6 receptor subunit alpha]
mwbbbce920 e8dd 4320 9386 fc94bfb2fc99[soluble in; interleukin-6 receptor subunit beta]
mwd31f52cc 04e7 40e0 885f c7b2d9e62215[soluble in; Interleukin-6 receptor subunit alpha]
mwab41493c 6349 45f1 a226 3030cfed0e06[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta; soluble in]
mw2e464cf3 a09c 4b7c 9f3c 06720016a48e[soluble in; Interleukin-6 receptor subunit alpha]
mw8c9107e6 f51d 442d b2dc 2bfdbb8482ca[interleukin-6 receptor subunit beta]
mwf405687b 7401 44ec a0d6 4a2b35c13e8a[Interleukin-6 receptor subunit alpha; Immunoglobulin; soluble in]
mw7becb5fe 8da8 4285 a821 0d77ad811b62[Interleukin-6; Interleukin-6 receptor subunit alpha; soluble in]
mw824bc3d4 1ac3 4912 9b51 8f14ff1c96b9[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta]
mw114aa90f 5f5b 4fe8 9406 361c8489b6a1[C-reactive protein]
mw2ba2b802 9f07 4f4d 94c6 24c8de1a95cf[soluble in; Interleukin-6; Interleukin-6 receptor subunit alpha; Immunoglobulin]
mwf626e95e 543f 41e4 aad4 c6bf60ab345b[Interleukin-6]
mw2c9b0499 3325 4394 8af3 bbf653a944a0[Interleukin-6]
mw6335d5d7 c7b0 4bc0 b883 f7ee4915c2c3[soluble in; Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta]
mw39c2e431 fdc3 4964 be29 6ca856620b1b[phosphorylation; signal transducer and activator of transcription 3]

Dwivedi2014 - Crohns IL6 Disease model - sgp130 activity: BIOMD0000000536v0.0.1

Dwivedi2014 - Crohns IL6 Disease model - sgp130 activityThis model is comprised of four models: [[BIOMD0000000534]](htt…

Details

In this study, we have developed a multiscale systems model of interleukin (IL)-6-mediated immune regulation in Crohn's disease, by integrating intracellular signaling with organ-level dynamics of pharmacological markers underlying the disease. This model was linked to a general pharmacokinetic model for therapeutic monoclonal antibodies and used to comparatively study various biotherapeutic strategies targeting IL-6-mediated signaling in Crohn's disease. Our work illustrates techniques to develop mechanistic models of disease biology to study drug-system interaction. Despite a sparse training data set, predictions of the model were qualitatively validated by clinical biomarker data from a pilot trial with tocilizumab. Model-based analysis suggests that strategies targeting IL-6, IL-6Rα, or the IL-6/sIL-6Rα complex are less effective at suppressing pharmacological markers of Crohn's than dual targeting the IL-6/sIL-6Rα complex in addition to IL-6 or IL-6Rα. The potential value of multiscale system pharmacology modeling in drug discovery and development is also discussed.CPT: Pharmacometrics & Systems Pharmacology (2014) 3, e89; doi:10.1038/psp.2013.64; advance online publication 8 January 2014. link: http://identifiers.org/pubmed/24402116

Parameters:

NameDescription
mw862f1480_c60c_4863_a565_b2c1c77e238e = 0.5Reaction: mwd5313618_89eb_4c8c_bc82_66f10f966349 => mw36ea78c1_ed71_4def_96d3_857a442d7195; mwd5313618_89eb_4c8c_bc82_66f10f966349, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mw862f1480_c60c_4863_a565_b2c1c77e238e*mwd5313618_89eb_4c8c_bc82_66f10f966349/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
Metabolite_80 = 221.06367608557Reaction: species_1 = (mw114aa90f_5f5b_4fe8_9406_361c8489b6a1-Metabolite_80)/(Metabolite_80/(-100)), Rate Law: missing
kRint = 1.96Reaction: mw7d86cc23_a1af_44c3_bdb9_71e9b1bb2a83 => ; mw7d86cc23_a1af_44c3_bdb9_71e9b1bb2a83, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRint*mw7d86cc23_a1af_44c3_bdb9_71e9b1bb2a83/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRsynth = 0.0685Reaction: => mw10315fa3_6f13_4618_bda8_a8694bd3c374, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRsynth/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mw9f83bdd3_3aa1_47ff_abd6_54e5ce60704a = 0.0104166666666667; mwa071fdbe_d498_4620_a7a4_940aa31c8161 = 0.0208333333333333Reaction: mwa2d8dd1c_bb9a_4552_8738_e24671651c1d => mw2f3d48e0_c9c4_4a0e_aca3_9241eb573296; mwa2d8dd1c_bb9a_4552_8738_e24671651c1d, mw2f3d48e0_c9c4_4a0e_aca3_9241eb573296, Rate Law: mw9f83bdd3_3aa1_47ff_abd6_54e5ce60704a*mwa2d8dd1c_bb9a_4552_8738_e24671651c1d-mwa071fdbe_d498_4620_a7a4_940aa31c8161*mw2f3d48e0_c9c4_4a0e_aca3_9241eb573296
mw08950572_81b0_4570_b2e4_b9c3462c1425 = 10.0; mw92d854a7_8aaf_458e_b5e2_20a63ce9b654 = 330.0Reaction: mwd5313618_89eb_4c8c_bc82_66f10f966349 = mw92d854a7_8aaf_458e_b5e2_20a63ce9b654*mw39c2e431_fdc3_4964_be29_6ca856620b1b/(mw08950572_81b0_4570_b2e4_b9c3462c1425+mw39c2e431_fdc3_4964_be29_6ca856620b1b), Rate Law: missing
kIL6Decay = 34.82Reaction: mwf626e95e_543f_41e4_aad4_c6bf60ab345b => ; mwf626e95e_543f_41e4_aad4_c6bf60ab345b, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*kIL6Decay*mwf626e95e_543f_41e4_aad4_c6bf60ab345b/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwfd291862_195f_4979_94b5_b4e5ae1b7d52 = 5.34; mwd36b0261_2480_4cab_9222_2cf8fb0e65dc = 0.62Reaction: mw48867e93_f170_44e8_ac7a_185b23e1bf3b => mw2b255f94_8018_4b99_bde8_918eeac45446; mw48867e93_f170_44e8_ac7a_185b23e1bf3b, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwd36b0261_2480_4cab_9222_2cf8fb0e65dc*mw48867e93_f170_44e8_ac7a_185b23e1bf3b/(mwfd291862_195f_4979_94b5_b4e5ae1b7d52+mw48867e93_f170_44e8_ac7a_185b23e1bf3b)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwe8fc1900_f07d_468b_b5c8_15400a583c3d = 219.0; mw9442cd0e_4d7c_4ba6_a695_f84919bdf569 = 145.0Reaction: mw42054cd7_17af_46da_970c_7f99151906ad + mw0eb6c959_d408_45a0_a450_928b8c5876bb => mw39c2e431_fdc3_4964_be29_6ca856620b1b + mw0eb6c959_d408_45a0_a450_928b8c5876bb; mw0eb6c959_d408_45a0_a450_928b8c5876bb, mw42054cd7_17af_46da_970c_7f99151906ad, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mw9442cd0e_4d7c_4ba6_a695_f84919bdf569*mw0eb6c959_d408_45a0_a450_928b8c5876bb*mw42054cd7_17af_46da_970c_7f99151906ad/(mwe8fc1900_f07d_468b_b5c8_15400a583c3d+mw42054cd7_17af_46da_970c_7f99151906ad)/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mwf44f7f27_5bb1_4c7f_8964_560fa5e1743a = 0.01Reaction: mw0eb6c959_d408_45a0_a450_928b8c5876bb => ; mw0eb6c959_d408_45a0_a450_928b8c5876bb, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mwf44f7f27_5bb1_4c7f_8964_560fa5e1743a*mw0eb6c959_d408_45a0_a450_928b8c5876bb/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mw43ccad8c_cabf_4eaf_90d5_e06ae43be2cb = 0.0208333333333333; mw640ca705_e089_4c64_a5f4_9562317e8c76 = 0.0208333333333333Reaction: mwa2d8dd1c_bb9a_4552_8738_e24671651c1d => mw1d9426a3_e1e9_49e0_ad77_eb6833be398a; mwa2d8dd1c_bb9a_4552_8738_e24671651c1d, mw1d9426a3_e1e9_49e0_ad77_eb6833be398a, Rate Law: mw640ca705_e089_4c64_a5f4_9562317e8c76*mwa2d8dd1c_bb9a_4552_8738_e24671651c1d-mw43ccad8c_cabf_4eaf_90d5_e06ae43be2cb*mw1d9426a3_e1e9_49e0_ad77_eb6833be398a
kIL6Synth = 0.0063Reaction: => mwf626e95e_543f_41e4_aad4_c6bf60ab345b, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*kIL6Synth/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
kgp130Off = 1.026; kgp130On = 20.52Reaction: mw4638f126_8cb8_4021_ab41_6ae195743ba0 + mw147d30ec_478e_4090_b496_128a131d29eb => mwab41493c_6349_45f1_a226_3030cfed0e06; mw147d30ec_478e_4090_b496_128a131d29eb, mw4638f126_8cb8_4021_ab41_6ae195743ba0, mwab41493c_6349_45f1_a226_3030cfed0e06, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*(kgp130On*mw4638f126_8cb8_4021_ab41_6ae195743ba0*mw147d30ec_478e_4090_b496_128a131d29eb-kgp130Off*mwab41493c_6349_45f1_a226_3030cfed0e06)/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRAct = 155.0Reaction: mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a => mw0eb6c959_d408_45a0_a450_928b8c5876bb; mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRAct*mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mwce10678d_8197_408c_ad47_1daec8104cd8 = 0.8473; mwc67e1333_079a_4bea_9b4f_0a1b15ddd7bb = 1.2125Reaction: mwf626e95e_543f_41e4_aad4_c6bf60ab345b => mw0adf3eb4_a196_4c48_b10d_4e9e9faaf9e1; mwf626e95e_543f_41e4_aad4_c6bf60ab345b, mw0adf3eb4_a196_4c48_b10d_4e9e9faaf9e1, Rate Law: mwc67e1333_079a_4bea_9b4f_0a1b15ddd7bb*mwf626e95e_543f_41e4_aad4_c6bf60ab345b-mwce10678d_8197_408c_ad47_1daec8104cd8*mw0adf3eb4_a196_4c48_b10d_4e9e9faaf9e1
mwbd1d5bc3_d4b9_4aec_9b86_6f776da20a30 = 0.0018Reaction: mw1d9426a3_e1e9_49e0_ad77_eb6833be398a => ; mw1d9426a3_e1e9_49e0_ad77_eb6833be398a, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mwbd1d5bc3_d4b9_4aec_9b86_6f776da20a30*mw1d9426a3_e1e9_49e0_ad77_eb6833be398a/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mw1667a8e0_9d20_4e59_ba51_596148aba787 = 0.525; mwfcf06900_5f2f_4bb3_bb1f_12023612b8a8 = 155.3Reaction: mw0eb6c959_d408_45a0_a450_928b8c5876bb => mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a; mw0eb6c959_d408_45a0_a450_928b8c5876bb, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mw1667a8e0_9d20_4e59_ba51_596148aba787*mw0eb6c959_d408_45a0_a450_928b8c5876bb/(mwfcf06900_5f2f_4bb3_bb1f_12023612b8a8+mw0eb6c959_d408_45a0_a450_928b8c5876bb)/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mw88a75379_f9a1_4acc_baeb_94c32bb736a5 = 0.3Reaction: mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc => ; mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw88a75379_f9a1_4acc_baeb_94c32bb736a5*mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw65c85954_5ca0_4df2_9e22_ff2aa3fbe3f1 = 0.42Reaction: => mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw65c85954_5ca0_4df2_9e22_ff2aa3fbe3f1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwf67caf9d_2f4b_4986_abf2_e6090bbb72ce = 3.47222222222222E-4; mw4aea26f6_8860_414c_97f5_40d325196f2e = 0.00173611111111111Reaction: mwf345ed7a_0622_403c_b816_c8749a2c9ded => mwbc2f5464_81e5_43fd_8b39_f5a2756af72f; mwf345ed7a_0622_403c_b816_c8749a2c9ded, mwbc2f5464_81e5_43fd_8b39_f5a2756af72f, Rate Law: mwf67caf9d_2f4b_4986_abf2_e6090bbb72ce*mwf345ed7a_0622_403c_b816_c8749a2c9ded-mw4aea26f6_8860_414c_97f5_40d325196f2e*mwbc2f5464_81e5_43fd_8b39_f5a2756af72f
kRdeg = 0.1561Reaction: mw10315fa3_6f13_4618_bda8_a8694bd3c374 => ; mw10315fa3_6f13_4618_bda8_a8694bd3c374, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRdeg*mw10315fa3_6f13_4618_bda8_a8694bd3c374/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRLOff = 1.92; kRLOn = 0.384Reaction: mw10315fa3_6f13_4618_bda8_a8694bd3c374 + mw0adf3eb4_a196_4c48_b10d_4e9e9faaf9e1 => mw7d86cc23_a1af_44c3_bdb9_71e9b1bb2a83; mw0adf3eb4_a196_4c48_b10d_4e9e9faaf9e1, mw10315fa3_6f13_4618_bda8_a8694bd3c374, mw7d86cc23_a1af_44c3_bdb9_71e9b1bb2a83, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*(kRLOn*mw10315fa3_6f13_4618_bda8_a8694bd3c374*mw0adf3eb4_a196_4c48_b10d_4e9e9faaf9e1-kRLOff*mw7d86cc23_a1af_44c3_bdb9_71e9b1bb2a83)/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mw5832a2dc_ee18_44df_aa59_ccb21cb74df2 = 0.0054Reaction: mw114aa90f_5f5b_4fe8_9406_361c8489b6a1 => mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc + mw114aa90f_5f5b_4fe8_9406_361c8489b6a1; mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw5832a2dc_ee18_44df_aa59_ccb21cb74df2*mw114aa90f_5f5b_4fe8_9406_361c8489b6a1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw1f41474c_c399_4a60_a53a_9926dd092e8d = 3.9Reaction: => mwbbbce920_e8dd_4320_9386_fc94bfb2fc99, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw1f41474c_c399_4a60_a53a_9926dd092e8d/mw53ffe9e6_beef_45c4_90a5_a79197ed506e

States:

NameDescription
mw1d9426a3 e1e9 49e0 ad77 eb6833be398a[Interleukin-6 receptor subunit alpha; Interleukin-6; Immunoglobulin; soluble in]
mw42054cd7 17af 46da 970c 7f99151906ad[signal transducer and activator of transcription 3]
mw810ff751 fa4e 4143 bd50 169b3e325e1e[Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta; soluble in]
mw7d86cc23 a1af 44c3 bdb9 71e9b1bb2a83[Interleukin-6 receptor subunit alpha; Interleukin-6]
species 1[C-reactive protein]
mwbc2f5464 81e5 43fd 8b39 f5a2756af72f[Immunoglobulin]
mwa2d8dd1c bb9a 4552 8738 e24671651c1d[Interleukin-6 receptor subunit alpha; Interleukin-6; Immunoglobulin; soluble in]
mwd2d9d93a 3bd1 4f17 bac1 baba9ef2d55a[Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta]
mw3667a5e1 02c9 44a0 acb4 b0431faa822d[Immunoglobulin]
mw03db56ac 8dc6 4931 ae82 fef706d2ee3d[Interleukin-6 receptor subunit alpha; Interleukin-6; soluble in]
mw0adf3eb4 a196 4c48 b10d 4e9e9faaf9e1[Interleukin-6]
mw48867e93 f170 44e8 ac7a 185b23e1bf3b[phosphorylation; signal transducer and activator of transcription 3]
mw6cce2109 0e32 4dd9 98ec 41173e8ef07d[SBO:0000286; active; Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta]
mwd5313618 89eb 4c8c bc82 66f10f966349[C-reactive protein]
mw30ae63db 6cd3 4b6f 93ad 3350cd360bcc[soluble in; Interleukin-6 receptor subunit alpha]
mwf345ed7a 0622 403c b816 c8749a2c9ded[Immunoglobulin]
mwf7796221 1fea 4274 a93e c00adbf5778c[Immunoglobulin]
mw147d30ec 478e 4090 b496 128a131d29eb[soluble in; interleukin-6 receptor subunit beta]
mw4638f126 8cb8 4021 ab41 6ae195743ba0[Interleukin-6 receptor subunit alpha; Interleukin-6; soluble in]
mw0eb6c959 d408 45a0 a450 928b8c5876bb[SBO:0000286; active; Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta]
mw10315fa3 6f13 4618 bda8 a8694bd3c374[Interleukin-6 receptor subunit alpha]
mwab41493c 6349 45f1 a226 3030cfed0e06[soluble in; Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta]
mwbbbce920 e8dd 4320 9386 fc94bfb2fc99[soluble in; interleukin-6 receptor subunit beta]
mw80848184 e2dd 47ce 86d7 7a21479342bd[interleukin-6 receptor subunit beta]
mw2b255f94 8018 4b99 bde8 918eeac45446[signal transducer and activator of transcription 3]
mw8c9107e6 f51d 442d b2dc 2bfdbb8482ca[interleukin-6 receptor subunit beta]
mw2f3d48e0 c9c4 4a0e aca3 9241eb573296[soluble in; Interleukin-6 receptor subunit alpha; Interleukin-6; Immunoglobulin]
mw114aa90f 5f5b 4fe8 9406 361c8489b6a1[C-reactive protein]
mw7becb5fe 8da8 4285 a821 0d77ad811b62[Interleukin-6; Interleukin-6 receptor subunit alpha; soluble in]
mw824bc3d4 1ac3 4912 9b51 8f14ff1c96b9[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta]
mwf626e95e 543f 41e4 aad4 c6bf60ab345b[Interleukin-6]
mw2c9b0499 3325 4394 8af3 bbf653a944a0[Interleukin-6]

Dwivedi2014 - Healthy Volunteer IL6 Model: BIOMD0000000534v0.0.1

Dwivedi2014 - Healthy Volunteer IL6 ModelThis model is comprised of four models: [[BIOMD0000000534]](http://www.ebi.ac.…

Details

In this study, we have developed a multiscale systems model of interleukin (IL)-6-mediated immune regulation in Crohn's disease, by integrating intracellular signaling with organ-level dynamics of pharmacological markers underlying the disease. This model was linked to a general pharmacokinetic model for therapeutic monoclonal antibodies and used to comparatively study various biotherapeutic strategies targeting IL-6-mediated signaling in Crohn's disease. Our work illustrates techniques to develop mechanistic models of disease biology to study drug-system interaction. Despite a sparse training data set, predictions of the model were qualitatively validated by clinical biomarker data from a pilot trial with tocilizumab. Model-based analysis suggests that strategies targeting IL-6, IL-6Rα, or the IL-6/sIL-6Rα complex are less effective at suppressing pharmacological markers of Crohn's than dual targeting the IL-6/sIL-6Rα complex in addition to IL-6 or IL-6Rα. The potential value of multiscale system pharmacology modeling in drug discovery and development is also discussed.CPT: Pharmacometrics & Systems Pharmacology (2014) 3, e89; doi:10.1038/psp.2013.64; advance online publication 8 January 2014. link: http://identifiers.org/pubmed/24402116

Parameters:

NameDescription
mw862f1480_c60c_4863_a565_b2c1c77e238e = 0.5Reaction: mwd5313618_89eb_4c8c_bc82_66f10f966349 => mw36ea78c1_ed71_4def_96d3_857a442d7195; mwd5313618_89eb_4c8c_bc82_66f10f966349, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mw862f1480_c60c_4863_a565_b2c1c77e238e*mwd5313618_89eb_4c8c_bc82_66f10f966349/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRsynth = 0.0685Reaction: => mw10315fa3_6f13_4618_bda8_a8694bd3c374, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRsynth/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRint = 1.96Reaction: mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a => ; mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRint*mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mw9f83bdd3_3aa1_47ff_abd6_54e5ce60704a = 0.0104166666666667; mwa071fdbe_d498_4620_a7a4_940aa31c8161 = 0.0208333333333333Reaction: mwf345ed7a_0622_403c_b816_c8749a2c9ded => mwf7796221_1fea_4274_a93e_c00adbf5778c; mwf345ed7a_0622_403c_b816_c8749a2c9ded, mwf7796221_1fea_4274_a93e_c00adbf5778c, Rate Law: mw9f83bdd3_3aa1_47ff_abd6_54e5ce60704a*mwf345ed7a_0622_403c_b816_c8749a2c9ded-mwa071fdbe_d498_4620_a7a4_940aa31c8161*mwf7796221_1fea_4274_a93e_c00adbf5778c
mw06241335_b5f2_47ed_bdcc_ef77b68a2b98 = 1.0Reaction: mw2c9b0499_3325_4394_8af3_bbf653a944a0 => ; mw2c9b0499_3325_4394_8af3_bbf653a944a0, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mw06241335_b5f2_47ed_bdcc_ef77b68a2b98*mw2c9b0499_3325_4394_8af3_bbf653a944a0/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mw08950572_81b0_4570_b2e4_b9c3462c1425 = 10.0; mw92d854a7_8aaf_458e_b5e2_20a63ce9b654 = 330.0Reaction: mw0083d743_836f_4238_a17f_4602193d5bc0 = mw92d854a7_8aaf_458e_b5e2_20a63ce9b654*mw48867e93_f170_44e8_ac7a_185b23e1bf3b/(mw08950572_81b0_4570_b2e4_b9c3462c1425+mw48867e93_f170_44e8_ac7a_185b23e1bf3b), Rate Law: missing
mwfd291862_195f_4979_94b5_b4e5ae1b7d52 = 5.34; mwd36b0261_2480_4cab_9222_2cf8fb0e65dc = 0.62Reaction: mw48867e93_f170_44e8_ac7a_185b23e1bf3b => mw2b255f94_8018_4b99_bde8_918eeac45446; mw48867e93_f170_44e8_ac7a_185b23e1bf3b, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwd36b0261_2480_4cab_9222_2cf8fb0e65dc*mw48867e93_f170_44e8_ac7a_185b23e1bf3b/(mwfd291862_195f_4979_94b5_b4e5ae1b7d52+mw48867e93_f170_44e8_ac7a_185b23e1bf3b)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwe8fc1900_f07d_468b_b5c8_15400a583c3d = 219.0; mw9442cd0e_4d7c_4ba6_a695_f84919bdf569 = 145.0Reaction: mw42054cd7_17af_46da_970c_7f99151906ad + mw0eb6c959_d408_45a0_a450_928b8c5876bb => mw39c2e431_fdc3_4964_be29_6ca856620b1b + mw0eb6c959_d408_45a0_a450_928b8c5876bb; mw0eb6c959_d408_45a0_a450_928b8c5876bb, mw42054cd7_17af_46da_970c_7f99151906ad, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mw9442cd0e_4d7c_4ba6_a695_f84919bdf569*mw0eb6c959_d408_45a0_a450_928b8c5876bb*mw42054cd7_17af_46da_970c_7f99151906ad/(mwe8fc1900_f07d_468b_b5c8_15400a583c3d+mw42054cd7_17af_46da_970c_7f99151906ad)/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mwf44f7f27_5bb1_4c7f_8964_560fa5e1743a = 0.01Reaction: mw0eb6c959_d408_45a0_a450_928b8c5876bb => ; mw0eb6c959_d408_45a0_a450_928b8c5876bb, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mwf44f7f27_5bb1_4c7f_8964_560fa5e1743a*mw0eb6c959_d408_45a0_a450_928b8c5876bb/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mw43ccad8c_cabf_4eaf_90d5_e06ae43be2cb = 0.0208333333333333; mw640ca705_e089_4c64_a5f4_9562317e8c76 = 0.0208333333333333Reaction: mwf345ed7a_0622_403c_b816_c8749a2c9ded => mw3667a5e1_02c9_44a0_acb4_b0431faa822d; mwf345ed7a_0622_403c_b816_c8749a2c9ded, mw3667a5e1_02c9_44a0_acb4_b0431faa822d, Rate Law: mw640ca705_e089_4c64_a5f4_9562317e8c76*mwf345ed7a_0622_403c_b816_c8749a2c9ded-mw43ccad8c_cabf_4eaf_90d5_e06ae43be2cb*mw3667a5e1_02c9_44a0_acb4_b0431faa822d
kgp130Off = 1.026; kgp130On = 20.52Reaction: mwd65b5b39_dc1b_4e77_a999_67277a880e5e + mw7becb5fe_8da8_4285_a821_0d77ad811b62 => mw6335d5d7_c7b0_4bc0_b883_f7ee4915c2c3; mw6335d5d7_c7b0_4bc0_b883_f7ee4915c2c3, mw7becb5fe_8da8_4285_a821_0d77ad811b62, mwd65b5b39_dc1b_4e77_a999_67277a880e5e, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*(kgp130On*mwd65b5b39_dc1b_4e77_a999_67277a880e5e*mw7becb5fe_8da8_4285_a821_0d77ad811b62-kgp130Off*mw6335d5d7_c7b0_4bc0_b883_f7ee4915c2c3)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwbd1d5bc3_d4b9_4aec_9b86_6f776da20a30 = 0.0018Reaction: mwf7796221_1fea_4274_a93e_c00adbf5778c => ; mwf7796221_1fea_4274_a93e_c00adbf5778c, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwbd1d5bc3_d4b9_4aec_9b86_6f776da20a30*mwf7796221_1fea_4274_a93e_c00adbf5778c/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwce10678d_8197_408c_ad47_1daec8104cd8 = 0.8473; mwc67e1333_079a_4bea_9b4f_0a1b15ddd7bb = 1.2125Reaction: mwf626e95e_543f_41e4_aad4_c6bf60ab345b => mw2c9b0499_3325_4394_8af3_bbf653a944a0; mwf626e95e_543f_41e4_aad4_c6bf60ab345b, mw2c9b0499_3325_4394_8af3_bbf653a944a0, Rate Law: mwc67e1333_079a_4bea_9b4f_0a1b15ddd7bb*mwf626e95e_543f_41e4_aad4_c6bf60ab345b-mwce10678d_8197_408c_ad47_1daec8104cd8*mw2c9b0499_3325_4394_8af3_bbf653a944a0
mw88a75379_f9a1_4acc_baeb_94c32bb736a5 = 0.3Reaction: mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc => ; mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw88a75379_f9a1_4acc_baeb_94c32bb736a5*mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
kRAct = 155.0Reaction: mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a => mw0eb6c959_d408_45a0_a450_928b8c5876bb; mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRAct*mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mw1667a8e0_9d20_4e59_ba51_596148aba787 = 0.525; mwfcf06900_5f2f_4bb3_bb1f_12023612b8a8 = 155.3Reaction: mw0eb6c959_d408_45a0_a450_928b8c5876bb => mwd2d9d93a_3bd1_4f17_bac1_baba9ef2d55a; mw0eb6c959_d408_45a0_a450_928b8c5876bb, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*mw1667a8e0_9d20_4e59_ba51_596148aba787*mw0eb6c959_d408_45a0_a450_928b8c5876bb/(mwfcf06900_5f2f_4bb3_bb1f_12023612b8a8+mw0eb6c959_d408_45a0_a450_928b8c5876bb)/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mwbcb5a310_9b67_405e_89ec_43d25e8cc93d = 1.0Reaction: mwbbbce920_e8dd_4320_9386_fc94bfb2fc99 => ; mwbbbce920_e8dd_4320_9386_fc94bfb2fc99, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mwbcb5a310_9b67_405e_89ec_43d25e8cc93d*mwbbbce920_e8dd_4320_9386_fc94bfb2fc99/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwa09d6284_843e_404e_abbb_052fbb535197 = 1000.0; mw1c4bc9c3_52ad_4ef7_bf7f_97b0e2101ead = 2.5Reaction: mwf7796221_1fea_4274_a93e_c00adbf5778c + mw2c9b0499_3325_4394_8af3_bbf653a944a0 => mw5d764bb8_5693_4ac8_9557_f65992cc5eb0; mw2c9b0499_3325_4394_8af3_bbf653a944a0, mw5d764bb8_5693_4ac8_9557_f65992cc5eb0, mwf7796221_1fea_4274_a93e_c00adbf5778c, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*(mwa09d6284_843e_404e_abbb_052fbb535197*mwf7796221_1fea_4274_a93e_c00adbf5778c*mw2c9b0499_3325_4394_8af3_bbf653a944a0-mw1c4bc9c3_52ad_4ef7_bf7f_97b0e2101ead*mw5d764bb8_5693_4ac8_9557_f65992cc5eb0)/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwa8d72918_f6c2_4d81_bf3b_fc2b464d5e69 = 0.0017Reaction: => mw2c9b0499_3325_4394_8af3_bbf653a944a0, Rate Law: mwe9501423_9fb4_494b_b5b6_288f3fcb17b5*mwa8d72918_f6c2_4d81_bf3b_fc2b464d5e69/mwe9501423_9fb4_494b_b5b6_288f3fcb17b5
mwc4c58db7_5535_4590_aaa5_bbc8ed53cdab = 0.1Reaction: => mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mwc4c58db7_5535_4590_aaa5_bbc8ed53cdab/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw65c85954_5ca0_4df2_9e22_ff2aa3fbe3f1 = 0.42Reaction: => mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw65c85954_5ca0_4df2_9e22_ff2aa3fbe3f1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mwf67caf9d_2f4b_4986_abf2_e6090bbb72ce = 3.47222222222222E-4; mw4aea26f6_8860_414c_97f5_40d325196f2e = 0.00173611111111111Reaction: mwf345ed7a_0622_403c_b816_c8749a2c9ded => mwbc2f5464_81e5_43fd_8b39_f5a2756af72f; mwf345ed7a_0622_403c_b816_c8749a2c9ded, mwbc2f5464_81e5_43fd_8b39_f5a2756af72f, Rate Law: mwf67caf9d_2f4b_4986_abf2_e6090bbb72ce*mwf345ed7a_0622_403c_b816_c8749a2c9ded-mw4aea26f6_8860_414c_97f5_40d325196f2e*mwbc2f5464_81e5_43fd_8b39_f5a2756af72f
kRdeg = 0.1561Reaction: mw10315fa3_6f13_4618_bda8_a8694bd3c374 => ; mw10315fa3_6f13_4618_bda8_a8694bd3c374, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*kRdeg*mw10315fa3_6f13_4618_bda8_a8694bd3c374/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
kRLOff = 1.92; kRLOn = 0.384Reaction: mw2e464cf3_a09c_4b7c_9f3c_06720016a48e + mw0adf3eb4_a196_4c48_b10d_4e9e9faaf9e1 => mw4638f126_8cb8_4021_ab41_6ae195743ba0; mw0adf3eb4_a196_4c48_b10d_4e9e9faaf9e1, mw2e464cf3_a09c_4b7c_9f3c_06720016a48e, mw4638f126_8cb8_4021_ab41_6ae195743ba0, Rate Law: mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e*(kRLOn*mw2e464cf3_a09c_4b7c_9f3c_06720016a48e*mw0adf3eb4_a196_4c48_b10d_4e9e9faaf9e1-kRLOff*mw4638f126_8cb8_4021_ab41_6ae195743ba0)/mw88ca8d9a_f5cf_41bf_9d9d_fc48f6e1a19e
mw5832a2dc_ee18_44df_aa59_ccb21cb74df2 = 0.0054Reaction: mw114aa90f_5f5b_4fe8_9406_361c8489b6a1 => mw30ae63db_6cd3_4b6f_93ad_3350cd360bcc + mw114aa90f_5f5b_4fe8_9406_361c8489b6a1; mw114aa90f_5f5b_4fe8_9406_361c8489b6a1, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw5832a2dc_ee18_44df_aa59_ccb21cb74df2*mw114aa90f_5f5b_4fe8_9406_361c8489b6a1/mw53ffe9e6_beef_45c4_90a5_a79197ed506e
mw1f41474c_c399_4a60_a53a_9926dd092e8d = 3.9Reaction: => mwbbbce920_e8dd_4320_9386_fc94bfb2fc99, Rate Law: mw53ffe9e6_beef_45c4_90a5_a79197ed506e*mw1f41474c_c399_4a60_a53a_9926dd092e8d/mw53ffe9e6_beef_45c4_90a5_a79197ed506e

States:

NameDescription
mw1d9426a3 e1e9 49e0 ad77 eb6833be398a[Interleukin-6; Interleukin-6 receptor subunit alpha; Immunoglobulin; soluble in]
mw810ff751 fa4e 4143 bd50 169b3e325e1e[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta; soluble in]
mw7d86cc23 a1af 44c3 bdb9 71e9b1bb2a83[Interleukin-6; Interleukin-6 receptor subunit alpha]
mwbc2f5464 81e5 43fd 8b39 f5a2756af72f[Immunoglobulin]
mwa2d8dd1c bb9a 4552 8738 e24671651c1d[Interleukin-6; Interleukin-6 receptor subunit alpha; Immunoglobulin; soluble in]
mwd2d9d93a 3bd1 4f17 bac1 baba9ef2d55a[Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta]
mw3667a5e1 02c9 44a0 acb4 b0431faa822d[Immunoglobulin]
mw03db56ac 8dc6 4931 ae82 fef706d2ee3d[Interleukin-6 receptor subunit alpha; Interleukin-6; soluble in]
mw0083d743 836f 4238 a17f 4602193d5bc0[CCO:U0000003]
mw0adf3eb4 a196 4c48 b10d 4e9e9faaf9e1[Interleukin-6]
mw48867e93 f170 44e8 ac7a 185b23e1bf3b[signal transducer and activator of transcription 3; phosphorylation]
mw6cce2109 0e32 4dd9 98ec 41173e8ef07d[SBO:0000286; active; Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta]
mw5d764bb8 5693 4ac8 9557 f65992cc5eb0[Interleukin-6; Immunoglobulin]
mw30ae63db 6cd3 4b6f 93ad 3350cd360bcc[Interleukin-6 receptor subunit alpha; soluble in]
mwf345ed7a 0622 403c b816 c8749a2c9ded[Immunoglobulin]
mwf7796221 1fea 4274 a93e c00adbf5778c[Immunoglobulin]
mw147d30ec 478e 4090 b496 128a131d29eb[soluble in; interleukin-6 receptor subunit beta]
mw1da111f2 a036 4392 8512 015005bdcbb7[Interleukin-6; Immunoglobulin]
mwd65b5b39 dc1b 4e77 a999 67277a880e5e[soluble in; interleukin-6 receptor subunit beta]
mw36ea78c1 ed71 4def 96d3 857a442d7195[C-reactive protein]
mw4638f126 8cb8 4021 ab41 6ae195743ba0[Interleukin-6 receptor subunit alpha; Interleukin-6; soluble in]
mw0eb6c959 d408 45a0 a450 928b8c5876bb[Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta; SBO:0000286; active]
mw10315fa3 6f13 4618 bda8 a8694bd3c374[Interleukin-6 receptor subunit alpha]
mwab41493c 6349 45f1 a226 3030cfed0e06[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta; soluble in]
mwd31f52cc 04e7 40e0 885f c7b2d9e62215[soluble in; Interleukin-6 receptor subunit alpha]
mw80848184 e2dd 47ce 86d7 7a21479342bd[interleukin-6 receptor subunit beta]
mwbbbce920 e8dd 4320 9386 fc94bfb2fc99[interleukin-6 receptor subunit beta; soluble in]
mw2e464cf3 a09c 4b7c 9f3c 06720016a48e[Interleukin-6 receptor subunit alpha; soluble in]
mw2b255f94 8018 4b99 bde8 918eeac45446[signal transducer and activator of transcription 3]
mw8c9107e6 f51d 442d b2dc 2bfdbb8482ca[interleukin-6 receptor subunit beta]
mwf405687b 7401 44ec a0d6 4a2b35c13e8a[Interleukin-6; Immunoglobulin]
mw2f3d48e0 c9c4 4a0e aca3 9241eb573296[soluble in; Interleukin-6; Interleukin-6 receptor subunit alpha; Immunoglobulin]
mw7becb5fe 8da8 4285 a821 0d77ad811b62[Interleukin-6 receptor subunit alpha; Interleukin-6; soluble in]
mw824bc3d4 1ac3 4912 9b51 8f14ff1c96b9[Interleukin-6 receptor subunit alpha; Interleukin-6; interleukin-6 receptor subunit beta]
mw114aa90f 5f5b 4fe8 9406 361c8489b6a1[C-reactive protein]
mwf626e95e 543f 41e4 aad4 c6bf60ab345b[Interleukin-6]
mw2c9b0499 3325 4394 8af3 bbf653a944a0[Interleukin-6]
mw6335d5d7 c7b0 4bc0 b883 f7ee4915c2c3[Interleukin-6; Interleukin-6 receptor subunit alpha; interleukin-6 receptor subunit beta; soluble in]

E


E2_PBPK: MODEL1711210001v0.0.1

Using scaling from PhysB model Blood flow in L/hr Compartments in Kg Baseline as ~0.013nM free E2 in plasma_venous E2 b…

Details

Estrogen is a vital hormone that regulates many biological functions within the body. These include roles in the development of the secondary sexual organs in both sexes, plus uterine angiogenesis and proliferation during the menstrual cycle and pregnancy in women. The varied biological roles of estrogens in human health also make them a therapeutic target for contraception, mitigation of the adverse effects of the menopause, and treatment of estrogen-responsive tumours. In addition, endogenous (e.g. genetic variation) and external (e.g. exposure to estrogen-like chemicals) factors are known to impact estrogen biology. To understand how these multiple factors interact to determine an individual's response to therapy is complex, and may be best approached through a systems approach.We present a physiologically-based pharmacokinetic model (PBPK) of estradiol, and validate it against plasma kinetics in humans following intravenous and oral exposure. We extend this model by replacing the intrinsic clearance term with: a detailed kinetic model of estrogen metabolism in the liver; or, a genome-scale model of liver metabolism. Both models were validated by their ability to reproduce clinical data on estradiol exposure. We hypothesise that the enhanced mechanistic information contained within these models will lead to more robust predictions of the biological phenotype that emerges from the complex interactions between estrogens and the body.To demonstrate the utility of these models we examine the known drug-drug interactions between phenytoin and oral estradiol. We are able to reproduce the approximate 50% reduction in area under the concentration-time curve for estradiol associated with this interaction. Importantly, the inclusion of a genome-scale metabolic model allows the prediction of this interaction without directly specifying it within the model. In addition, we predict that PXR activation by drugs results in an enhanced ability of the liver to excrete glucose. This has important implications for the relationship between drug treatment and metabolic syndrome.We demonstrate how the novel coupling of PBPK models with genome-scale metabolic networks has the potential to aid prediction of drug action, including both drug-drug interactions and changes to the metabolic landscape that may predispose an individual to disease development. link: http://identifiers.org/pubmed/29246152

Earm1990_CalciumDynamics_Cardiac: MODEL1006230070v0.0.1

This a model from the article: A model of the single atrial cell: relation between calcium current and calcium release…

Details

The hypothesis that calcium release from the sarcoplasmic reticulum in cardiac muscle is induced by rises in free cytosolic calcium (Fabiato 1983, Am. J. Physiol 245) allows the possibility that the release could be at least partly regenerative. There would then be a non-linear relation between calcium current and calcium release. We have investigated this possibility in a single-cell version of the rabbit-atrial model developed by Hilgemann & Noble (1987, Proc. R. Soc. Lond. B 230). The model predicts different voltage ranges of activation for calcium-dependent processes (like the sodium-calcium exchange current, contraction or Fura-2 signals) and the calcium current, in agreement with the experimental results obtained by Earm et al. (1990, Proc. R. Soc. Lond. B 240) on exchange current tails, Cannell et al. (1987, Science, Wash. 238) by using Fura-2 signals, and Fedida et al. (1987, J. Physiol., Lond. 385) and Talo et al. (1988, Biology of isolated adult cardiac myocytes) by using contraction. However, when the Fura-2 concentration is sufficiently high (greater than 200 microM) the activation ranges become very similar as the buffering properties of Fura-2 are sufficient to remove the regenerative effect. It is therefore important to allow for the buffering properties of calcium indicators when investigating the correlation between calcium current and calcium release. link: http://identifiers.org/pubmed/1972993

Edelstein1996 - EPSP ACh event: BIOMD0000000001v0.0.1

Edelstein1996 - EPSP ACh eventModel of a nicotinic Excitatory Post-Synaptic Potential in a Torpedo electric organ. Acet…

Details

Nicotinic acetylcholine receptors are transmembrane oligomeric proteins that mediate interconversions between open and closed channel states under the control of neurotransmitters. Fast in vitro chemical kinetics and in vivo electrophysiological recordings are consistent with the following multi-step scheme. Upon binding of agonists, receptor molecules in the closed but activatable resting state (the Basal state, B) undergo rapid transitions to states of higher affinities with either open channels (the Active state, A) or closed channels (the initial Inactivatable and fully Desensitized states, I and D). In order to represent the functional properties of such receptors, we have developed a kinetic model that links conformational interconversion rates to agonist binding and extends the general principles of the Monod-Wyman-Changeux model of allosteric transitions. The crucial assumption is that the linkage is controlled by the position of the interconversion transition states on a hypothetical linear reaction coordinate. Application of the model to the peripheral nicotine acetylcholine receptor (nAChR) accounts for the main properties of ligand-gating, including single-channel events, and several new relationships are predicted. Kinetic simulations reveal errors inherent in using the dose-response analysis, but justify its application under defined conditions. The model predicts that (in order to overcome the intrinsic stability of the B state and to produce the appropriate cooperativity) channel activation is driven by an A state with a Kd in the 50 nM range, hence some 140-fold stronger than the apparent affinity of the open state deduced previously. According to the model, recovery from the desensitized states may occur via rapid transit through the A state with minimal channel opening, thus without necessarily undergoing a distinct recovery pathway, as assumed in the standard 'cycle' model. Transitions to the desensitized states by low concentration 'pre-pulses' are predicted to occur without significant channel opening, but equilibrium values of IC50 can be obtained only with long pre-pulse times. Predictions are also made concerning allosteric effectors and their possible role in coincidence detection. In terms of future developments, the analysis presented here provides a physical basis for constructing more biologically realistic models of synaptic modulation that may be applied to artificial neural networks. link: http://identifiers.org/pubmed/8983160

Parameters:

NameDescription
kr_12 = 4.0; kf_12 = 3000.0Reaction: D => DL, Rate Law: comp1*(kf_12*D-kr_12*DL)
kr_2 = 700.0; kf_2 = 30000.0Reaction: BLL => ALL, Rate Law: comp1*(kf_2*BLL-kr_2*ALL)
kf_5 = 0.54; kr_5 = 10800.0Reaction: B => A, Rate Law: comp1*(kf_5*B-kr_5*A)
kr_6 = 2740.0; kf_6 = 130.0Reaction: BL => AL, Rate Law: comp1*(kf_6*BL-kr_6*AL)
kr_16 = 0.0012; kf_16 = 0.05Reaction: ILL => DLL, Rate Law: comp1*(kf_16*ILL-kr_16*DLL)
kr_7 = 4.0; kf_7 = 3000.0Reaction: I => IL, Rate Law: comp1*(kf_7*I-kr_7*IL)
kf_13 = 1500.0; kr_13 = 8.0Reaction: DL => DLL, Rate Law: comp1*(kf_13*DL-kr_13*DLL)
kf_4 = 1500.0; kr_4 = 17.28Reaction: AL => ALL, Rate Law: comp1*(kf_4*AL-kr_4*ALL)
kf_11 = 20.0; kr_11 = 0.81Reaction: ALL => ILL, Rate Law: comp1*(kf_11*ALL-kr_11*ILL)
kf_14 = 0.05; kr_14 = 0.0012Reaction: I => D, Rate Law: comp1*(kf_14*I-kr_14*D)
kr_1 = 16000.0; kf_1 = 1500.0Reaction: BL => BLL, Rate Law: comp1*(kf_1*BL-kr_1*BLL)
kr_15 = 0.0012; kf_15 = 0.05Reaction: IL => DL, Rate Law: comp1*(kf_15*IL-kr_15*DL)
kf_9 = 19.7; kr_9 = 3.74Reaction: A => I, Rate Law: comp1*(kf_9*A-kr_9*I)
kf_0 = 3000.0; kr_0 = 8000.0Reaction: B => BL, Rate Law: comp1*(kf_0*B-kr_0*BL)
kr_3 = 8.64; kf_3 = 3000.0Reaction: A => AL, Rate Law: comp1*(kf_3*A-kr_3*AL)
kf_10 = 19.85; kr_10 = 1.74Reaction: AL => IL, Rate Law: comp1*(kf_10*AL-kr_10*IL)
kf_8 = 1500.0; kr_8 = 8.0Reaction: IL => ILL, Rate Law: comp1*(kf_8*IL-kr_8*ILL)

States:

NameDescription
BLL[IPR002394; acetylcholine-gated channel complex]
DLL[IPR002394; acetylcholine-gated channel complex]
A[IPR002394; acetylcholine-gated channel complex]
BL[IPR002394; acetylcholine-gated channel complex]
D[IPR002394; acetylcholine-gated channel complex]
ALL[IPR002394; acetylcholine-gated channel complex]
B[IPR002394; acetylcholine-gated channel complex]
I[IPR002394; acetylcholine-gated channel complex]
AL[IPR002394; acetylcholine-gated channel complex]
DL[IPR002394; acetylcholine-gated channel complex]
ILL[IPR002394; acetylcholine-gated channel complex]
IL[IPR002394; acetylcholine-gated channel complex]

Edelstein1996 - EPSP ACh species: BIOMD0000000002v0.0.1

Edelstein1996 - EPSP ACh speciesModel of a nicotinic Excitatory Post-Synaptic Potential in a Torpedo electric organ. Ace…

Details

Nicotinic acetylcholine receptors are transmembrane oligomeric proteins that mediate interconversions between open and closed channel states under the control of neurotransmitters. Fast in vitro chemical kinetics and in vivo electrophysiological recordings are consistent with the following multi-step scheme. Upon binding of agonists, receptor molecules in the closed but activatable resting state (the Basal state, B) undergo rapid transitions to states of higher affinities with either open channels (the Active state, A) or closed channels (the initial Inactivatable and fully Desensitized states, I and D). In order to represent the functional properties of such receptors, we have developed a kinetic model that links conformational interconversion rates to agonist binding and extends the general principles of the Monod-Wyman-Changeux model of allosteric transitions. The crucial assumption is that the linkage is controlled by the position of the interconversion transition states on a hypothetical linear reaction coordinate. Application of the model to the peripheral nicotine acetylcholine receptor (nAChR) accounts for the main properties of ligand-gating, including single-channel events, and several new relationships are predicted. Kinetic simulations reveal errors inherent in using the dose-response analysis, but justify its application under defined conditions. The model predicts that (in order to overcome the intrinsic stability of the B state and to produce the appropriate cooperativity) channel activation is driven by an A state with a Kd in the 50 nM range, hence some 140-fold stronger than the apparent affinity of the open state deduced previously. According to the model, recovery from the desensitized states may occur via rapid transit through the A state with minimal channel opening, thus without necessarily undergoing a distinct recovery pathway, as assumed in the standard 'cycle' model. Transitions to the desensitized states by low concentration 'pre-pulses' are predicted to occur without significant channel opening, but equilibrium values of IC50 can be obtained only with long pre-pulse times. Predictions are also made concerning allosteric effectors and their possible role in coincidence detection. In terms of future developments, the analysis presented here provides a physical basis for constructing more biologically realistic models of synaptic modulation that may be applied to artificial neural networks. link: http://identifiers.org/pubmed/8983160

Parameters:

NameDescription
kr_3 = 8.64; kf_3 = 3.0E8Reaction: A + L => AL, Rate Law: comp1*(kf_3*A*L-kr_3*AL)
kr_6 = 2740.0; kf_6 = 130.0Reaction: BL => AL, Rate Law: comp1*(kf_6*BL-kr_6*AL)
kf_5 = 0.54; kr_5 = 10800.0Reaction: B => A, Rate Law: comp1*(kf_5*B-kr_5*A)
kf_4 = 1.5E8; kr_4 = 17.28Reaction: AL + L => ALL, Rate Law: comp1*(kf_4*AL*L-kr_4*ALL)
kr_2 = 700.0; kf_2 = 30000.0Reaction: BLL => ALL, Rate Law: comp1*(kf_2*BLL-kr_2*ALL)
kf_13 = 1.5E8; kr_13 = 8.0Reaction: DL + L => DLL, Rate Law: comp1*(kf_13*DL*L-kr_13*DLL)
kr_16 = 0.0012; kf_16 = 0.05Reaction: ILL => DLL, Rate Law: comp1*(kf_16*ILL-kr_16*DLL)
kr_1 = 16000.0; kf_1 = 1.5E8Reaction: BL + L => BLL, Rate Law: comp1*(kf_1*BL*L-kr_1*BLL)
kf_11 = 20.0; kr_11 = 0.81Reaction: ALL => ILL, Rate Law: comp1*(kf_11*ALL-kr_11*ILL)
kf_14 = 0.05; kr_14 = 0.0012Reaction: I => D, Rate Law: comp1*(kf_14*I-kr_14*D)
kr_12 = 4.0; kf_12 = 3.0E8Reaction: D + L => DL, Rate Law: comp1*(kf_12*D*L-kr_12*DL)
kr_15 = 0.0012; kf_15 = 0.05Reaction: IL => DL, Rate Law: comp1*(kf_15*IL-kr_15*DL)
kf_0 = 3.0E8; kr_0 = 8000.0Reaction: B + L => BL, Rate Law: comp1*(kf_0*B*L-kr_0*BL)
kf_9 = 19.7; kr_9 = 3.74Reaction: A => I, Rate Law: comp1*(kf_9*A-kr_9*I)
kf_10 = 19.85; kr_10 = 1.74Reaction: AL => IL, Rate Law: comp1*(kf_10*AL-kr_10*IL)
kf_8 = 1.5E8; kr_8 = 8.0Reaction: IL + L => ILL, Rate Law: comp1*(kf_8*IL*L-kr_8*ILL)
kr_7 = 4.0; kf_7 = 3.0E8Reaction: I + L => IL, Rate Law: comp1*(kf_7*I*L-kr_7*IL)

States:

NameDescription
BLL[IPR002394; acetylcholine-gated channel complex]
DLL[acetylcholine-gated channel complex]
A[acetylcholine-gated channel complex]
BL[IPR002394; acetylcholine-gated channel complex]
D[IPR002394; acetylcholine-gated channel complex]
L[acetylcholine; Acetylcholine]
ALL[IPR002394; acetylcholine-gated channel complex]
B[IPR002394; acetylcholine-gated channel complex]
I[IPR002394; acetylcholine-gated channel complex]
AL[IPR002394; acetylcholine-gated channel complex]
DL[IPR002394; acetylcholine-gated channel complex]
ILL[IPR002394; acetylcholine-gated channel complex]
IL[IPR002394; acetylcholine-gated channel complex]

Eftimie2010 - immunity to melanoma: BIOMD0000000768v0.0.1

The paper describes a model of immunity to melanoma. Created by COPASI 4.25 (Build 207) This model is described in…

Details

Recent experiments indicate that CD4(+) Th2 cells can reject skin tumors in mice, while CD4(+) Th1 cells cannot (Mattes et al., 2003; Zhang et al., 2009). These results are surprising because CD4(+) Th1 cells are typically considered to be capable of tumor rejection. We used mathematical models to investigate this unexpected outcome. We found that neither CD4(+) Th1 nor CD4(+) Th2 cells could eliminate the cancer cells when acting alone, but that tumor elimination could be induced by recruitment of eosinophils by the Th2 cells. These recruited eosinophils had unexpected indirect effects on the decay rate of type 2 cytokines and the rate at which Th2 cells are inactivated through interactions with cancer cells. Strikingly, the presence of eosinophils impacted tumor growth more significantly than the release of tumor-suppressing cytokines such as IFN-gamma and TNF-alpha. Our simulations suggest that novel strategies to enhance eosinophil recruitment into skin tumors may improve cancer immunotherapies. link: http://identifiers.org/pubmed/20450922

Parameters:

NameDescription
cth = 0.1 1/d; f = 0.0 1Reaction: Th =>, Rate Law: tme*cth*f
atum = 0.514 1/d; kp = 1.0 1; ktum = 1.02E-9 1; ks = 1.0 1Reaction: => T; Cp, Cs, Rate Law: tme*atum*T*(1+kp*Cp)*(1-ktum*T)/(1+ks*Cs)
h2 = 1000.0 1; i31 = 3.8E-4 1/d; c = 1.0 1/dReaction: => Cp; Th, T, Rate Law: tme*(i31*Th+c)*T/(h2+T)
kp = 1.0 1; kth = 1.0E-8 1; bth = 0.09 1/dReaction: => Th; Cp, Rate Law: tme*bth*Th*(1-kth*Th)/(1+kp*Cp)
dth = 1.0E-7 1/dReaction: Th => ; T, Rate Law: tme*dth*T*Th
kp = 1.0 1; h2 = 1000.0 1; i21 = 8.6 1/d; c = 1.0 1/dReaction: => Cs; Th, T, Cp, Rate Law: tme*(i21*Th+c)*T/((h2+T)*(1+kp*Cp))
j0 = 34.0 1/dReaction: C =>, Rate Law: tme*j0*C
jts = 34.0 1/dReaction: Cs =>, Rate Law: tme*jts*Cs
kp = 1.0 1; ath = 0.008 1/d; h2 = 1000.0 1Reaction: => Th; C, T, Cp, Rate Law: tme*ath*C*T/((h2+T)*(1+kp*Cp))
h1 = 1000000.0 1; i3t = 10.0 1/dReaction: => Cp; T, Rate Law: tme*i3t*T^2/(h1^2+T^2)
kp = 1.0 1; h2 = 1000.0 1; i11 = 5.4 1/d; c = 1.0 1/dReaction: => C; Th, T, Cp, Rate Law: tme*(i11*Th+c)*T/((h2+T)*(1+kp*Cp))
h0 = 100000.0 1; gtum = 0.2 1/dReaction: T => ; Cs, Rate Law: tme*gtum*Cs*T/(h0+T)
jtp = 34.0 1/dReaction: Cp =>, Rate Law: tme*jtp*Cp

States:

NameDescription
Th[helper T cell]
Cs[Cytokine]
T[malignant cell]
C[Cytokine]
Cp[Cytokine]

Eftimie2017/1 - interaction of Th and macrophage: BIOMD0000000770v0.0.1

The paper describes a model of interaction of Th cells and macrophage in melanoma. Created by COPASI 4.25 (Build 207)…

Details

It is generally accepted that tumour cells can be eliminated by M1 anti-tumour macrophages and CD8+ T cells. However, experimental results over the past 10-15 years have shown that B16 mouse melanoma cells can be eliminated by the CD4+ T cells alone (either Th1 or Th2 sub-types), in the absence of CD8+ T cells. In some studies, elimination of B16 melanoma was associated with a Th1 immune response (i.e., elimination occurred in the presence of cytokines produced by Th1 cells), while in other studies melanoma elimination was associated with a Th2 immune response (i.e., elimination occurred in the presence of cytokines produced by Th2 cells). Moreover, macrophages have been shown to be present inside the tumours, during both Th1 and Th2 immune responses. To investigate the possible biological mechanisms behind these apparently contradictory results, we develop a class of mathematical models for the dynamics of Th1 and Th2 cells, and M1 and M2 macrophages in the presence/absence of tumour cells. Using this mathematical model, we show that depending on the re-polarisation rates between M1 and M2 macrophages, we obtain tumour elimination in the presence of a type-I immune response (i.e., more Th1 and M1 cells, compared to the Th2 and M2 cells), or in the presence of a type-II immune response (i.e., more Th2 and M2 cells). Moreover, tumour elimination is also possible in the presence of a mixed type-I/type-II immune response. Tumour growth always occurs in the presence of a type-II immune response, as observed experimentally. Finally, tumour dormancy is the result of a delicate balance between the pro-tumour effects of M2 cells and the anti-tumour effects of M1 and Th1 cells. link: http://identifiers.org/pubmed/28219660

Parameters:

NameDescription
am1 = 0.001 1/dReaction: => M1; H1, Rate Law: tme*am1*H1
em2 = 0.02 1/dReaction: M2 =>, Rate Law: tme*em2*M2
em1 = 0.02 1/dReaction: M1 =>, Rate Law: tme*em1*M1
ah1 = 0.008 1/dReaction: => H1; M1, Rate Law: tme*ah1*M1
rm1 = 0.09 1/dReaction: M2 => M1, Rate Law: tme*rm1*M2
pm1 = 0.02 1/d; m2 = 1.0E9 1Reaction: => M1; M2, Rate Law: tme*pm1*M1*(1-(M1+M2)/m2)
pm2 = 0.02 1/d; m2 = 1.0E9 1Reaction: => M2; H2, M1, Rate Law: tme*pm2*M2*H2*(1-(M2+M1)/m2)
m1 = 1.0E8 1; ph2 = 0.09 1/dReaction: => H2; M2, H1, Rate Law: tme*ph2*H2*M2*(1-(H2+H1)/m1)
m1 = 1.0E8 1; ph1 = 0.09 1/dReaction: => H1; M1, H2, Rate Law: tme*ph1*H1*M1*(1-(H1+H2)/m1)
am2 = 0.001 1/dReaction: => M2; H2, Rate Law: tme*am2*H2
eh2 = 0.03 1/dReaction: H2 =>, Rate Law: tme*eh2*H2
ah2 = 0.001 1/dReaction: => H2; M2, Rate Law: tme*ah2*M2
eh1 = 0.03 1/dReaction: H1 =>, Rate Law: tme*eh1*H1
rm2 = 0.05 1/dReaction: M1 => M2, Rate Law: tme*rm2*M1

States:

NameDescription
H1[T-helper 1 cell]
M2[M2 Macrophage]
M1[M1 Macrophage]
H2[T-helper 2 cell]

Eftimie2017/2 - interaction of Th and macrophage in melanoma: BIOMD0000000769v0.0.1

The paper describes a model of interaction of Th cells and macrophage in melanoma. Created by COPASI 4.25 (Build 207)…

Details

It is generally accepted that tumour cells can be eliminated by M1 anti-tumour macrophages and CD8+ T cells. However, experimental results over the past 10-15 years have shown that B16 mouse melanoma cells can be eliminated by the CD4+ T cells alone (either Th1 or Th2 sub-types), in the absence of CD8+ T cells. In some studies, elimination of B16 melanoma was associated with a Th1 immune response (i.e., elimination occurred in the presence of cytokines produced by Th1 cells), while in other studies melanoma elimination was associated with a Th2 immune response (i.e., elimination occurred in the presence of cytokines produced by Th2 cells). Moreover, macrophages have been shown to be present inside the tumours, during both Th1 and Th2 immune responses. To investigate the possible biological mechanisms behind these apparently contradictory results, we develop a class of mathematical models for the dynamics of Th1 and Th2 cells, and M1 and M2 macrophages in the presence/absence of tumour cells. Using this mathematical model, we show that depending on the re-polarisation rates between M1 and M2 macrophages, we obtain tumour elimination in the presence of a type-I immune response (i.e., more Th1 and M1 cells, compared to the Th2 and M2 cells), or in the presence of a type-II immune response (i.e., more Th2 and M2 cells). Moreover, tumour elimination is also possible in the presence of a mixed type-I/type-II immune response. Tumour growth always occurs in the presence of a type-II immune response, as observed experimentally. Finally, tumour dormancy is the result of a delicate balance between the pro-tumour effects of M2 cells and the anti-tumour effects of M1 and Th1 cells. link: http://identifiers.org/pubmed/28219660

Parameters:

NameDescription
gm2 = 2.3E-10 1/dReaction: => T; M2, Rate Law: tme*gm2*T*M2
nm2 = 1.0E-7 1/dReaction: => M2; T, Rate Law: tme*nm2*M2*T
em1 = 0.02 1/dReaction: M1 =>, Rate Law: tme*em1*M1
gh1 = 4.8E-9 1/dReaction: T => ; H1, Rate Law: tme*gh1*T*H1
pm1 = 0.02 1/d; m2 = 1.0E9 1Reaction: => M1; M2, Rate Law: tme*pm1*M1*(1-(M1+M2)/m2)
m1 = 1.0E8 1; ph1 = 0.09 1/dReaction: => H1; M1, H2, Rate Law: tme*ph1*H1*M1*(1-(H1+H2)/m1)
am2 = 0.001 1/dReaction: => M2; H2, Rate Law: tme*am2*H2
eh2 = 0.03 1/dReaction: H2 =>, Rate Law: tme*eh2*H2
ah2 = 0.001 1/dReaction: => H2; M2, Rate Law: tme*ah2*M2
nh2 = 1.0E-7 1/dReaction: H2 => ; T, Rate Law: tme*nh2*H2*T
nm1 = 1.0E-7 1/dReaction: M1 => ; T, Rate Law: tme*nm1*M1*T
eh1 = 0.03 1/dReaction: H1 =>, Rate Law: tme*eh1*H1
gm1 = 6.0E-9 1/dReaction: T => ; M1, Rate Law: tme*gm1*T*M1
nh1 = 1.0E-7 1/dReaction: H1 => ; T, Rate Law: tme*nh1*H1*T
am1 = 0.001 1/dReaction: => M1; H1, Rate Law: tme*am1*H1
em2 = 0.02 1/dReaction: M2 =>, Rate Law: tme*em2*M2
ah1 = 0.008 1/dReaction: => H1; M1, Rate Law: tme*ah1*M1
rm1 = 0.09 1/dReaction: M2 => M1, Rate Law: tme*rm1*M2
b = 1.0E9 1; a = 0.69 1/dReaction: => T, Rate Law: tme*a*T*(1-T/b)
pm2 = 0.02 1/d; m2 = 1.0E9 1Reaction: => M2; H2, M1, Rate Law: tme*pm2*M2*H2*(1-(M2+M1)/m2)
m1 = 1.0E8 1; ph2 = 0.09 1/dReaction: => H2; M2, H1, Rate Law: tme*ph2*H2*M2*(1-(H2+H1)/m1)
gh2 = 1.0E-9 1/dReaction: T => ; H2, Rate Law: tme*gh2*T*H2
f = 1.0E-8 1/dReaction: T =>, Rate Law: tme*f*T
rm2 = 0.05 1/dReaction: M1 => M2, Rate Law: tme*rm2*M1

States:

NameDescription
H1[T-helper 1 cell]
T[malignant cell]
M2[M2 Macrophage]
M1[M1 Macrophage]
H2[T-helper 2 cell]

Eftimie2018 - Cancer and Immune biomarkers: BIOMD0000000741v0.0.1

The paper describes a model on the detection of cancer based on cancer and immune biomarkers. Created by COPASI 4.25 (B…

Details

Background: Early cancer diagnosis is one of the most important challenges of cancer research, since in many can- cers it can lead to cure for patients with early stage diseases. For epithelial ovarian cancer (which is the leading cause of death among gynaecologic malignancies) the classical detection approach is based on measurements of CA-125 biomarker. However, the poor sensitivity and specificity of this biomarker impacts the detection of early-stage cancers. Methods: Here we use a computational approach to investigate the effect of combining multiple biomarkers for ovarian cancer (e.g., CA-125 and IL-7), to improve early cancer detection. Results: We show that this combined biomarkers approach could lead indeed to earlier cancer detection. However, the immune response (which influences the level of secreted IL-7 biomarker) plays an important role in improving and/or delaying cancer detection. Moreover, the detection level of IL-7 immune biomarker could be in a range that would not allow to distinguish between a healthy state and a cancerous state. In this case, the construction of solu- tion diagrams in the space generated by the IL-7 and CA-125 biomarkers could allow us predict the long-term evolu- tion of cancer biomarkers, thus allowing us to make predictions on cancer detection times. Conclusions: Combining cancer and immune biomarkers could improve cancer detection times, and any predic- tions that could be made (at least through the use of CA-125/IL-7 biomarkers) are patient specific. Keywords: Ovarian cancer, Mathematical model, CA-125 biomarker, IL-7 biomarker, Cancer detection times link: http://identifiers.org/doi/10.1186/s12967-018-1432-8

Parameters:

NameDescription
kei = 2.14 1/ksReaction: BI =>, Rate Law: compartment*kei*BI
fhirhinh = 19548.0 1/ksReaction: => BI, Rate Law: compartment*fhirhinh
ft = 0.1 1; rt = 4.5E-5 1/ksReaction: => BT; NT, Rate Law: compartment*ft*rt*NT
kgr = 0.00578 1/ksReaction: => NT, Rate Law: compartment*kgr*NT
fhtrhtnh = 4560.0 1/ksReaction: => BT, Rate Law: compartment*fhtrhtnh
M = 1.0E9 1; ai = 2.0794 1/ksReaction: => NI; NT, Rate Law: compartment*ai*NT*(1-NI/M)
firi = 1.0925E-6 1/ksReaction: => BI; NI, Rate Law: compartment*firi*NI
hi = 1000.0 1; dt = 1.0E-6 1/ksReaction: NT => ; NI, Rate Law: compartment*dt*NT*NI/(hi+NI)
di = 0.4 1/ksReaction: NI =>, Rate Law: compartment*di*NI
ket = 0.11 1/ksReaction: BT =>, Rate Law: compartment*ket*BT

States:

NameDescription
BT[Mucin-16; Biomarker]
NT[malignant cell; Malignant Cell]
BI[Interleukin-7; Biomarker]
NI[lymphocyte; Lymphocyte]

Eftimie2019-Macrophages Plasticity: BIOMD0000000806v0.0.1

This paper describes the complex interactions between two extreme types of macrophages (M1 and M2 cells), effector T cel…

Details

Over the last few years, oncolytic virus therapy has been recognised as a promising approach in cancer treatment, due to the potential of these viruses to induce systemic anti-tumour immunity and selectively killing tumour cells. However, the effectiveness of these viruses depends significantly on their interactions with the host immune responses, both innate (e.g., macrophages, which accumulate in high numbers inside solid tumours) and adaptive (e.g., [Formula: see text] T cells). In this article, we consider a mathematical approach to investigate the possible outcomes of the complex interactions between two extreme types of macrophages (M1 and M2 cells), effector [Formula: see text] T cells and an oncolytic Vesicular Stomatitis Virus (VSV), on the growth/elimination of B16F10 melanoma. We discuss, in terms of VSV, [Formula: see text] and macrophages levels, two different types of immune responses which could ensure tumour control and eventual elimination. We show that both innate and adaptive anti-tumour immune responses, as well as the oncolytic virus, could be very important in delaying tumour relapse and eventually eliminating the tumour. Overall this study supports the use mathematical modelling to increase our understanding of the complex immune interaction following oncolytic virotherapies. However, the complexity of the model combined with a lack of sufficient data for model parametrisation has an impact on the possibility of making quantitative predictions. link: http://identifiers.org/pubmed/31410657

Parameters:

NameDescription
delta_i = 0.475 1/ms; b = 2500.0Reaction: => Virus_Xv; Infected_Tumour_Cells_Xi, Rate Law: compartment*delta_i*b*Infected_Tumour_Cells_Xi
d_em2 = 0.2 1/msReaction: M2_Macrophage_Xm2 =>, Rate Law: compartment*d_em2*M2_Macrophage_Xm2
v_d_m1 = 1.5 1/ms; h_m = 1000.0Reaction: Infected_Tumour_Cells_Xi => ; Infected_Tumour_Cells_Xi, M1_Macrophage_Xm1, M2_Macrophage_Xm2, Rate Law: compartment*v_d_m1*Infected_Tumour_Cells_Xi*M1_Macrophage_Xm1/(h_m+M2_Macrophage_Xm2)
d_em1 = 0.2Reaction: M1_Macrophage_Xm1 =>, Rate Law: compartment*d_em1*M1_Macrophage_Xm1
h_e = 1.0; v_d_u = 4.4 1/msReaction: Infected_Tumour_Cells_Xi => ; Infected_Tumour_Cells_Xi, Effector_Cytotoxic_CD8_TCells__Xe, Rate Law: compartment*v_d_u*Infected_Tumour_Cells_Xi*Effector_Cytotoxic_CD8_TCells__Xe/(h_e+Effector_Cytotoxic_CD8_TCells__Xe)
omega = 2.0 1/msReaction: Virus_Xv => ; Virus_Xv, Rate Law: compartment*omega*Virus_Xv
r = 0.924 1/ms; K = 3.3E9 1Reaction: => UnInfected_Tumour_Cells_Xu, Rate Law: compartment*r*UnInfected_Tumour_Cells_Xu*(1-UnInfected_Tumour_Cells_Xu/K)
p_e = 2070.0 1/ms; h_m = 1000.0Reaction: => Effector_Cytotoxic_CD8_TCells__Xe; M1_Macrophage_Xm1, M2_Macrophage_Xm2, Rate Law: compartment*p_e*M1_Macrophage_Xm1/(h_m+M2_Macrophage_Xm2)
p_m2 = 0.22 1/ms; M = 1.0E8Reaction: => M2_Macrophage_Xm2; M1_Macrophage_Xm1, Rate Law: compartment*p_m2*M2_Macrophage_Xm2*(1-(M1_Macrophage_Xm1+M2_Macrophage_Xm2)/M)
u_a_1 = 3.0E-6 1/msReaction: => M1_Macrophage_Xm1; UnInfected_Tumour_Cells_Xu, Rate Law: compartment*u_a_1*UnInfected_Tumour_Cells_Xu
d_u = 0.44; h_e = 1.0Reaction: UnInfected_Tumour_Cells_Xu => ; Effector_Cytotoxic_CD8_TCells__Xe, Rate Law: compartment*d_u*UnInfected_Tumour_Cells_Xu*Effector_Cytotoxic_CD8_TCells__Xe/(h_e+Effector_Cytotoxic_CD8_TCells__Xe)
v_h_u = 100000.0; d_v = 0.011Reaction: UnInfected_Tumour_Cells_Xu => Infected_Tumour_Cells_Xi; Virus_Xv, Rate Law: compartment*d_v*Virus_Xv*UnInfected_Tumour_Cells_Xu/(v_h_u+UnInfected_Tumour_Cells_Xu)
p_m1 = 0.22 1/ms; M = 1.0E8Reaction: => M1_Macrophage_Xm1; M2_Macrophage_Xm2, Rate Law: compartment*p_m1*M1_Macrophage_Xm1*(1-(M1_Macrophage_Xm1+M2_Macrophage_Xm2)/M)
d_t = 1.0E-10 1/msReaction: Effector_Cytotoxic_CD8_TCells__Xe => ; UnInfected_Tumour_Cells_Xu, Rate Law: compartment*d_t*UnInfected_Tumour_Cells_Xu*Effector_Cytotoxic_CD8_TCells__Xe
o_r_m1 = 0.001 1/ms; u_r_m1 = 4.0 1/ms; h_u = 5.0E9Reaction: M1_Macrophage_Xm1 => M2_Macrophage_Xm2; UnInfected_Tumour_Cells_Xu, Rate Law: compartment*M1_Macrophage_Xm1*(o_r_m1+u_r_m1*UnInfected_Tumour_Cells_Xu/(h_u+UnInfected_Tumour_Cells_Xu))
d_ee = 0.4 1/msReaction: Effector_Cytotoxic_CD8_TCells__Xe =>, Rate Law: compartment*d_ee*Effector_Cytotoxic_CD8_TCells__Xe
delta_i = 0.475 1/msReaction: Infected_Tumour_Cells_Xi =>, Rate Law: compartment*delta_i*Infected_Tumour_Cells_Xi
h_m = 1000.0; d_m1 = 0.01Reaction: UnInfected_Tumour_Cells_Xu => ; M1_Macrophage_Xm1, M2_Macrophage_Xm2, Rate Law: compartment*d_m1*UnInfected_Tumour_Cells_Xu*M1_Macrophage_Xm1/(h_m+M2_Macrophage_Xm2)
v_a_1 = 1.0E-6 1/msReaction: => M1_Macrophage_Xm1; Infected_Tumour_Cells_Xi, Virus_Xv, Rate Law: compartment*v_a_1*(Infected_Tumour_Cells_Xi+Virus_Xv)
d_m2 = 0.4; h_m = 1000.0Reaction: => UnInfected_Tumour_Cells_Xu; M2_Macrophage_Xm2, Rate Law: compartment*d_m2*UnInfected_Tumour_Cells_Xu*M2_Macrophage_Xm2/(h_m+M2_Macrophage_Xm2)
u_a_2 = 4.0E-8 1/msReaction: => M2_Macrophage_Xm2; UnInfected_Tumour_Cells_Xu, Rate Law: compartment*u_a_2*UnInfected_Tumour_Cells_Xu
H = 0.0Reaction: => Virus_Xv, Rate Law: compartment*H
h_v = 0.105636; v_r_m2 = 0.5 1/ms; o_r_m2 = 0.001 1/msReaction: M2_Macrophage_Xm2 => M1_Macrophage_Xm1; Virus_Xv, Rate Law: compartment*M2_Macrophage_Xm2*(o_r_m2+v_r_m2*Virus_Xv/(h_v+Virus_Xv))

States:

NameDescription
Effector Cytotoxic CD8 TCells Xe[CD8-positive, alpha-beta cytotoxic T cell]
M1 Macrophage Xm1[M1 Macrophage]
M2 Macrophage Xm2[M2 Macrophage]
UnInfected Tumour Cells Xu[cancer; B16-F10 cell]
Infected Tumour Cells Xi[B16-F10 cell; EFO:0000311; infected cell]
Virus Xv[Vesicular stomatitis virus]

Egli2004_ProlactinRhythmicSecretion: MODEL0912452142v0.0.1

This a model from the article: Rhythmic secretion of prolactin in rats: action of oxytocin coordinated by vasoactive i…

Details

Prolactin (PRL) is secreted from lactotrophs of the anterior pituitary gland of rats in a unique pattern in response to uterine cervical stimulation (CS) during mating. Surges of PRL secretion occur in response to relief from hypothalamic dopaminergic inhibition and stimulation by hypothalamic releasing neurohormones. In this study, we characterized the role of oxytocin (OT) in this system and the involvement of vasoactive intestinal polypeptide (VIP) from the suprachiasmatic nucleus (SCN) in controlling OT and PRL secretion of CS rats. The effect of OT on PRL secretion was demonstrated in cultured lactotrophs showing simultaneous enhanced secretion rate and increased intracellular Ca(2+). Neurosecretory OT cells of the hypothalamic paraventricular nucleus that express VIP receptors were identified by using immunocytochemical techniques in combination with the retrogradely transported neuronal tracer Fluoro-Gold (iv injected). OT measurements of serial blood samples obtained from ovariectomized (OVX) CS rats displayed a prominent increase at the time of the afternoon PRL peak. The injection of VIP antisense oligonucleotides into the SCN abolished the afternoon increase of OT and PRL in CS-OVX animals. These findings suggest that VIP from the SCN contributes to the regulation of OT and PRL secretion in CS rats. We propose that in CS rats the regulatory mechanism(s) for PRL secretion comprise coordinated action of neuroendocrine dopaminergic and OT cells, both governed by the daily rhythm of VIP-ergic output from the SCN. This hypothesis is illustrated with a mathematical model. link: http://identifiers.org/pubmed/15033917

Ehrenstein1997 - The choline-leakage hypothesis in Alzheimer's disease: BIOMD0000000553v0.0.1

Ehrenstein1997 - The choline-leakage hypothesis in Alzheimer's diseaseThis model is described in the article: [The chol…

Details

We present a hypothesis for the loss of acetylcholine in Alzheimer's disease that is based on two recent experimental results: that beta-amyloid causes leakage of choline across cell membranes and that decreased production of acetylcholine increases the production of beta-amyloid. According to the hypothesis, an increase in beta-amyloid concentration caused by proteolysis of the amyloid precursor protein results in an increase in the leakage of choline out of cells. This leads to a reduction in intracellular choline concentration and hence a reduction in acetylcholine production. The reduction in acetylcholine production, in turn, causes an increase in the concentration of beta-amyloid. The resultant positive feedback between decreased acetylcholine and increased beta-amyloid accelerates the loss of acetylcholine. We compare the predictions of the choline-leakage hypothesis with a number of experimental observations. We also approximate it with a pair of ordinary differential equations. The solutions of these equations indicate that the loss of acetylcholine is very sensitive to the initial rate of beta-amyloid production. link: http://identifiers.org/pubmed/9284295

Parameters:

NameDescription
k1 = 0.007Reaction: a => ; b, a, b, Rate Law: Brain*k1*a*b
k2 = 0.33Reaction: => b, Rate Law: Brain*k2
k3 = 0.0042Reaction: b => ; a, a, Rate Law: Brain*k3*a
k4 = 0.01Reaction: b => ; b, Rate Law: Brain*k4*b

States:

NameDescription
b[Amyloid beta A4 protein]
aRel[acetylcholine]
a[acetylcholine]

Ehrenstein2000 - Positive-Feedback model for the loss of acetylcholine in Alzheimer's disease: BIOMD0000000552v0.0.1

Ehrenstein2000 - Positive-Feedback model for the loss of acetylcholine in Alzheimer's diseaseCurated model derived from…

Details

We describe a two-component positive-feedback system that could account for the large reduction of acetylcholine that is characteristic of patients with Alzheimer's disease (AD). One component is beta-amyloid-induced apoptosis of cholinergic cells, leading to a decrease in acetylcholine. The other component is an increase in the concentration of beta-amyloid in response to a decrease in acetylcholine. We describe each mechanism with a differential equation, and then solve the two equations numerically. The solution provides a description of the time course of the reduction of acetylcholine in AD patients that is consistent with epidemiological data. This model may also provide an explanation for the significant, but lesser, decrease of other neurotransmitters that is characteristic of AD. link: http://identifiers.org/pubmed/10863547

Parameters:

NameDescription
k1 = 0.007Reaction: a => ; b, a, b, Rate Law: Brain*k1*a*b
k2 = 0.33Reaction: => b, Rate Law: Brain*k2
k3 = 0.0042Reaction: b => ; a, a, Rate Law: Brain*k3*a
k4 = 0.01Reaction: b => ; b, Rate Law: Brain*k4*b

States:

NameDescription
b[Amyloid beta A4 protein]
aRel[acetylcholine]
a[acetylcholine]

Elowitz2000 - Repressilator: BIOMD0000000012v0.0.1

Elowitz2000 - RepressilatorThis model describes the deterministic version of the repressilator system. The authors of t…

Details

Networks of interacting biomolecules carry out many essential functions in living cells, but the 'design principles' underlying the functioning of such intracellular networks remain poorly understood, despite intensive efforts including quantitative analysis of relatively simple systems. Here we present a complementary approach to this problem: the design and construction of a synthetic network to implement a particular function. We used three transcriptional repressor systems that are not part of any natural biological clock to build an oscillating network, termed the repressilator, in Escherichia coli. The network periodically induces the synthesis of green fluorescent protein as a readout of its state in individual cells. The resulting oscillations, with typical periods of hours, are slower than the cell-division cycle, so the state of the oscillator has to be transmitted from generation to generation. This artificial clock displays noisy behaviour, possibly because of stochastic fluctuations of its components. Such 'rational network design may lead both to the engineering of new cellular behaviours and to an improved understanding of naturally occurring networks. link: http://identifiers.org/pubmed/10659856

Parameters:

NameDescription
k_tl = NaNReaction: => PX; X, Rate Law: k_tl*X
a0_tr = NaN; a_tr = NaN; n = 2.0; KM = 40.0Reaction: => X; PZ, Rate Law: a0_tr+a_tr*KM^n/(KM^n+PZ^n)
kd_prot = NaNReaction: PX =>, Rate Law: kd_prot*PX
kd_mRNA = NaNReaction: Z =>, Rate Law: kd_mRNA*Z

States:

NameDescription
Y[messenger RNA; RNA; Tetracycline repressor protein class B from transposon Tn10]
Z[messenger RNA; RNA; Repressor protein cI]
PY[Tetracycline repressor protein class B from transposon Tn10]
PX[Lactose operon repressor]
X[messenger RNA; RNA; Lactose operon repressor]
PZ[Repressor protein cI]

Endresen1997_SinoatrialActionPotential: MODEL0912388235v0.0.1

This a model from the article: Chaos in weakly-coupled pacemaker cells. Endresen LP. J Theor Biol 1997 Jan 7;184(1)…

Details

A model of the rabbit sinoatrial action potential is introduced, based on a model by Morris & Lecar. One cell is described by two nonlinear first-order ordinary differential equations, with ten constant parameters. The model is much simpler than most other models in use, but can reproduce perfectly experimentally recorded action potentials. The dynamics of two coupled cells, with and without the presence of periodic acetylcholine pulses, shows examples of bifurcations and strange attractors, mathematical phenomena characterizing chaotic motion. It remains to be clarified whether such dynamics is actually observed, for example in the small irregular variations of the normal heart rate. link: http://identifiers.org/pubmed/9039399

Erguler2013 - Unfolded protein stress response: BIOMD0000000446v0.0.1

Erguler2013 - Unfolded protein stress responseThe model investigates the mechanism by which UPR (unfolded protein respon…

Details

The unfolded protein response (UPR) is a major signalling cascade acting in the quality control of protein folding in the endoplasmic reticulum (ER). The cascade is known to play an accessory role in a range of genetic and environmental disorders including neurodegenerative and cardiovascular diseases, diabetes and kidney diseases. The three major receptors of the ER stress involved with the UPR, i.e. IRE1 α, PERK and ATF6, signal through a complex web of pathways to convey an appropriate response. The emerging behaviour ranges from adaptive to maladaptive depending on the severity of unfolded protein accumulation in the ER; however, the decision mechanism for the switch and its timing have so far been poorly understood.Here, we propose a mechanism by which the UPR outcome switches between survival and death. We compose a mathematical model integrating the three signalling branches, and perform a comprehensive bifurcation analysis to investigate possible responses to stimuli. The analysis reveals three distinct states of behaviour, low, high and intermediate activity, associated with stress adaptation, tolerance, and the initiation of apoptosis. The decision to adapt or destruct can, therefore, be understood as a dynamic process where the balance between the stress and the folding capacity of the ER plays a pivotal role in managing the delivery of the most appropriate response. The model demonstrates for the first time that the UPR is capable of generating oscillations in translation attenuation and the apoptotic signals, and this is supplemented with a Bayesian sensitivity analysis identifying a set of parameters controlling this behaviour.This work contributes largely to the understanding of one of the most ubiquitous signalling pathways involved in protein folding quality control in the metazoan ER. The insights gained have direct consequences on the management of many UPR-related diseases, revealing, in addition, an extended list of candidate disease modifiers. Demonstration of stress adaptation sheds light to how preconditioning might be beneficial in manifesting the UPR outcome to prevent untimely apoptosis, and paves the way to novel approaches for the treatment of many UPR-related conditions. link: http://identifiers.org/pubmed/23433609

Parameters:

NameDescription
kmbc = 0.03 acu1 = acu^-1; kfbc = 10.0 aru = acu.atu^-1Reaction: => BCL2T; CHOP, CHOP, Rate Law: kfbc/(1+kmbc*CHOP)
BiP = 0.0 acu; tmr = 10.0 dimensionless; UFP = 0.0 acu; kf = 10.0 aru2 = acu^-1.atu^-1Reaction: => BiUFP, Rate Law: tmr*kf*BiP*UFP
kdATF4 = 0.1 aru1 = atu^-1Reaction: ATF4 => ; ATF4, Rate Law: kdATF4*ATF4
BH3 = 0.0 acu; kas3 = 10.0 aru2 = acu^-1.atu^-1; BCL2 = 0.0 acuReaction: => BH3BCL2, Rate Law: kas3*BH3*BCL2
kdBiP = 0.01 aru1 = atu^-1Reaction: BiPT => ; BiPT, Rate Law: kdBiP*BiPT
kdXS = 0.1 aru1 = atu^-1Reaction: Xbp1s => ; Xbp1s, Rate Law: kdXS*Xbp1s
kff = 10.0 aru3 = acu^-3.atu^-1; IRE1 = 0.0 acu; tmr = 10.0 dimensionless; n = 4.0 dimensionlessReaction: => IRE1A, Rate Law: tmr*kff*IRE1^n
ktrATF6 = 1.0 aru1 = atu^-1; mATF6T = 5.0 acuReaction: => ATF6T, Rate Law: ktrATF6*mATF6T
kbu = 0.0 aru2 = acu^-1.atu^-1Reaction: UFPT => ; BiUFP, BiUFP, UFPT, Rate Law: kbu*BiUFP*UFPT
kdsx = 0.05 aru1 = atu^-1Reaction: BAXmBCL2 => ; BAXmBCL2, Rate Law: kdsx*BAXmBCL2
kdATF6p50 = 0.1 aru1 = atu^-1Reaction: ATF6p50 => ; ATF6p50, Rate Law: kdATF6p50*ATF6p50
BAXm = 0.0 acu; BCL2 = 0.0 acu; kasx = 90.0 aru2 = acu^-1.atu^-1Reaction: => BAXmBCL2, Rate Law: kasx*BAXm*BCL2
ktrUFP = 1.0 aru1 = atu^-1; extATT = 0.0 dimensionless; mUFPT = 0.0 acu; eIF2aT = 1.0 acu; eIF2a = 0.0 acuReaction: => UFPT, Rate Law: ktrUFP*mUFPT*piecewise(eIF2a/eIF2aT, extATT == 1, 1)
kmXbp = 10.0 dimensionless; krcBiP = 5.0 acu; trcBiP = 1.0 aru = acu.atu^-1; basalBiP = 1.0 acu; kmAtfsBiP = 1.0 dimensionlessReaction: => mBiPT; Xbp1s, ATF6p50, Xbp1s, ATF6p50, Rate Law: trcBiP*(basalBiP+kmXbp*Xbp1s+kmAtfsBiP*ATF6p50)/(krcBiP+basalBiP+kmXbp*Xbp1s+kmAtfsBiP*ATF6p50)
ktrans = 1.0 aru1 = atu^-1; ATF6 = 0.0 acuReaction: ATF6T => ATF6GB, Rate Law: ktrans*ATF6
kff = 10.0 aru3 = acu^-3.atu^-1; switch = 0.0 dimensionless; PERK = 0.0 acu; tmr = 10.0 dimensionless; n = 4.0 dimensionless; UFP = 0.0 acuReaction: => PERKA, Rate Law: tmr*kff*piecewise(UFP, switch == 1, 1)*PERK^n
BH3 = 0.0 acu; kfxp = 3.0 aru2 = acu^-1.atu^-1Reaction: BAXmT => ; BAXmT, Rate Law: kfxp*BH3*BAXmT
kdmBiP = 1.0 aru1 = atu^-1Reaction: mBiPT => ; mBiPT, Rate Law: kdmBiP*mBiPT
kdGADD34 = 0.1 aru1 = atu^-1Reaction: GADD34 => ; GADD34, Rate Law: kdGADD34*GADD34
ktrCHOP = 1.0 aru1 = atu^-1Reaction: => CHOP; mCHOP, mCHOP, Rate Law: ktrCHOP*mCHOP
extPERK = 0.0 acu1 = acu^-1; kr = 1.0 aru1 = atu^-1; tmr = 10.0 dimensionless; UFP = 0.0 acuReaction: PERKA => ; PERKA, Rate Law: tmr*kr*PERKA/(1+extPERK*UFP)
kdATF6 = 0.1 aru1 = atu^-1Reaction: ATF6T => ; ATF6T, Rate Law: kdATF6*ATF6T
kfx = 1.0 aru1 = atu^-1Reaction: BAXmT => ; BAXmT, Rate Law: kfx*BAXmT
spliceRate = 0.0 aru = acu.atu^-1Reaction: mXbp1u => mXbp1s, Rate Law: spliceRate
kdCHOP = 0.1 aru1 = atu^-1Reaction: CHOP => ; CHOP, Rate Law: kdCHOP*CHOP
BiP = 0.0 acu; IRE1 = 0.0 acu; tmr = 10.0 dimensionless; kf = 10.0 aru2 = acu^-1.atu^-1Reaction: => BiRE1, Rate Law: tmr*kf*BiP*IRE1
krcGADD34 = 1.0 acu; kmChop = 0.05 dimensionless; trcGADD34 = 1.0 aru = acu.atu^-1Reaction: => mGADD34; CHOP, CHOP, Rate Law: trcGADD34*kmChop*CHOP/(krcGADD34+kmChop*CHOP)
kdmWFS = 1.0 aru1 = atu^-1Reaction: mWFS1 => ; mWFS1, Rate Law: kdmWFS*mWFS1
kds3 = 0.01 aru1 = atu^-1Reaction: BH3BCL2 => ; BH3BCL2, Rate Law: kds3*BH3BCL2
kr = 1.0 aru1 = atu^-1; tmr = 10.0 dimensionlessReaction: BiUFP => ; BiUFP, Rate Law: tmr*kr*BiUFP
ks3 = 0.1 aru = acu.atu^-1Reaction: => BH3T, Rate Law: ks3
ktrATF4 = 1.0 aru1 = atu^-1; kATF4 = 0.1 acu; mATF4 = 1.0 acu; nh = 2.0 dimensionless; eIF2a = 0.0 acuReaction: => ATF4, Rate Law: ktrATF4*mATF4/(1+(eIF2a/kATF4)^nh)
ktrBiP = 1.0 aru1 = atu^-1Reaction: => BiPT; mBiPT, mBiPT, Rate Law: ktrBiP*mBiPT
kdWFS = 0.1 aru1 = atu^-1Reaction: WFS1 => ; WFS1, Rate Law: kdWFS*WFS1
kdATF6GB = 0.1 aru1 = atu^-1Reaction: ATF6GB => ; ATF6GB, Rate Law: kdATF6GB*ATF6GB
ktrWFS = 1.0 aru1 = atu^-1Reaction: => WFS1; mWFS1, mWFS1, Rate Law: ktrWFS*mWFS1
ks3p = 0.6 aru1 = atu^-1; kstr = 0.2 dimensionlessReaction: => BH3T; CHOP, CHOP, Rate Law: ks3p*kstr*CHOP
BAXT = 100.0 acu; BH3 = 0.0 acu; kfxp = 3.0 aru2 = acu^-1.atu^-1Reaction: => BAXmT, Rate Law: kfxp*BH3*BAXT
krcWFS = 1.0 acu; trcWFS = 1.0 aru = acu.atu^-1Reaction: => mWFS1; ATF6p50, ATF6p50, Rate Law: trcWFS*ATF6p50/(krcWFS+ATF6p50)
BiP = 0.0 acu; tmr = 10.0 dimensionless; ATF6 = 0.0 acu; kf = 10.0 aru2 = acu^-1.atu^-1Reaction: => BiATF, Rate Law: tmr*kf*BiP*ATF6
BAXT = 100.0 acu; kfx = 1.0 aru1 = atu^-1Reaction: => BAXmT, Rate Law: kfx*BAXT
ktrXS = 1.0 aru1 = atu^-1Reaction: => Xbp1s; mXbp1s, mXbp1s, Rate Law: ktrXS*mXbp1s
kdmCHOP = 1.0 aru1 = atu^-1Reaction: mCHOP => ; mCHOP, Rate Law: kdmCHOP*mCHOP
kbx = 2.0 aru1 = atu^-1Reaction: BAXmT => ; BAXmT, Rate Law: kbx*BAXmT
kdeAW = 1.0 aru2 = acu^-1.atu^-1Reaction: ATF6T => ; WFS1, WFS1, ATF6T, Rate Law: kdeAW*WFS1*ATF6T
ktrGADD34 = 1.0 aru1 = atu^-1Reaction: => GADD34; mGADD34, mGADD34, Rate Law: ktrGADD34*mGADD34
kdmGADD34 = 1.0 aru1 = atu^-1Reaction: mGADD34 => ; mGADD34, Rate Law: kdmGADD34*mGADD34
kcleave = 10.0 aru1 = atu^-1Reaction: ATF6GB => ATF6p50; ATF6GB, Rate Law: kcleave*ATF6GB
kd3 = 0.01 aru1 = atu^-1Reaction: BH3T => ; BH3T, Rate Law: kd3*BH3T
krcCHOP = 1.0 acu; kmAtff = 0.05 dimensionless; kmAtfs = 0.1 dimensionless; trcCHOP = 1.0 aru = acu.atu^-1Reaction: => mCHOP; ATF4, ATF6p50, ATF4, ATF6p50, Rate Law: trcCHOP*(kmAtff*ATF4+kmAtfs*ATF6p50)/(krcCHOP+kmAtff*ATF4+kmAtfs*ATF6p50)
BiP = 0.0 acu; PERK = 0.0 acu; tmr = 10.0 dimensionless; kf = 10.0 aru2 = acu^-1.atu^-1Reaction: => BiPER, Rate Law: tmr*kf*BiP*PERK
kdbc = 0.1 aru1 = atu^-1Reaction: BCL2T => ; BCL2T, Rate Law: kdbc*BCL2T
kdmXS = 1.0 aru1 = atu^-1Reaction: mXbp1s => ; mXbp1s, Rate Law: kdmXS*mXbp1s
kdUFP = 0.1 aru1 = atu^-1Reaction: UFPT => ; UFPT, Rate Law: kdUFP*UFPT
krcXU = 5.0 acu; trcXU = 1.0 aru = acu.atu^-1; kmAtfsXBP = 10.0 dimensionless; basalXBP = 1.0 acuReaction: => mXbp1u; ATF6p50, ATF6p50, Rate Law: trcXU*(basalXBP+kmAtfsXBP*ATF6p50)/(krcXU+basalXBP+kmAtfsXBP*ATF6p50)
kdmXU = 1.0 aru1 = atu^-1Reaction: mXbp1u => ; mXbp1u, Rate Law: kdmXU*mXbp1u

States:

NameDescription
mWFS1mWFS1
UFPTUFPT
ATF6TATF6T
ATF4[Cyclic AMP-dependent transcription factor ATF-4]
BiPERBiPER
BCL2TBCL2T
IRE1AIRE1A
BiUFPBiUFP
WFS1[Wolframin]
CHOP[DNA damage-inducible transcript 3 protein]
BH3TBH3T
mXbp1umXbp1u
PERKAPERKA
ATF6GBATF6GB
BAXmBCL2BAXmBCL2
mBiPTmBiPT
BiPTBiPT
mCHOPmCHOP
BiATFBiATF
BH3BCL2BH3BCL2
BiRE1BiRE1
ATF6p50ATF6p50
Xbp1s[X-box-binding protein 1]
GADD34[Protein phosphatase 1 regulatory subunit 15A]
mGADD34mGADD34
BAXmTBAXmT
mXbp1smXbp1s

Eungdamrong2007_Ras_Activation: BIOMD0000000161v0.0.1

The model reproduces the time profiles of Golgi Ras-GTP and plasma membrane Ras-GTP, subjected to a palmitoylation rate…

Details

Imaging experiments have shown that cell signaling components such as Ras can be activated by growth factors at distinct subcellular locations. Trafficking between these subcellular locations is a regulated dynamic process. The effects of trafficking and the molecular mechanisms underlying compartment-specific Ras activation were studied using numerical simulations of an ordinary differential equation-based multi-compartment model. The simulations show that interplay between two distinct mechanisms, a palmitoylation cycle that controls Ras trafficking and a phospholipase C-epsilon (PLC-epsilon) driven feedback loop, can convert a transient calcium signal into prolonged Ras activation at the Golgi. Detailed analysis of the network identified PLC-epsilon as a key determinant of "compartment switching". Modulation of PLC-epsilon activity switches the location of activated Ras between the plasma membrane and Golgi through a new mechanism termed "kinetic scaffolding". These simulations indicate that multiple biochemical mechanisms, when appropriately coupled, can give rise to an intracellular compartment-specific sustained Ras activation in response to stimulation of growth factor receptors at the plasma membrane. link: http://identifiers.org/pubmed/17098795

Parameters:

NameDescription
KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; Kf=50.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); Kr=10.0 s^(-1)Reaction: buffer_cyt + Ca => ca_buffer_cyt, Rate Law: (Kf*0.00166112956810631*buffer_cyt*0.00166112956810631*Ca+(-Kr*0.00166112956810631*ca_buffer_cyt))*cyt*1*1/KMOLE
Kr_Ca_binds_IP3R = NaN s^(-1); I=0.0 dimensionless*A*m^(-2); Kf=1000.0 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: Ract + Ca => RactCa, Rate Law: (Ract*Kf*0.00166112956810631*Ca+(-Kr_Ca_binds_IP3R*RactCa))*erMembrane
I=0.0 dimensionless*A*m^(-2); Kr=0.0 s^(-1); Kf=0.2 s^(-1)Reaction: PLC_act_PM => PLC_PM, Rate Law: (Kf*PLC_act_PM+(-Kr*PLC_PM))*PM
Ratebasal_PIPsyn_PIP_synthesis = NaN s^(-1); Ratestim_PIPsyn_PIP_synthesis = NaN s^(-1); I=0.0 dimensionless*A*m^(-2)Reaction: PI_PM => PIP_PM, Rate Law: (Ratebasal_PIPsyn_PIP_synthesis+Ratestim_PIPsyn_PIP_synthesis)*PI_PM*PM
KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; Kr=0.0168 s^(-1); Kf=0.025 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: Sos_cyt + Grb2_cyt => SosGrb2_cyt, Rate Law: (Kf*0.00166112956810631*Sos_cyt*0.00166112956810631*Grb2_cyt+(-Kr*0.00166112956810631*SosGrb2_cyt))*cyt*1*1/KMOLE
Kr=0.0 1E12*dimensionless*item*m^(-2)*s^(-1); I=0.0 dimensionless*A*m^(-2); Kf=0.002 s^(-1)Reaction: Activated_EGFR_PM =>, Rate Law: Kf*Activated_EGFR_PM*PM
Kr=0.5 s^(-1); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; Kf=0.1 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: Ca + CAPRI_cyt => CaCAPRI_cyt, Rate Law: (Kf*0.00166112956810631*Ca*0.00166112956810631*CAPRI_cyt+(-Kr*0.00166112956810631*CaCAPRI_cyt))*cyt*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; vL=3.16E-5 1E-18*dimensionless*item^(-1)*m^3*s^(-1)Reaction: Ca => Ca_ER; ER_erMembrane, Rate Law: (-ER_erMembrane*(0.00166112956810631*Ca_ER+(-0.00166112956810631*Ca))*vL)*erMembrane*1*1/KMOLE
Kr=0.0 s^(-1); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; Kf=1.0E-4 s^(-1)Reaction: RasGTP_pal_cyt => RasGDP_pal_cyt, Rate Law: (Kf*0.00166112956810631*RasGTP_pal_cyt+(-Kr*0.00166112956810631*RasGDP_pal_cyt))*cyt*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Km=410.0 1E12*dimensionless*item*m^(-2); Vmax_EGF_act_PLCgamma = NaN 1E12*dimensionless*item*m^(-2)*s^(-1)Reaction: PLC_PM => PLC_act_PM; Activated_EGFR_PM, Rate Law: Vmax_EGF_act_PLCgamma*PLC_PM*1/(Km+PLC_PM)*PM
Kf=0.015 s^(-1); I=0.0 dimensionless*A*m^(-2); Kr=1.0E-5 1E15*dimensionless*item*m*mol^(-1)*s^(-1)Reaction: RasGDP_Golgi_GM => RasGDP_pal_cyt, Rate Law: (Kf*RasGDP_Golgi_GM+(-Kr*0.00166112956810631*RasGDP_pal_cyt))*GM
IP3_basal=0.0 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; kIP3deg=0.5 s^(-1)Reaction: IP3 =>, Rate Law: kIP3deg*(0.00166112956810631*IP3+(-IP3_basal))*cyt*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Kr=0.1 s^(-1); Kf=120.0 1E15*dimensionless*item*m*mol^(-1)*s^(-1)Reaction: CaCAPRI_cyt => CaCAPRI_PM_PM, Rate Law: (Kf*0.00166112956810631*CaCAPRI_cyt+(-Kr*CaCAPRI_PM_PM))*PM
I=0.0 dimensionless*A*m^(-2); Kf=1.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); Kr=0.01 s^(-1)Reaction: EGF_EC + EGFR_PM => Activated_EGFR_PM, Rate Law: (Kf*0.00166112956810631*EGF_EC*EGFR_PM+(-Kr*Activated_EGFR_PM))*PM
I=0.0 dimensionless*A*m^(-2); Kr=0.0 s^(-1); Kf=1.0E-4 s^(-1)Reaction: RasGTP_PM => RasGDP_PM, Rate Law: (Kf*RasGTP_PM+(-Kr*RasGDP_PM))*PM
Km=600.0 1E12*dimensionless*item*m^(-2); I=0.0 dimensionless*A*m^(-2); Vmax_RasGRP_DAG_GEF = NaN 1E12*dimensionless*item*m^(-2)*s^(-1)Reaction: RasGDP_Golgi_GM => RasGTP_Golgi_GM; RasGRP_DAG_GM, Rate Law: Vmax_RasGRP_DAG_GEF*RasGDP_Golgi_GM*1/(Km+RasGDP_Golgi_GM)*GM
I=0.0 dimensionless*A*m^(-2); Km=1200.0 1E12*dimensionless*item*m^(-2); Vmax_CaRasGRP_act_RasGM = NaN 1E12*dimensionless*item*m^(-2)*s^(-1)Reaction: RasGDP_Golgi_GM => RasGTP_Golgi_GM; Ca_RasGRP_GM_GM, Rate Law: Vmax_CaRasGRP_act_RasGM*RasGDP_Golgi_GM*1/(Km+RasGDP_Golgi_GM)*GM
I=0.0 dimensionless*A*m^(-2); Km=1032.0 1E12*dimensionless*item*m^(-2); Vmax_Shc_phosphorylation = NaN 1E12*dimensionless*item*m^(-2)*s^(-1)Reaction: Shc_PM => Shc_star_PM; Activated_EGFR_PM, Rate Law: Vmax_Shc_phosphorylation*Shc_PM*1/(Km+Shc_PM)*PM
Kf=0.25 s^(-1); Kr=0.0 1E12*dimensionless*item*m^(-2)*s^(-1); I=0.0 dimensionless*A*m^(-2)Reaction: DAG_GM_GM =>, Rate Law: Kf*DAG_GM_GM*GM
kact=1.18 1E-12*dimensionless*item^(-1)*m^2*s^(-1); I=0.0 dimensionless*A*m^(-2)Reaction: PIP2_GM_GM => DAG_GM_GM + IP3; Ras_CaPLCe_GM, Rate Law: kact*PIP2_GM_GM*Ras_CaPLCe_GM*GM
I=0.0 dimensionless*A*m^(-2); k_PIP2hyd=1.188 1E-12*dimensionless*item^(-1)*m^2*s^(-1)Reaction: PIP2_PM => DAG_PM + IP3; PLC_act_PM, Rate Law: k_PIP2hyd*PIP2_PM*PLC_act_PM*PM
Km=600.0 1E12*dimensionless*item*m^(-2); I=0.0 dimensionless*A*m^(-2); Vmax=1.0 1E12*dimensionless*item*m^(-2)*s^(-1)Reaction: RasGTP_Golgi_GM => RasGDP_Golgi_GM, Rate Law: Vmax*RasGTP_Golgi_GM*1/(Km+RasGTP_Golgi_GM)*GM
I=0.0 dimensionless*A*m^(-2); Kf=1.0E-4 s^(-1); Kr=0.0 1E15*dimensionless*item*m*mol^(-1)*s^(-1)Reaction: RasGTP_PM => RasGTP_depal_cyt, Rate Law: (Kf*RasGTP_PM+(-Kr*0.00166112956810631*RasGTP_depal_cyt))*PM
I=0.0 dimensionless*A*m^(-2); Kf=1.58489319246111E-4 s^(-1); Kr=1.0E-5 1E15*dimensionless*item*m*mol^(-1)*s^(-1)Reaction: RasGTP_Golgi_GM => RasGTP_pal_cyt, Rate Law: (Kf*RasGTP_Golgi_GM+(-Kr*0.00166112956810631*RasGTP_pal_cyt))*GM
Km=600.0 1E12*dimensionless*item*m^(-2); I=0.0 dimensionless*A*m^(-2); Vmax_Sos_act_RasPM = NaN 1E12*dimensionless*item*m^(-2)*s^(-1)Reaction: RasGDP_PM => RasGTP_PM; SGS_PM, Rate Law: Vmax_Sos_act_RasPM*RasGDP_PM*1/(Km+RasGDP_PM)*PM
I=0.0 dimensionless*A*m^(-2); Kf=120.0 1E15*dimensionless*item*m*mol^(-1)*s^(-1); Kr=0.01 s^(-1)Reaction: RasGDP_depal_cyt => RasGDP_Golgi_GM, Rate Law: (Kf*0.00166112956810631*RasGDP_depal_cyt+(-Kr*RasGDP_Golgi_GM))*GM
I=0.0 dimensionless*A*m^(-2); Km=1200.0 1E12*dimensionless*item*m^(-2); Vmax_CAPRI_GAP = NaN 1E12*dimensionless*item*m^(-2)*s^(-1)Reaction: RasGTP_PM => RasGDP_PM; CaCAPRI_PM_PM, Rate Law: Vmax_CAPRI_GAP*RasGTP_PM*1/(Km+RasGTP_PM)*PM
KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; Kf=3.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); Kr=1.0 s^(-1)Reaction: Ca + PLCe_cyt => Ca_PLCe_cyt, Rate Law: (Kf*0.00166112956810631*Ca*0.00166112956810631*PLCe_cyt+(-Kr*0.00166112956810631*Ca_PLCe_cyt))*cyt*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; singleChanFlux=4.69 1E-30*dimensionless*item^(-2)*m^5*s^(-1); dI=0.8 0.001*dimensionless*m^(-3)*molReaction: Ca => Ca_ER; ER_erMembrane, RactCa, Ract, IP3, Rinh, RinhCa, Rate Law: (-0.25*ER_erMembrane*(RactCa+Ract)*(0.00166112956810631*Ca_ER+(-0.00166112956810631*Ca))*(0.00166112956810631*IP3*RactCa*Rinh*1/(0.00166112956810631*IP3+dI)*1/(RactCa+Ract)*1/(RinhCa+Rinh))^3*singleChanFlux)*erMembrane*1*1/KMOLE
Kon_reaction2 = 2.1 1000*dimensionless*m^3*mol^(-1)*s^(-1); I=0.0 dimensionless*A*m^(-2); Koff_reaction2 = NaN s^(-1)Reaction: Rinh + Ca => RinhCa, Rate Law: (Kon_reaction2*Rinh*0.00166112956810631*Ca+(-Koff_reaction2*RinhCa))*erMembrane
I=0.0 dimensionless*A*m^(-2); Kr=0.1 s^(-1); Kf=90.0 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: SosGrb2_cyt + Shc_star_PM => SGS_PM, Rate Law: (Kf*0.00166112956810631*SosGrb2_cyt*Shc_star_PM+(-Kr*SGS_PM))*PM
I=0.0 dimensionless*A*m^(-2); Kr=0.1 s^(-1); Kf=0.5 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: DAG_GM_GM + RasGRP_cyt => RasGRP_DAG_GM, Rate Law: (Kf*DAG_GM_GM*0.00166112956810631*RasGRP_cyt+(-Kr*RasGRP_DAG_GM))*GM
I=0.0 dimensionless*A*m^(-2); Kr=0.0 s^(-1); Kf=0.5 s^(-1)Reaction: Shc_star_PM => Shc_PM, Rate Law: (Kf*Shc_star_PM+(-Kr*Shc_PM))*PM
vP=0.0664 1E-33*dimensionless*item^(-2)*m^2*mol*s^(-1); I=0.0 dimensionless*A*m^(-2); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; kP=0.27Reaction: Ca => Ca_ER; ER_erMembrane, serca, Rate Law: ER_erMembrane*serca*vP*0.00166112956810631*Ca*0.00166112956810631*Ca*1/(kP*kP+0.00166112956810631*Ca*0.00166112956810631*Ca)*erMembrane*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Kf=10.0 1E15*dimensionless*item*m*mol^(-1)*s^(-1); Kr=5.0 s^(-1)Reaction: CaRasGRP1_cyt => Ca_RasGRP_GM_GM, Rate Law: (Kf*0.00166112956810631*CaRasGRP1_cyt+(-Kr*Ca_RasGRP_GM_GM))*GM
I=0.0 dimensionless*A*m^(-2); Rate_PIP2Synbasal_PIP2_synthesis = NaN s^(-1); Rate_PIP2SynStim_PIP2_synthesis = NaN s^(-1)Reaction: PIP_PM => PIP2_PM, Rate Law: (Rate_PIP2Synbasal_PIP2_synthesis+Rate_PIP2SynStim_PIP2_synthesis)*PIP_PM*PM
I=0.0 dimensionless*A*m^(-2); Kf=15.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); Kr=1.0 s^(-1)Reaction: RasGTP_Golgi_GM + Ca_PLCe_cyt => Ras_CaPLCe_GM, Rate Law: (Kf*RasGTP_Golgi_GM*0.00166112956810631*Ca_PLCe_cyt+(-Kr*Ras_CaPLCe_GM))*GM

States:

NameDescription
Ras CaPLCe GM[calcium(2+); Ras-related protein R-Ras2; Calcium cation]
DAG PM[diglyceride; Diacylglycerol]
RinhRinh
EGF ECEGF_EC
Ca ER[calcium(2+); Calcium cation]
RasGTP Golgi GM[Ras-related protein R-Ras2]
EGFR PM[Epidermal growth factor receptor]
PI PM[1-phosphatidyl-1D-myo-inositol; 1-Phosphatidyl-D-myo-inositol]
SosGrb2 cyt[Growth factor receptor-bound protein 2; Son of sevenless homolog 1]
ca buffer cytca_buffer_cyt
RasGTP pal cyt[Ras-related protein R-Ras2]
RasGDP depal cyt[Ras-related protein R-Ras2]
RasGRP cyt[RAS guanyl-releasing protein 1]
RasGDP Golgi GM[Ras-related protein R-Ras2]
RinhCa[calcium(2+); Calcium cation]
Activated EGFR PM[Epidermal growth factor receptor]
Ca[calcium(2+); Calcium cation]
RasGDP pal cyt[Ras-related protein R-Ras2]
CaRasGRP1 cyt[calcium(2+); RAS guanyl-releasing protein 1; Calcium cation]
RasGTP PM[Ras-related protein R-Ras2]
PLCe cytPLCe_cyt
RactCaRactCa
PLC act PMPLC_act_PM
Shc star PM[SHC-transforming protein 2]
PLC PMPLC_PM
buffer cytbuffer_cyt
RasGDP PM[Ras-related protein R-Ras2]
IP3[1D-myo-inositol 1,4,5-trisphosphate; D-myo-Inositol 1,4,5-trisphosphate]
CAPRI cytCAPRI_cyt
Sos cyt[Son of sevenless homolog 1]
CaCAPRI cyt[calcium(2+); Calcium cation]
Shc PM[SHC-transforming protein 2]
SGS PMSGS_PM
PIP2 GM GM[1-phosphatidyl-1D-myo-inositol 4,5-bisphosphate; 1-Phosphatidyl-D-myo-inositol 4,5-bisphosphate]
Grb2 cyt[Growth factor receptor-bound protein 2]
PIP2 PM[1-phosphatidyl-1D-myo-inositol 4,5-bisphosphate; 1-Phosphatidyl-D-myo-inositol 4,5-bisphosphate]
CaCAPRI PM PM[calcium(2+); Calcium cation]
RactRact
PIP PM[1-phosphatidyl-1D-myo-inositol 4-phosphate; 1-Phosphatidyl-1D-myo-inositol 4-phosphate]
Ca RasGRP GM GM[calcium(2+); RAS guanyl-releasing protein 1; Calcium cation]
DAG GM GM[diglyceride; Diacylglycerol]
RasGTP depal cyt[Ras-related protein R-Ras2]
RasGRP DAG GM[RAS guanyl-releasing protein 1]
Ca PLCe cyt[calcium(2+); Calcium cation]

Evans2004 - Cell based mathematical model of topotecan: BIOMD0000000945v0.0.1

A two compartment mathematical model of the antineoplastic compound topotecan

Details

In this paper a compartmental modelling approach is applied to provide a mathematical description of the activity of the anti-cancer agent topotecan, and delivery to its nuclear DNA target following administration. The activity of topotecan in defined buffers is first modelled using a linear two compartment model that then forms the basis of a cell based model for drug activity in live cell experiments. An identifiability analysis is performed before parameter estimation to ensure that the model output (i.e., continuous, perfect and noise-free data) uniquely determines the parameters. Parameter estimation is performed using experimental data which offers concentrations of active and inactive forms of topotecan from high performance liquid chromatography methods. link: http://identifiers.org/pubmed/15094319

Parameters:

NameDescription
k_c_c = 0.18637; k_i = 3.09E-4; k_b = 8.5341E-4; k_d = 4.4489; k_e = 1.014; v_2 = 0.393244873341375; v_1 = 6.0313630880579; B_T = 28.9; k_o_c = 0.026553Reaction: L_c = (((k_i*v_1*L_m-(k_e+k_o_c)*L_c)+k_c_c*H_c)-k_b*(B_T-L_n)*L_c)+v_2*k_d*L_n, Rate Law: (((k_i*v_1*L_m-(k_e+k_o_c)*L_c)+k_c_c*H_c)-k_b*(B_T-L_n)*L_c)+v_2*k_d*L_n
k_0_m = 0.0289; k_c_m = 1.06E-4Reaction: H_m = k_0_m*L_m-k_c_m*H_m, Rate Law: k_0_m*L_m-k_c_m*H_m
k_c_c = 0.18637; k_o_c = 0.026553Reaction: H_c = k_o_c*L_c-k_c_c*H_c, Rate Law: k_o_c*L_c-k_c_c*H_c
k_i = 3.09E-4; k_0_m = 0.0289; k_c_m = 1.06E-4; k_e = 1.014; v_1 = 6.0313630880579Reaction: L_m = (-(k_0_m+k_i))*L_m+k_c_m*H_m+k_e*v_1*L_c, Rate Law: (-(k_0_m+k_i))*L_m+k_c_m*H_m+k_e*v_1*L_c
k_b = 8.5341E-4; k_d = 4.4489; v_2 = 0.393244873341375; B_T = 28.9Reaction: L_n = k_b*v_2*(B_T-L_n)*L_c-k_d*L_n, Rate Law: k_b*v_2*(B_T-L_n)*L_c-k_d*L_n

States:

NameDescription
H m[hydroxy monocarboxylic acid; C11158; extracellular region; inactive]
L c[lactone; C11158; intracellular; antineoplastic agent]
H c[hydroxy monocarboxylic acid; C11158; intracellular; inactive]
L m[lactone; C11158; extracellular region; antineoplastic agent]
L n[deoxyribonucleic acid; lactone; C11158; nucleus; antineoplastic agent; protein-DNA complex assembly]

Evans2005 - Compartmental model for antineoplastic drug topotecan in breast cancer cells: BIOMD0000000946v0.0.1

Compartment model for the antineoplastic drug topotecan. Modelling drug in its active lactone and inactive hydroxy acid…

Details

A compartmental model for the in vitro uptake kinetics of the anti-cancer agent topotecan is proposed. This model provides a description of the activity of the drug, and subsequent delivery of active form to the nuclear DNA target. The unknown model parameters are estimated from two-photon laser-scanning microscopy data, which provide concentrations of topotecan (active plus inactive forms) in the extracellular region containing live human breast tumour cells (MCF-7 cell line), the cytoplasm and the nucleus. This determines an output structure for which the model is uniquely identifiable, that is, the unknown parameters are uniquely determined from noise-free, continuous and perfect data. The model allows in silico predictions of the dose dependence of target binding. Copyright 2005 John Wiley & Sons, Ltd. link: http://identifiers.org/doi/10.1002/acs.856

Parameters:

NameDescription
v2 = 0.393244873341375; k_oc = 1.2913E-4; k_dh = 1.5639E-7; k_cc = 3.1578E-4Reaction: H_c = (k_oc*L_c-k_cc*H_c)+k_dh*v2*L_n, Rate Law: (k_oc*L_c-k_cc*H_c)+k_dh*v2*L_n
v1 = 0.362955164339553; k_i = 0.02211; k_dl = 0.055522; v2 = 0.393244873341375; k_e = 0.0078915; k_oc = 1.2913E-4; B_T = 89.972; k_cc = 3.1578E-4; k_b = 3.8085E-4Reaction: L_c = ((k_i*v1*L_e-(k_e+k_oc)*L_c)+k_cc*H_c+k_dl*v2*L_n)-k_b*(B_T-L_n)*L_c, Rate Law: ((k_i*v1*L_e-(k_e+k_oc)*L_c)+k_cc*H_c+k_dl*v2*L_n)-k_b*(B_T-L_n)*L_c
v1 = 0.362955164339553; k_i = 0.02211; k_mi = 1.3974E-6; k_mo = 0.085551; k_e = 0.0078915; k_om = 1.5599E-4; v0 = 1.5045E-5; k_cm = 2.9553E-4Reaction: L_e = (k_mi/v0*L_m-(k_mo+k_om+k_i)*L_e)+k_cm*H_e+k_e/v1*L_c, Rate Law: (k_mi/v0*L_m-(k_mo+k_om+k_i)*L_e)+k_cm*H_e+k_e/v1*L_c
k_mi = 1.3974E-6; k_mo = 0.085551; k_om = 1.5599E-4; v0 = 1.5045E-5; k_cm = 2.9553E-4Reaction: H_e = (k_mi/v0*H_m+k_om*L_e)-(k_cm+k_mo)*H_e, Rate Law: (k_mi/v0*H_m+k_om*L_e)-(k_cm+k_mo)*H_e
k_dl = 0.055522; v2 = 0.393244873341375; B_T = 89.972; k_dh = 1.5639E-7; k_b = 3.8085E-4Reaction: L_n = k_b/v2*(B_T-L_n)*L_c-(k_dl+k_dh)*L_n, Rate Law: k_b/v2*(B_T-L_n)*L_c-(k_dl+k_dh)*L_n
k_mi = 1.3974E-6; k_mo = 0.085551; k_om = 1.5599E-4; k_cm = 2.9553E-4; v0 = 1.5045E-5Reaction: L_m = (-(k_om+k_mi))*L_m+k_cm*H_m+k_mo*v0*L_e, Rate Law: (-(k_om+k_mi))*L_m+k_cm*H_m+k_mo*v0*L_e

States:

NameDescription
H m[hydroxy monocarboxylic acid; C11158; culture medium; inactive]
L c[lactone; C11158; cytoplasm; antineoplastic agent]
H c[hydroxy monocarboxylic acid; C11158; cytoplasm; inactive]
L e[lactone; C11158; extracellular region; antineoplastic agent]
H e[hydroxy monocarboxylic acid; C11158; extracellular region; inactive]
L m[lactone; C11158; culture medium; antineoplastic agent]
L n[lactone; deoxyribonucleic acid; C11158; protein-DNA complex assembly; nucleus; antineoplastic agent]

F


Faber2000_LRmodel_CardiacMyocytes: MODEL0912153452v0.0.1

This a model from the article: Action potential and contractility changes in [Na(+)](i) overloaded cardiac myocytes: a…

Details

Sodium overload of cardiac cells can accompany various pathologies and induce fatal cardiac arrhythmias. We investigate effects of elevated intracellular sodium on the cardiac action potential (AP) and on intracellular calcium using the Luo-Rudy model of a mammalian ventricular myocyte. The results are: 1) During rapid pacing, AP duration (APD) shortens in two phases, a rapid phase without Na(+) accumulation and a slower phase that depends on Na(+). 2) The rapid APD shortening is due to incomplete deactivation (accumulation) of I(Ks). 3) The slow phase is due to increased repolarizing currents I(NaK) and reverse-mode I(NaCa), secondary to elevated Na(+). 4) Na(+)-overload slows the rate of AP depolarization, allowing time for greater I(Ca(L)) activation; it also enhances reverse-mode I(NaCa). The resulting increased Ca(2+) influx triggers a greater Ca(2+) transient. 5) Reverse-mode I(NaCa) alone can trigger Ca(2+) release in a voltage and Na(+)-dependent manner. 6) During I(NaK) block, Na(+) and Ca(2+) accumulate and APD shortens due to enhanced reverse-mode I(NaCa); contribution of I(K(Na)) to APD shortening is negligible. By slowing AP depolarization (hence velocity) and shortening APD, Na(+)-overload acts to enhance inducibility of reentrant arrhythmias. Shortened APD with elevated Ca(2+) (secondary to Na(+)-overload) also predisposes the myocardium to arrhythmogenic delayed afterdepolarizations. link: http://identifiers.org/pubmed/10777735

Fallon2000 - Interleukin-2 dynamics: BIOMD0000000665v0.0.1

This a model from the article: Computational model for effects of ligand/receptor binding properties on interleukin-2…

Details

Multisubunit cytokine receptors such as the heterotrimeric receptor for interleukin-2 (IL-2) are ubiquitous in hematopoeitic cell types of importance in biotechnology and are crucial regulators of cell proliferation and differentiation behavior. Dynamics of cytokine/receptor endocytic trafficking can significantly impact cell responses through effects of receptor down-regulation and ligand depletion, and in turn are governed by ligand/receptor binding properties. We describe here a computational model for trafficking dynamics of the IL-2 receptor (IL-2R) system, which is able to predict T cell proliferation responses to IL-2. This model comprises kinetic equations describing binding, internalization, and postendocytic sorting of IL-2 and IL-2R, including an experimentally derived dependence of cell proliferation rate on these properties. Computational results from this model predict that IL-2 depletion can be reduced by decreasing its binding affinity for the IL-2R betagamma subunit relative to the alpha subunit at endosomal pH, as a result of enhanced ligand sorting to recycling vis-à-vis degradation, and that an IL-2 analogue with such altered binding properties should exhibit increased potency for stimulating the T cell proliferation response. These results are in agreement with our recent experimental findings for the IL-2 analogue termed 2D1 [Fallon, E. M. et al. J. Biol. Chem. 2000, 275, 6790-6797]. Thus, this type of model may enable prediction of beneficial cytokine/receptor binding properties to aid development of molecular design criteria for improvements in applications such as in vivo cytokine therapies and in vitro hematopoietic cell bioreactors. link: http://identifiers.org/pubmed/11027188

Parameters:

NameDescription
ksyn = 0.0011 1/(0.0166667*s)Reaction: => Rs_0; Cs_0, Rate Law: COMpartment*ksyn*Cs_0
kt = 0.007 1/(0.0166667*s)Reaction: Rs_0 => Ri_0, Rate Law: COMpartment*kt*Rs_0
kf = 0.00124324324324324 0.06*nl/(mol*s); NA = 6.022E11 1/Tmol; Ve = 1.0E-14 l; kr = 0.0138 1/(0.0166667*s); kx = 0.15 1/(0.0166667*s)Reaction: L_0 => ; Rs_0, Cs_0, Li_0, Y_0, Rate Law: COMpartment*((kf*L_0*Rs_0-kr*Cs_0)-kx*Li_0*Ve*NA)*Y_0/NA
ke = 0.04 1/(0.0166667*s)Reaction: Cs_0 => Ci_0, Rate Law: COMpartment*ke*Cs_0
kh = 0.035 1/(0.0166667*s)Reaction: Ri_0 =>, Rate Law: COMpartment*kh*Ri_0
kre = 0.1104 1/(0.0166667*s)Reaction: Ci_0 => Ri_0, Rate Law: COMpartment*kre*Ci_0
kf = 0.00124324324324324 0.06*nl/(mol*s); NA = 6.022E11 1/Tmol; kre = 0.1104 1/(0.0166667*s); Ve = 1.0E-14 lReaction: Li_0 => ; Ri_0, Ci_0, Rate Law: COMpartment*(kf*Li_0*Ri_0-kre*Ci_0)/(Ve*NA)
kf = 0.00124324324324324 0.06*nl/(mol*s)Reaction: Rs_0 => Cs_0; L_0, Rate Law: COMpartment*kf*L_0*Rs_0
kr = 0.0138 1/(0.0166667*s)Reaction: Cs_0 => Rs_0, Rate Law: COMpartment*kr*Cs_0
kfe = 1.104E-4 0.06*nl/(mol*s)Reaction: Ri_0 => Ci_0; Li_0, Rate Law: COMpartment*kfe*Li_0*Ri_0
kx = 0.15 1/(0.0166667*s)Reaction: Li_0 =>, Rate Law: COMpartment*kx*Li_0
Vs = 11.0 1/(0.0166667*s)Reaction: => Rs_0, Rate Law: COMpartment*Vs

States:

NameDescription
Ld 0[Interleukin-2]
Y 0[T-lymphocyte]
Ci 0[Cytokine receptor common subunit gamma; Interleukin-2; Interleukin-2 receptor subunit beta; interleukin-2 receptor complex; intracellular part]
L 0[Interleukin-2; extracellular space]
Rs 0[Interleukin-2 receptor subunit beta; Cytokine receptor common subunit gamma; Interleukin-2 Receptor; plasma membrane]
Ri 0[Cytokine receptor common subunit gamma; Interleukin-2 receptor subunit beta; Interleukin-2 Receptor; intracellular part]
Li 0[Interleukin-2; intracellular part]
Cs 0[Cytokine receptor common subunit gamma; Interleukin-2; Interleukin-2 receptor subunit beta; interleukin-2 receptor complex; plasma membrane]

Fang2010 - Genome-scale metabolic network of Mycobacterium tuberculosis (iNJ661m): MODEL1507180018v0.0.1

Fang2010 - Genome-scale metabolic network of Mycobacterium tuberculosis (iNJ661m)This model is described in the article:…

Details

BACKGROUND: During infection, Mycobacterium tuberculosis confronts a generally hostile and nutrient-poor in vivo host environment. Existing models and analyses of M. tuberculosis metabolic networks are able to reproduce experimentally measured cellular growth rates and identify genes required for growth in a range of different in vitro media. However, these models, under in vitro conditions, do not provide an adequate description of the metabolic processes required by the pathogen to infect and persist in a host. RESULTS: To better account for the metabolic activity of M. tuberculosis in the host environment, we developed a set of procedures to systematically modify an existing in vitro metabolic network by enhancing the agreement between calculated and in vivo-measured gene essentiality data. After our modifications, the new in vivo network contained 663 genes, 838 metabolites, and 1,049 reactions and had a significantly increased sensitivity (0.81) in predicted gene essentiality than the in vitro network (0.31). We verified the modifications generated from the purely computational analysis through a review of the literature and found, for example, that, as the analysis suggested, lipids are used as the main source for carbon metabolism and oxygen must be available for the pathogen under in vivo conditions. Moreover, we used the developed in vivo network to predict the effects of double-gene deletions on M. tuberculosis growth in the host environment, explore metabolic adaptations to life in an acidic environment, highlight the importance of different enzymes in the tricarboxylic acid-cycle under different limiting nutrient conditions, investigate the effects of inhibiting multiple reactions, and look at the importance of both aerobic and anaerobic cellular respiration during infection. CONCLUSIONS: The network modifications we implemented suggest a distinctive set of metabolic conditions and requirements faced by M. tuberculosis during host infection compared with in vitro growth. Likewise, the double-gene deletion calculations highlight the importance of specific metabolic pathways used by the pathogen in the host environment. The newly constructed network provides a quantitative model to study the metabolism and associated drug targets of M. tuberculosis under in vivo conditions. link: http://identifiers.org/pubmed/21092312

Fang2011 - Genome-scale metabolic network of Burkholderia cenocepacia (iKF1028): MODEL1507180051v0.0.1

Fang2011 - Genome-scale metabolic network of Burkholderia cenocepacia (iKF1028)This model is described in the article:…

Details

BACKGROUND: Burkholderia cenocepacia is a threatening nosocomial epidemic pathogen in patients with cystic fibrosis (CF) or a compromised immune system. Its high level of antibiotic resistance is an increasing concern in treatments against its infection. Strain B. cenocepacia J2315 is the most infectious isolate from CF patients. There is a strong demand to reconstruct a genome-scale metabolic network of B. cenocepacia J2315 to systematically analyze its metabolic capabilities and its virulence traits, and to search for potential clinical therapy targets. RESULTS: We reconstructed the genome-scale metabolic network of B. cenocepacia J2315. An iterative reconstruction process led to the establishment of a robust model, iKF1028, which accounts for 1,028 genes, 859 internal reactions, and 834 metabolites. The model iKF1028 captures important metabolic capabilities of B. cenocepacia J2315 with a particular focus on the biosyntheses of key metabolic virulence factors to assist in understanding the mechanism of disease infection and identifying potential drug targets. The model was tested through BIOLOG assays. Based on the model, the genome annotation of B. cenocepacia J2315 was refined and 24 genes were properly re-annotated. Gene and enzyme essentiality were analyzed to provide further insights into the genome function and architecture. A total of 45 essential enzymes were identified as potential therapeutic targets. CONCLUSIONS: As the first genome-scale metabolic network of B. cenocepacia J2315, iKF1028 allows a systematic study of the metabolic properties of B. cenocepacia and its key metabolic virulence factors affecting the CF community. The model can be used as a discovery tool to design novel drugs against diseases caused by this notorious pathogen. link: http://identifiers.org/pubmed/21609491

Fang2020 - SEIR model of COVID-19 transmission considering government interventions in Wuhan: BIOMD0000000984v0.0.1

Using the parameterized susceptible‐exposed‐infectious‐recovered model, we simulated the spread dynamics of coronavirus…

Details

Using the parameterized susceptible-exposed-infectious-recovered model, we simulated the spread dynamics of coronavirus disease 2019 (COVID-19) outbreak and impact of different control measures, conducted the sensitivity analysis to identify the key factor, plotted the trend curve of effective reproductive number (R), and performed data fitting after the simulation. By simulation and data fitting, the model showed the peak existing confirmed cases of 59 769 arriving on 15 February 2020, with the coefficient of determination close to 1 and the fitting bias 3.02%, suggesting high precision of the data-fitting results. More rigorous government control policies were associated with a slower increase in the infected population. Isolation and protective procedures would be less effective as more cases accrue, so the optimization of the treatment plan and the development of specific drugs would be of more importance. There was an upward trend of R in the beginning, followed by a downward trend, a temporary rebound, and another continuous decline. The feature of high infectiousness for severe acute respiratory syndrome coronavirus 2(SARS-CoV-2) led to an upward trend, and government measures contributed to the temporary rebound and declines. The declines of R could be exploited as strong evidence for the effectiveness of the interventions. Evidence from the four-phase stringent measures showed that it was significant to ensure early detection, early isolation, early treatment, adequate medical supplies, patients&#39; being admitted to designated hospitals, and comprehensive therapeutic strategy. Collaborative efforts are required to combat the novel coronavirus, focusing on both persistent strict domestic interventions and vigilance against exogenous imported cases. link: http://identifiers.org/pubmed/32141624

FangeElf2006_MinSystem_MesoRD: MODEL5974712823v0.0.1

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

Details

The spatiotemporal oscillations of the Escherichia coli proteins MinD and MinE direct cell division to the region between the chromosomes. Several quantitative models of the Min system have been suggested before, but no one of them accounts for the behavior of all documented mutant phenotypes. We analyzed the stochastic reaction-diffusion kinetics of the Min proteins for several E. coli mutants and compared the results to the corresponding deterministic mean-field description. We found that wild-type (wt) and filamentous (ftsZ-) cells are well characterized by the mean-field model, but that a stochastic model is necessary to account for several of the characteristics of the spherical (rodA-) and phospathedylethanolamide-deficient (PE-) phenotypes. For spherical cells, the mean-field model is bistable, and the system can get trapped in a non-oscillatory state. However, when the intrinsic noise is considered, only the experimentally observed oscillatory behavior remains. The stochastic model also reproduces the change in oscillation directions observed in the spherical phenotype and the occasional gliding of the MinD region along the inner membrane. For the PE- mutant, the stochastic model explains the appearance of randomly localized and dense MinD clusters as a nucleation phenomenon, in which the stochastic kinetics at low copy number causes local discharges of the high MinD(ATP) to MinD(ADP) potential. We find that a simple five-reaction model of the Min system can explain all documented Min phenotypes, if stochastic kinetics and three-dimensional diffusion are accounted for. Our results emphasize that local copy number fluctuation may result in phenotypic differences although the total number of molecules of the relevant species is high. link: http://identifiers.org/pubmed/16846247

Faratian2009 - Role of PTEN in Trastuzumab resistance: BIOMD0000000424v0.0.1

Faratian2009 - Role of PTEN in Trastuzumab resistanceThis model is described in the article: [Systems biology reveals n…

Details

Resistance to targeted cancer therapies such as trastuzumab is a frequent clinical problem not solely because of insufficient expression of HER2 receptor but also because of the overriding activation states of cell signaling pathways. Systems biology approaches lend themselves to rapid in silico testing of factors, which may confer resistance to targeted therapies. Inthis study, we aimed to develop a new kinetic model that could be interrogated to predict resistance to receptor tyrosine kinase (RTK) inhibitor therapies and directly test predictions in vitro and in clinical samples. The new mathematical model included RTK inhibitor antibody binding, HER2/HER3 dimerization and inhibition, AKT/mitogen-activated protein kinase cross-talk, and the regulatory properties of PTEN. The model was parameterized using quantitative phosphoprotein expression data from cancer cell lines using reverse-phase protein microarrays. Quantitative PTEN protein expression was found to be the key determinant of resistance to anti-HER2 therapy in silico, which was predictive of unseen experiments in vitro using the PTEN inhibitor bp(V). When measured in cancer cell lines, PTEN expression predicts sensitivity to anti-HER2 therapy; furthermore, this quantitative measurement is more predictive of response (relative risk, 3.0; 95% confidence interval, 1.6-5.5; P < 0.0001) than other pathway components taken in isolation and when tested by multivariate analysis in a cohort of 122 breast cancers treated with trastuzumab. For the first time, a systems biology approach has successfully been used to stratify patients for personalized therapy in cancer and is further compelling evidence that PTEN, appropriately measured in the clinical setting, refines clinical decision making in patients treated with anti-HER2 therapies. link: http://identifiers.org/pubmed/19638581

Parameters:

NameDescription
k41 = 3.0; Kd_41 = 0.1Reaction: Akt_PI_PP + PP2A => Akt_PI_PP_PP2A, Rate Law: k41*(Akt_PI_PP*PP2A-Kd_41*Akt_PI_PP_PP2A)
k28 = 300.0; k_28 = 0.0Reaction: E23HP_PI3K => E23HP_PI3Ka, Rate Law: k28*(E23HP_PI3K-k_28*E23HP_PI3Ka)
k3 = 1.0; Kd_3 = 0.1Reaction: E23H_C => E23HP, Rate Law: k3*(E23H_C-Kd_3*E23HP)
k_50 = 0.012; k50 = 0.6Reaction: E2_Per => E2Per, Rate Law: k50*E2_Per-k_50*E2Per
V12 = 3.0; K12 = 0.1Reaction: RasGTP => RasGDP, Rate Law: V12*RasGTP/(K12+RasGTP)
k34 = 3.6Reaction: PTEN_PI => PI2 + PTEN, Rate Law: k34*PTEN_PI
k15 = 2.1; K15 = 1.0Reaction: MEK => MEKP; Rafa, Rate Law: k15*MEK*Rafa/(K15+MEK)
K24 = 10.0; V24 = 1.8Reaction: ERKP => ERK, Rate Law: V24*ERKP/(K24+ERKP)
k27 = 3.0; Kd_27 = 1.0Reaction: E23HP + PI3K => E23HP_PI3K, Rate Law: k27*(E23HP*PI3K-Kd_27*E23HP_PI3K)
k13 = 1.0; K13 = 11.7Reaction: Raf => Rafa; RasGTP, Rate Law: k13*Raf*RasGTP/(K13+Raf)
k23 = 1.2; K23 = 10.0Reaction: ERK => ERKP; MEKPP, Rate Law: k23*ERK*MEKPP/(K23+ERK)
Kd_5 = 1.0; k5 = 0.06Reaction: E23HP + Shc => E23HP_Shc, Rate Law: k5*(E23HP*Shc-Kd_5*E23HP_Shc)
k36 = 1.0; Kd_36 = 2.2Reaction: PTEN + PTENP => PTENP_PTEN, Rate Law: k36*(PTEN*PTENP-Kd_36*PTENP_PTEN)
k38 = 150.0Reaction: PTEN_PTEN => PTEN, Rate Law: k38*PTEN_PTEN
k_9 = 0.0; k9 = 35.0Reaction: ShGS => GS + ShcP, Rate Law: k9*(ShGS-k_9*ShcP*GS)
k16 = 0.06; Kd_16 = 1.0Reaction: MEKPP + PP2A => MEKPP_PP2A, Rate Law: k16*(PP2A*MEKPP-Kd_16*MEKPP_PP2A)
K11 = 0.18; k11 = 6.0Reaction: RasGDP => RasGTP; ShGS, Rate Law: k11*RasGDP*ShGS/(K11+RasGDP)
E_raf = 7.0; K14 = 50.0; k14 = 0.6Reaction: Rafa => Raf; Akt_PI_PP, Rate Law: k14*Rafa*(Akt_PI_PP+E_raf)/(Rafa+K14)
Kd_49 = 20000.0; k49 = 0.003Reaction: E2 + Per => E2_Per, Rate Law: k49*(Per*E2-Kd_49*E2_Per)
k16 = 0.06Reaction: MEKP + PP2A => MEKP_PP2A, Rate Law: k16*MEKP*PP2A
k22 = 0.06Reaction: MEKP_PP2A => MEKP + PP2A, Rate Law: k22*MEKP_PP2A
Kd_39 = 20.0; k39 = 15000.0Reaction: Akt + PIP3 => Akt_PIP3, Rate Law: k39*(PIP3*Akt-Kd_39*Akt_PIP3)
k48 = 0.001Reaction: E23HP =>, Rate Law: k48*E23HP
k56 = 30.0Reaction: PI3Ka_PIP3 => PI3Ka + PIP3, Rate Law: k56*PI3Ka_PIP3
k_6 = 3.0; k6 = 12.0Reaction: E23HP_Shc => E23HP_ShcP, Rate Law: k6*E23HP_Shc-k_6*E23HP_ShcP
bpV = 0.0; k57 = 100.0; Kd_57 = 10.0Reaction: PTEN => PTEN_bpV, Rate Law: k57*(PTEN*bpV-Kd_57*PTEN_bpV)
Kd_7 = 9.0; k7 = 36.0Reaction: E23HP_ShcP + GS => E23HP_ShGS, Rate Law: k7*(E23HP_ShcP*GS-Kd_7*E23HP_ShGS)
K4 = 50.0; V4 = 10.0Reaction: E23HP => E23H, Rate Law: V4*E23HP/(K4+E23HP)
k55 = 30.0Reaction: PI3Ka_PI => PI3Ka_PIP3, Rate Law: k55*PI3Ka_PI
K_d31 = 100.0; k31 = 0.03Reaction: PI2 + PI3Ka => PI3Ka_PI, Rate Law: k31*(PI2*PI3Ka-K_d31*PI3Ka_PI)
k43 = 30.0Reaction: Akt_PIP3_PP2A => Akt_PIP3 + PP2A, Rate Law: k43*Akt_PIP3_PP2A
k47 = 0.3Reaction: Akt_PI_P_PP2A => Akt_PI_P + PP2A, Rate Law: k47*Akt_PI_P_PP2A
K35 = 2.0; V35 = 150.0Reaction: PTEN => PTENP, Rate Law: V35*PTEN/(K35+PTEN)
V30 = 900.0Reaction: PI3Ka => PI3K, Rate Law: V30*PI3Ka
k_29 = 0.0; k29 = 13520.0Reaction: E23HP_PI3Ka => E23HP + PI3Ka, Rate Law: k29*E23HP_PI3Ka-k_29*E23HP*PI3Ka
k53 = 0.01Reaction: E23H => E23H_C, Rate Law: k53*E23H
k37 = 150.0Reaction: PTENP_PTEN => PTEN_PTEN, Rate Law: k37*PTENP_PTEN
k42 = 45.0Reaction: Akt_PI_P_PP2A => Akt_PIP3_PP2A, Rate Law: k42*Akt_PI_P_PP2A
k33 = 15.0Reaction: PTEN_PIP3 => PTEN_PI, Rate Law: k33*PTEN_PIP3
Kd_8 = 0.1; k8 = 12.0Reaction: E23HP_ShGS => E23HP + ShGS, Rate Law: k8*(E23HP_ShGS-Kd_8*E23HP*ShGS)
k41 = 3.0Reaction: Akt_PI_P + PP2A => Akt_PI_P_PP2A, Rate Law: k41*Akt_PI_P*PP2A
V10 = 0.0154; K10 = 340.0Reaction: ShcP => Shc, Rate Law: V10*ShcP/(K10+ShcP)
V40 = 15000.0; K40 = 0.1Reaction: Akt_PI_P => Akt_PI_PP, Rate Law: V40*Akt_PI_P/(K40+Akt_PI_P)
k18 = 0.6Reaction: MEK_PP2A => MEK + PP2A, Rate Law: k18*MEK_PP2A
Kd_2 = 10.0; k2 = 10.0Reaction: E2 + E3H_C => E23H, Rate Law: k2*(E3H_C*E2-Kd_2*E23H)
k16_kat = 0.6Reaction: MEKPP_PP2A => MEKP_PP2A, Rate Law: k16_kat*MEKPP_PP2A
Kd_32 = 0.01; k32 = 8000.0Reaction: PIP3 + PTEN => PTEN_PIP3, Rate Law: k32*(PIP3*PTEN-Kd_32*PTEN_PIP3)

States:

NameDescription
Rafa[RAF proto-oncogene serine/threonine-protein kinase]
PTEN[Histidine kinase 4; Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN; Phosphatidylinositol-3,4,5-trisphosphate]
Shc[SHC-transforming protein 1]
E23HP Shc[Phosphoprotein; Protein sevenless; Receptor tyrosine-protein kinase erbB-3; Pro-neuregulin-1, membrane-bound isoform; Receptor tyrosine-protein kinase erbB-2; SHC-transforming protein 1]
E23HP PI3K[Phosphoprotein; phosphatidylinositol 3-kinase complex; Protein sevenless; Receptor tyrosine-protein kinase erbB-3; Pro-neuregulin-1, membrane-bound isoform; Receptor tyrosine-protein kinase erbB-2]
E23HP PI3Ka[Phosphoprotein; phosphatidylinositol 3-kinase complex; Receptor tyrosine-protein kinase erbB-3; Pro-neuregulin-1, membrane-bound isoform; Receptor tyrosine-protein kinase erbB-2]
MEKP[Dual specificity mitogen-activated protein kinase kinase 1; Phosphoprotein]
Akt PI P PP2A[Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; RAC-alpha serine/threonine-protein kinase; 24755492; Phosphoprotein]
PIP3[1-phosphatidyl-1D-myo-inositol 3,4,5-trisphosphate; Phosphatidylinositol-3,4,5-trisphosphate; 24755492]
Akt PIP3 PP2A[RAC-alpha serine/threonine-protein kinase; Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; 24755492]
PI2[prostaglandin I2; Prostaglandin I2]
PI3K[phosphatidylinositol 3-kinase complex]
MEK[Dual specificity mitogen-activated protein kinase kinase 1]
ERKPP[Mitogen-activated protein kinase 3; Phosphoprotein]
PTEN bpV[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN; AAH05821.1; 16760324]
E23HP[Protein sevenless; Receptor tyrosine-protein kinase erbB-2; Pro-neuregulin-1, membrane-bound isoform; Receptor tyrosine-protein kinase erbB-3; Phosphoprotein]
PP2A[Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform]
MEKPP[Dual specificity mitogen-activated protein kinase kinase 1; Phosphoprotein]
PI3Ka PIP3[phosphatidylinositol 3-kinase complex; 24755492]
PTENP[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN; Phosphoprotein]
Akt PI PP PP2A[RAC-alpha serine/threonine-protein kinase; Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; 24755492; Phosphoprotein]
Raf[RAF proto-oncogene serine/threonine-protein kinase]
RasGTP[GTP; GTPase KRas]
E23H C[Protein sevenless; Receptor tyrosine-protein kinase erbB-2; Pro-neuregulin-1, membrane-bound isoform; Receptor tyrosine-protein kinase erbB-3; cytoplasm]
E2 Per[Protein sevenless; Receptor tyrosine-protein kinase erbB-2; AAC37531.1; CHEMBL2007641]
MEK PP2A[Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; Dual specificity mitogen-activated protein kinase kinase 1]
Akt[RAC-alpha serine/threonine-protein kinase]
RasGDP[GDP; GTPase KRas]
PI3Ka[phosphatidylinositol 3-kinase complex]
PTEN PTEN[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN; protein complex]
Akt PI PP[RAC-alpha serine/threonine-protein kinase; 24755492; Phosphoprotein]
PTEN PI[prostaglandin I2; Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN]
E23HP ShcP[Protein sevenless; SHC-transforming protein 1; Receptor tyrosine-protein kinase erbB-2; Pro-neuregulin-1, membrane-bound isoform; Receptor tyrosine-protein kinase erbB-3; Phosphoprotein]
Akt PI P[Phosphoprotein; RAC-alpha serine/threonine-protein kinase; 24755492]
E2[Protein sevenless; Receptor tyrosine-protein kinase erbB-2]
ERKP[Mitogen-activated protein kinase 3; Phosphoprotein]
PTEN PIP3[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN; 24755492]
GS[Growth factor receptor-bound protein 2; Son of sevenless homolog 1; Son of sevenless homolog 2]
E23H[Protein sevenless; Receptor tyrosine-protein kinase erbB-3; Pro-neuregulin-1, membrane-bound isoform; Receptor tyrosine-protein kinase erbB-2]
E23HP ShGS[Protein sevenless; SHC-transforming protein 1; Receptor tyrosine-protein kinase erbB-2; Pro-neuregulin-1, membrane-bound isoform; Receptor tyrosine-protein kinase erbB-3; Son of sevenless homolog 2; Son of sevenless homolog 1; Growth factor receptor-bound protein 2; Phosphoprotein]
PTENP PTEN[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN; Phosphoprotein; protein complex]
ERK[Mitogen-activated protein kinase 3]
MEKP PP2A[Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; Dual specificity mitogen-activated protein kinase kinase 1; Phosphoprotein]

Farhy2007_hGHregulation: MODEL0912096133v0.0.1

This a model from the article: Model-projected mechanistic bases for sex differences in growth hormone regulation in h…

Details

Models of physiological systems facilitate rational experimental design, inference, and prediction. A recent construct of regulated growth hormone (GH) secretion interlinks the actions of GH-releasing hormone (GHRH), somatostatin (SRIF), and GH secretagogues (GHS) with GH feedback in the rat (Farhy LS, Veldhuis JD. Am J Physiol Regul Integr Comp Physiol 288: R1649-R1663, 2005). In contrast, no comparable formalism exists to explicate GH dynamics in any other species. The present analyses explore whether a unifying model structure can represent species- and sex-defined distinctions in the human and rodent. The consensus principle that GHRH and GHS synergize in vivo but not in vitro was explicable by assuming that GHS 1) evokes GHRH release from the brain, 2) opposes inhibition by SRIF both in the hypothalamus and on the pituitary gland, and 3) stimulates pituitary GH release directly and additively with GHRH. The gender-selective principle that GH pulses are larger and more irregular in women than men was conferrable by way of 4) higher GHRH potency and 5) greater GHS efficacy. The overall construct predicts GHRH/GHS synergy in the human only in the presence of SRIF when the brain-pituitary nexus is intact, larger and more irregular GH pulses in women, and observed gender differences in feedback by GH and the single and paired actions of GHRH, GHS, and SRIF. The proposed model platform should enhance the framing and interpretation of novel clinical hypotheses and create a basis for interspecies generalization of GH-axis regulation. link: http://identifiers.org/pubmed/17185408

Farhy2009_GlucagonCounterRegulationModel: MODEL1112110002v0.0.1

This a model from the article: Pancreatic network control of glucagon secretion and counterregulation. Farhy LS, McC…

Details

Glucagon counterregulation (GCR) is a key protection against hypoglycemia compromised in insulinopenic diabetes by an unknown mechanism. In this work, we present an interdisciplinary approach to the analysis of the GCR control mechanisms. Our results indicate that a pancreatic network which unifies a few explicit interactions between the major islet peptides and blood glucose (BG) can replicate the normal GCR axis and explain its impairment in diabetes. A key and novel component of this network is an alpha-cell auto-feedback, which drives glucagon pulsatility and mediates triggering of pulsatile GCR by hypoglycemia via a switch-off of the beta-cell suppression of the alpha-cells. We have performed simulations based on our models of the endocrine pancreas which explain the in vivo GCR response to hypoglycemia of the normal pancreas and the enhancement of defective pulsatile GCR in beta-cell deficiency by switch-off of intrapancreatic alpha-cell suppressing signals. The models also predicted that reduced insulin secretion decreases and delays the GCR. In conclusion, based on experimental data we have developed and validated a model of the normal GCR control mechanisms and their dysregulation in insulin deficient diabetes. One advantage of this construct is that all model components are clinically measurable, thereby permitting its transfer, validation, and application to the study of the GCR abnormalities of the human endocrine pancreas in vivo. link: http://identifiers.org/pubmed/19897107

Fassoni2019 - Oncogenesis encompassing mutations and genetic instability: BIOMD0000000807v0.0.1

This model describes the multistep process that transform a normal cell and its descendants into a malignant tumour by c…

Details

Tumorigenesis has been described as a multistep process, where each step is associated with a genetic alteration, in the direction to progressively transform a normal cell and its descendants into a malignant tumour. Into this work, we propose a mathematical model for cancer onset and development, considering three populations: normal, premalignant and cancer cells. The model takes into account three hallmarks of cancer: self-sufficiency on growth signals, insensibility to anti-growth signals and evading apoptosis. By using a nonlinear expression to describe the mutation from premalignant to cancer cells, the model includes genetic instability as an enabling characteristic of tumour progression. Mathematical analysis was performed in detail. Results indicate that apoptosis and tissue repair system are the first barriers against tumour progression. One of these mechanisms must be corrupted for cancer to develop from a single mutant cell. The results also show that the presence of aggressive cancer cells opens way to survival of less adapted premalignant cells. Numerical simulations were performed with parameter values based on experimental data of breast cancer, and the necessary time taken for cancer to reach a detectable size from a single mutant cell was estimated with respect to some parameters. We find that the rates of apoptosis and mutations have a large influence on the pace of tumour progression and on the time it takes to become clinically detectable. link: http://identifiers.org/pubmed/29947770

Parameters:

NameDescription
xi_G = 0.01 1/msReaction: normalized_pre_cancer_cell_g =>, Rate Law: compartment*xi_G*normalized_pre_cancer_cell_g
r_N = 1000000.0 1/ms; K = 1.0E8Reaction: => normalized_normal_cell_n, Rate Law: compartment*r_N/K
mu_A = 0.01 1/msReaction: normalized_cancer_cell_a =>, Rate Law: compartment*mu_A*normalized_cancer_cell_a
r_G = 0.05 1/msReaction: => normalized_pre_cancer_cell_g, Rate Law: compartment*r_G*normalized_pre_cancer_cell_g
beta_1 = 3.5E-10 1/ms; K = 1.0E8Reaction: normalized_normal_cell_n => ; normalized_cancer_cell_a, Rate Law: compartment*beta_1*K*normalized_normal_cell_n*normalized_cancer_cell_a
beta_4 = 0.0 1/msReaction: normalized_normal_cell_n => ; normalized_pre_cancer_cell_g, Rate Law: compartment*beta_4*normalized_normal_cell_n*normalized_pre_cancer_cell_g
beta_6 = 0.0 1/msReaction: normalized_cancer_cell_a => ; normalized_pre_cancer_cell_g, Rate Law: compartment*beta_6*normalized_cancer_cell_a*normalized_pre_cancer_cell_g
beta_3 = 3.5E-10 1/ms; K = 1.0E8Reaction: normalized_cancer_cell_a => ; normalized_normal_cell_n, Rate Law: compartment*beta_3*K*normalized_normal_cell_n*normalized_cancer_cell_a
beta_2 = 3.5E-10 1/ms; K = 1.0E8Reaction: normalized_pre_cancer_cell_g => ; normalized_normal_cell_n, Rate Law: compartment*beta_2*K*normalized_normal_cell_n*normalized_pre_cancer_cell_g
beta_5 = 0.0 1/msReaction: normalized_pre_cancer_cell_g => ; normalized_cancer_cell_a, Rate Law: compartment*beta_5*normalized_cancer_cell_a*normalized_pre_cancer_cell_g
mu_G = 0.01 1/msReaction: normalized_pre_cancer_cell_g =>, Rate Law: compartment*mu_G*normalized_pre_cancer_cell_g
K_A = 1.0E7 1; r_A = 0.05 1/ms; K = 1.0E8Reaction: => normalized_cancer_cell_a, Rate Law: compartment*r_A*normalized_cancer_cell_a*(1-normalized_cancer_cell_a/(K_A/K))
delta = 1.0E-5 1/ms; xi = 1000.0 1; K = 1.0E8Reaction: normalized_pre_cancer_cell_g => normalized_cancer_cell_a, Rate Law: compartment*delta*normalized_pre_cancer_cell_g^2/(xi/K+normalized_pre_cancer_cell_g)
mu_N = 0.01 1/msReaction: normalized_normal_cell_n =>, Rate Law: compartment*mu_N*normalized_normal_cell_n
xi_A = 0.006 1/msReaction: normalized_cancer_cell_a =>, Rate Law: compartment*xi_A*normalized_cancer_cell_a

States:

NameDescription
normalized normal cell n[Cell; Healthy]
normalized pre cancer cell g[Breast Cancer Cell; Prior]
normalized cancer cell a[Breast Cancer Cell]

FatehiChenar2018 - Mathematical model of immune response to hepatitis B: BIOMD0000000848v0.0.1

This is a mathematical model describing the dynamics of the immune response to hepatitis B, which takes into account con…

Details

A new detailed mathematical model for dynamics of immune response to hepatitis B is proposed, which takes into account contributions from innate and adaptive immune responses, as well as cytokines. Stability analysis of different steady states is performed to identify parameter regions where the model exhibits clearance of infection, maintenance of a chronic infection, or periodic oscillations. Effects of nucleoside analogues and interferon treatments are analysed, and the critical drug efficiency is determined. link: http://identifiers.org/pubmed/29574141

Parameters:

NameDescription
q = 5.0Reaction: => A; V, Rate Law: compartment*q*V
c = 0.67Reaction: V =>, Rate Law: compartment*c*V
s_2 = 0.6; mu_2 = 0.14; s_1_prime = 1.9; s_2_prime = 2.0; mu_1 = 5.0; s_1 = 1.5Reaction: I => ; F_1, F_2, N, E, Rate Law: compartment*(mu_1*(1+s_1*F_1+s_2*F_2)*N+mu_2*(1+s_1_prime*F_1+s_2_prime*F_2)*E)*I
delta_2 = 5.16Reaction: F_2 =>, Rate Law: compartment*delta_2*F_2
r_e = 0.5Reaction: => E, Rate Law: compartment*r_e*E*(1-E)
d = 0.003Reaction: => T, Rate Law: compartment*d*(1-T)
psi_2 = 21.0Reaction: I => R; F_2, Rate Law: compartment*psi_2*I*F_2
psi_1 = 14.0Reaction: T => R; F_1, F_2, Rate Law: compartment*psi_1*T*(F_1+F_2)
delta_1 = 4.9Reaction: F_1 =>, Rate Law: compartment*delta_1*F_1
q_1 = 0.8; q_2 = 0.6Reaction: => N; F_1, F_2, Rate Law: compartment*(q_1*F_1+q_2*F_2)*N
k = 8.0Reaction: V + A =>, Rate Law: compartment*k*V*A
p_1 = 1.0Reaction: => F_1; I, Rate Law: compartment*p_1*I
p_2 = 0.5Reaction: => F_2; E, Rate Law: compartment*p_2*E
rho = 5.0Reaction: R => T, Rate Law: compartment*rho*R
delta = 0.56Reaction: I =>, Rate Law: compartment*delta*I
p = 20.0; s_3 = 1.7; s_4 = 1.0Reaction: => V; I, F_1, F_2, Rate Law: compartment*I*p/(1+s_3*F_1+s_4*F_2)
p_3 = 3.0Reaction: => F_2; N, Rate Law: compartment*p_3*N
beta = 7.0Reaction: T => I; V, Rate Law: compartment*beta*V*T
alpha = 1.0Reaction: => E; I, Rate Law: compartment*alpha*I*E
d_a = 0.332Reaction: => A, Rate Law: compartment*d_a*(1-A)

States:

NameDescription
I[C14215; infected cell]
F 2[PR:000024990]
T[hepatocyte]
A[C62795]
N[natural killer cell]
V[C14215]
F 1[PR:000025848]
E[cytotoxic T cell]
R[hepatocyte; C38014]

Fatma2018 - Model of central carbon metabolism of E. coli with additional reactions to investigate potential routes of alkane synthesis: MODEL1802080001v0.0.1

Model of cetnral carbon metabolism of E. coli with additional reactions to investigate potential routes of alkane synthe…

Details

Biologically-derived hydrocarbons are considered to have great potential as next-generation biofuels owing to the similarity of their chemical properties to contemporary diesel and jet fuels. However, the low yield of these hydrocarbons in biotechnological production is a major obstacle for commercialization. Several genetic and process engineering approaches have been adopted to increase the yield of hydrocarbon, but a model driven approach has not been implemented so far. Here, we applied a constraint-based metabolic modeling approach in which a variable demand for alkane biosynthesis was imposed, and co-varying reactions were considered as potential targets for further engineering of an E. coli strain already expressing cyanobacterial enzymes towards higher chain alkane production. The reactions that co-varied with the imposed alkane production were found to be mainly associated with the pentose phosphate pathway (PPP) and the lower half of glycolysis. An optimal modeling solution was achieved by imposing increased flux through the reaction catalyzed by glucose-6-phosphate dehydrogenase (zwf) and iteratively removing 7 reactions from the network, leading to an alkane yield of 94.2% of the theoretical maximum conversion determined by in silico analysis at a given biomass rate. To validate the in silico findings, we first performed pathway optimization of the cyanobacterial enzymes in E. coli via different dosages of genes, promoting substrate channelling through protein fusion and inducing substantial equivalent protein expression, which led to a 36-fold increase in alka(e)ne production from 2.8mg/L to 102mg/L. Further, engineering of E. coli based on in silico findings, including biomass constraint, led to an increase in the alka(e)ne titer to 425mg/L (major components being 249mg/L pentadecane and 160mg/L heptadecene), a 148.6-fold improvement over the initial strain, respectively; with a yield of 34.2% of the theoretical maximum. The impact of model-assisted engineering was also tested for the production of long chain fatty alcohol, another commercially important molecule sharing the same pathway while differing only at the terminal reaction, and a titer of 1506mg/L was achieved with a yield of 86.4% of the theoretical maximum. Moreover, the model assisted engineered strains had produced 2.54g/L and 12.5g/L of long chain alkane and fatty alcohol, respectively, in the bioreactor under fed-batch cultivation condition. Our study demonstrated successful implementation of a combined in silico modeling approach along with the pathway and process optimization in achieving the highest reported titers of long chain hydrocarbons in E. coli. link: http://identifiers.org/pubmed/29408291

Faville2008_UPdepolarization: MODEL0912044015v0.0.1

This a model from the article: A biophysically based mathematical model of unitary potential activity in interstitial…

Details

Unitary potential (UP) depolarizations are the basic intracellular events responsible for pacemaker activity in interstitial cells of Cajal (ICCs), and are generated at intracellular sites termed "pacemaker units". In this study, we present a mathematical model of the transmembrane ion flows and intracellular Ca(2+) dynamics from a single ICC pacemaker unit acting at near-resting membrane potential. This model quantitatively formalizes the framework of a novel ICC pacemaking mechanism that has recently been proposed. Model simulations produce spontaneously rhythmic UP depolarizations with an amplitude of approximately 3 mV at a frequency of 0.05 Hz. The model predicts that the main inward currents, carried by a Ca(2+)-inhibited nonselective cation conductance, are activated by depletion of sub-plasma-membrane [Ca(2+)] caused by sarcoendoplasmic reticulum calcium ATPase Ca(2+) sequestration. Furthermore, pacemaker activity predicted by our model persists under simulated voltage clamp and is independent of [IP(3)] oscillations. The model presented here provides a basis to quantitatively analyze UP depolarizations and the biophysical mechanisms underlying their production. link: http://identifiers.org/pubmed/18339738

Feala2007_dros_mel_central_metabolism: MODEL2784700357v0.0.1

Model described in: **Flexibility in energy metabolism supports hypoxia tolerance in Drosophila flight muscle: metabol…

Details

The fruitfly Drosophila melanogaster offers promise as a genetically tractable model for studying adaptation to hypoxia at the cellular level, but the metabolic basis for extreme hypoxia tolerance in flies is not well known. Using (1)H NMR spectroscopy, metabolomic profiles were collected under hypoxia. Accumulation of lactate, alanine, and acetate suggested that these are the major end products of anaerobic metabolism in the fly. A constraint-based model of ATP-producing pathways was built using the annotated genome, existing models, and the literature. Multiple redundant pathways for producing acetate and alanine were added and simulations were run in order to find a single optimal strategy for producing each end product. System-wide adaptation to hypoxia was then investigated in silico using the refined model. Simulations supported the hypothesis that the ability to flexibly convert pyruvate to these three by-products might convey hypoxia tolerance by improving the ATP/H(+) ratio and efficiency of glucose utilization. link: http://identifiers.org/pubmed/17437024

Feist2006_methanogenesis_OptiAcetate: MODEL5662377562v0.0.1

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

Details

We present a genome-scale metabolic model for the archaeal methanogen Methanosarcina barkeri. We characterize the metabolic network and compare it to reconstructions from the prokaryotic, eukaryotic and archaeal domains. Using the model in conjunction with constraint-based methods, we simulate the metabolic fluxes and resulting phenotypes induced by different environmental and genetic conditions. This represents the first large-scale simulation of either a methanogen or an archaeal species. Model predictions are validated by comparison to experimental growth measurements and phenotypes of M. barkeri on different substrates. The predicted growth phenotypes for wild type and mutants of the methanogenic pathway have a high level of agreement with experimental findings. We further examine the efficiency of the energy-conserving reactions in the methanogenic pathway, specifically the Ech hydrogenase reaction, and determine a stoichiometry for the nitrogenase reaction. This work demonstrates that a reconstructed metabolic network can serve as an analysis platform to predict cellular phenotypes, characterize methanogenic growth, improve the genome annotation and further uncover the metabolic characteristics of methanogenesis. link: http://identifiers.org/pubmed/16738551