SBMLBioModels: S - T

S


Stavrum2013 - Tryptophan Metabolism in Liver: BIOMD0000000602v0.0.1

Stavrum2013 - Tryptophan Metabolism in LiverThis model is described in the article: [Model of tryptophan metabolism, re…

Details

Tryptophan is utilized in various metabolic routes including protein synthesis, serotonin, and melatonin synthesis and the kynurenine pathway. Perturbations in these pathways have been associated with neurodegenerative diseases and cancer. Here we present a comprehensive kinetic model of the complex network of human tryptophan metabolism based upon existing kinetic data for all enzymatic conversions and transporters. By integrating tissue-specific expression data, modeling tryptophan metabolism in liver and brain returned intermediate metabolite concentrations in the physiological range. Sensitivity and metabolic control analyses identified expected key enzymes to govern fluxes in the branches of the network. Combining tissue-specific models revealed a considerable impact of the kynurenine pathway in liver on the concentrations of neuroactive derivatives in the brain. Moreover, using expression data from a cancer study predicted metabolite changes that resembled the experimental observations. We conclude that the combination of the kinetic model with expression data represents a powerful diagnostic tool to predict alterations in tryptophan metabolism. The model is readily scalable to include more tissues, thereby enabling assessment of organismal tryptophan metabolism in health and disease. link: http://identifiers.org/pubmed/24129579

Parameters:

NameDescription
AADAT_Km_Lkynr = 4.7; AADAT_Km_hLkynr = 3.8; AADAT_kcat_Lkynr = 9.76; scaling = 1.0; AADAT_E_T_kat3 = 15588.2099609375Reaction: M_Lkynr_c + M_akg_c => M_kynate_c + M_glu_DASH_L_c; M_hLkynr_c, M_Lkynr_c, Rate Law: Cytosol*AADAT_E_T_kat3*AADAT_kcat_Lkynr*scaling*M_Lkynr_c*AADAT_Km_hLkynr/(AADAT_Km_Lkynr*AADAT_Km_hLkynr+AADAT_Km_Lkynr*M_hLkynr_c+AADAT_Km_hLkynr*M_Lkynr_c)
AFMID_Km_Lfmkynr = 0.05; kcat_A=100.0; AFMID_Km_nformanth = 0.211; scaling = 1.0; AFMID_Km_5hoxnfky = 0.4; AFMID_E_T = 15820.2158203125Reaction: M_Lfmkynr_c + M_h2o_c => M_for_c + M_Lkynr_c; M_5hoxnfkyn_c, M_nformanth_c, M_Lfmkynr_c, Rate Law: Cytosol*AFMID_E_T*kcat_A*scaling*M_Lfmkynr_c*AFMID_Km_5hoxnfky*AFMID_Km_nformanth/(AFMID_Km_Lfmkynr*AFMID_Km_5hoxnfky*AFMID_Km_nformanth+AFMID_Km_5hoxnfky*AFMID_Km_nformanth*M_Lfmkynr_c+AFMID_Km_Lfmkynr*AFMID_Km_nformanth*M_5hoxnfkyn_c+AFMID_Km_Lfmkynr*AFMID_Km_5hoxnfky*M_nformanth_c)
MAO_Km_srtn = 0.43; MAOA_E_T = 137204.8125; kcat_B=18.6; scaling = 1.0; MAO_Km_trypta = 0.033Reaction: M_srtn_c + M_h2o_c + M_o2_c => M_5hoxindact_c + M_h2o2_c + M_nh4_c; M_trypta_c, M_5hxkyn_c, M_srtn_c, Rate Law: Cytosol*MAOA_E_T*kcat_B*scaling*M_srtn_c*MAO_Km_trypta/(MAO_Km_srtn*MAO_Km_trypta+MAO_Km_srtn*M_trypta_c+MAO_Km_trypta*M_srtn_c)
AADAT_E_T_kat2 = 7744.3154296875; AADAT_Km_Lkynr = 4.7; AADAT_Km_hLkynr = 3.8; AADAT_kcat_hLkynr = 1.7; scaling = 1.0Reaction: M_hLkynr_c + M_akg_c => M_Xanthurenate + M_glu_DASH_L_c; M_Lkynr_c, M_hLkynr_c, Rate Law: Cytosol*AADAT_E_T_kat2*AADAT_kcat_hLkynr*scaling*M_hLkynr_c*AADAT_Km_Lkynr/(AADAT_Km_hLkynr*AADAT_Km_Lkynr+AADAT_Km_hLkynr*M_Lkynr_c+AADAT_Km_Lkynr*M_hLkynr_c)
Kb=0.273; kcat=0.18; E_T=235.128; scaling = 1.0; Ka=0.0403Reaction: M_thbpt + M_trp_DASH_L_c + M_o2_c => M_5htrp_c + M_dhbpt_c + M_h2o_c; M_o2_c, M_thbpt, M_trp_DASH_L_c, Rate Law: Cytosol*kcat*E_T*M_trp_DASH_L_c*M_o2_c*M_thbpt*scaling/(Ka*Kb+Kb*M_trp_DASH_L_c+Ka*M_o2_c+M_trp_DASH_L_c*M_o2_c)
e2=4.0; e1=2.0; k1=5.6667E-5Reaction: M_3hanthrn_c + M_o2_c => M_Cinnavalininate_c + M_o2s_c + M_h2o2_c + M_h_c; M_3hanthrn_c, M_o2_c, Rate Law: Cytosol*k1*M_3hanthrn_c^e1*M_o2_c^e2
IMNT_Km_trypta = 0.27; IMNT_E_T = 4186.5874023438; IMNT_Km_nmtrpta = 0.086; scaling = 1.0; IMNT_Km_srtn = 1.38; kcat_A=0.4Reaction: M_amet_c + M_trypta_c => M_ahcys_c + M_nmtrpta_c; M_nmtrpta_c, M_srtn_c, M_trypta_c, Rate Law: Cytosol*IMNT_E_T*kcat_A*scaling*M_trypta_c*IMNT_Km_nmtrpta*IMNT_Km_srtn/(IMNT_Km_trypta*IMNT_Km_nmtrpta*IMNT_Km_srtn+IMNT_Km_nmtrpta*IMNT_Km_srtn*M_trypta_c+IMNT_Km_trypta*IMNT_Km_srtn*M_nmtrpta_c+IMNT_Km_trypta*IMNT_Km_nmtrpta*M_srtn_c)
Transporter_E_T_Slc7a8 = 2226.3728027344; Transporter_Km_Lkynr = 0.032; Transporter_kcat_Trp = 1.3; Transporter_Km_Trp_Slc7a8 = 0.0573; scaling = 1.0Reaction: TRP_ex => M_trp_DASH_L_c; M_Lkynr_ex, M_Lkynr_c, M_trp_DASH_L_c, TRP_ex, Rate Law: Cytosol*scaling*(Transporter_E_T_Slc7a8*Transporter_kcat_Trp*TRP_ex/Transporter_Km_Trp_Slc7a8-Transporter_E_T_Slc7a8*Transporter_kcat_Trp*M_trp_DASH_L_c/Transporter_Km_Trp_Slc7a8)/(1+TRP_ex/Transporter_Km_Trp_Slc7a8+M_Lkynr_ex/Transporter_Km_Lkynr+M_trp_DASH_L_c/Transporter_Km_Trp_Slc7a8+M_Lkynr_c/Transporter_Km_Lkynr)
AADAT_E_T_kat2 = 7744.3154296875; AADAT_Km_Lkynr = 4.7; AADAT_Km_hLkynr = 3.8; AADAT_kcat_Lkynr = 9.76; scaling = 1.0Reaction: M_Lkynr_c + M_akg_c => M_kynate_c + M_glu_DASH_L_c; M_hLkynr_c, M_Lkynr_c, Rate Law: Cytosol*AADAT_E_T_kat2*AADAT_kcat_Lkynr*scaling*M_Lkynr_c*AADAT_Km_hLkynr/(AADAT_Km_Lkynr*AADAT_Km_hLkynr+AADAT_Km_Lkynr*M_hLkynr_c+AADAT_Km_hLkynr*M_Lkynr_c)
AADAT_Km_Lkynr = 4.7; AADAT_Km_hLkynr = 3.8; AADAT_kcat_hLkynr = 1.7; scaling = 1.0; AADAT_E_T_kat3 = 15588.2099609375Reaction: M_hLkynr_c + M_akg_c => M_Xanthurenate + M_glu_DASH_L_c; M_Lkynr_c, M_hLkynr_c, Rate Law: Cytosol*AADAT_E_T_kat3*AADAT_kcat_hLkynr*scaling*M_hLkynr_c*AADAT_Km_Lkynr/(AADAT_Km_hLkynr*AADAT_Km_Lkynr+AADAT_Km_hLkynr*M_Lkynr_c+AADAT_Km_Lkynr*M_hLkynr_c)
k1=2.5E-4Reaction: M_cmusa_c => M_quln_c; M_cmusa_c, Rate Law: Cytosol*k1*M_cmusa_c
Transporter_Km_Lkynr = 0.032; Transporter_kcat_Trp = 1.3; Transporter_Km_Trp_Slc7a5 = 0.019; scaling = 1.0; Transporter_E_T_Slc7a5 = 1961.5135498047Reaction: TRP_ex => M_trp_DASH_L_c; M_Lkynr_ex, M_Lkynr_c, M_trp_DASH_L_c, TRP_ex, Rate Law: Cytosol*scaling*(Transporter_E_T_Slc7a5*Transporter_kcat_Trp*TRP_ex/Transporter_Km_Trp_Slc7a5-Transporter_E_T_Slc7a5*Transporter_kcat_Trp*M_trp_DASH_L_c/Transporter_Km_Trp_Slc7a5)/(1+TRP_ex/Transporter_Km_Trp_Slc7a5+M_Lkynr_ex/Transporter_Km_Lkynr+M_trp_DASH_L_c/Transporter_Km_Trp_Slc7a5+M_Lkynr_c/Transporter_Km_Lkynr)
kcat=64.0; Ka=0.016; Kb=0.615; scaling = 1.0; E_T=10308.4Reaction: M_3hanthrn_c + M_o2_c => M_cmusa_c; M_quln_c, M_anth_c, M_3hanthrn_c, M_o2_c, Rate Law: Cytosol*kcat*E_T*M_3hanthrn_c*M_o2_c*scaling/(Ka*Kb+Ka*M_o2_c+Kb*M_3hanthrn_c+M_3hanthrn_c*M_o2_c)
E_T=943912.0; Ka=0.222; scaling = 1.0; Kb=0.037; kcat=1.4Reaction: M_trp_DASH_L_c + M_o2_c => M_Lfmkynr_c; M_o2_c, M_trp_DASH_L_c, Rate Law: Cytosol*kcat*E_T*M_trp_DASH_L_c*M_o2_c*scaling/(Ka*Kb+Ka*M_o2_c+Kb*M_trp_DASH_L_c+M_trp_DASH_L_c*M_o2_c)
AADAT_E_T_kat1 = 9455.1357421875; AADAT_Km_Lkynr = 4.7; AADAT_Km_hLkynr = 3.8; AADAT_kcat_hLkynr = 1.7; scaling = 1.0Reaction: M_hLkynr_c + M_akg_c => M_Xanthurenate + M_glu_DASH_L_c; M_Lkynr_c, M_hLkynr_c, Rate Law: Cytosol*AADAT_E_T_kat1*AADAT_kcat_hLkynr*scaling*M_hLkynr_c*AADAT_Km_Lkynr/(AADAT_Km_hLkynr*AADAT_Km_Lkynr+AADAT_Km_hLkynr*M_Lkynr_c+AADAT_Km_Lkynr*M_hLkynr_c)
E_T=4658.65; kcat=42.9; scaling = 1.0; Km=0.0677Reaction: M_atp_c + M_h_c + M_nicrnt_c => M_dnad_c + M_ppi_c; M_atp_c, M_h_c, M_nicrnt_c, Rate Law: Cytosol*E_T*kcat*M_nicrnt_c*M_h_c*M_atp_c*scaling/(Km+M_nicrnt_c)
KYNU_E_T = 56601.7578125; KYNU_Km_Lkynr = 0.495; KYNU_Km_Lfmkynr = 2.2; kcat_A=0.013; scaling = 1.0; KYNU_Km_hLkynr = 0.028Reaction: M_Lfmkynr_c + M_h2o_c => M_nformanth_c + M_ala_DASH_L_c; M_Lkynr_c, M_hLkynr_c, M_Lfmkynr_c, Rate Law: Cytosol*KYNU_E_T*kcat_A*scaling*M_Lfmkynr_c*KYNU_Km_Lkynr*KYNU_Km_hLkynr/(KYNU_Km_Lfmkynr*KYNU_Km_Lkynr*KYNU_Km_hLkynr+KYNU_Km_Lkynr*KYNU_Km_hLkynr*M_Lfmkynr_c+KYNU_Km_Lfmkynr*KYNU_Km_hLkynr*M_Lkynr_c+KYNU_Km_Lfmkynr*KYNU_Km_Lkynr*M_hLkynr_c)
IMNT_Km_trypta = 0.27; IMNT_E_T = 4186.5874023438; IMNT_Km_nmtrpta = 0.086; scaling = 1.0; IMNT_Km_srtn = 1.38; kcat_A=0.176Reaction: M_amet_c + M_nmtrpta_c => M_ahcys_c + M_nndmtrpta_c; M_srtn_c, M_trypta_c, M_nmtrpta_c, Rate Law: Cytosol*IMNT_E_T*kcat_A*scaling*M_nmtrpta_c*IMNT_Km_srtn*IMNT_Km_trypta/(IMNT_Km_nmtrpta*IMNT_Km_srtn*IMNT_Km_trypta+IMNT_Km_srtn*IMNT_Km_trypta*M_nmtrpta_c+IMNT_Km_nmtrpta*IMNT_Km_trypta*M_srtn_c+IMNT_Km_nmtrpta*IMNT_Km_srtn*M_trypta_c)
Transporter_Km_Lkynr = 0.032; Transporter_Km_Trp_Slc7a5 = 0.019; scaling = 1.0; Transporter_kcat_Lkynr = 1.3; Transporter_E_T_Slc7a5 = 1961.5135498047Reaction: M_Lkynr_c => M_Lkynr_ex; M_trp_DASH_L_c, TRP_ex, M_Lkynr_c, M_Lkynr_ex, Rate Law: Cytosol*scaling*(Transporter_E_T_Slc7a5*Transporter_kcat_Lkynr*M_Lkynr_c/Transporter_Km_Lkynr-Transporter_E_T_Slc7a5*Transporter_kcat_Lkynr*M_Lkynr_ex/Transporter_Km_Lkynr)/(1+M_Lkynr_c/Transporter_Km_Lkynr+M_trp_DASH_L_c/Transporter_Km_Trp_Slc7a5+M_Lkynr_ex/Transporter_Km_Lkynr+TRP_ex/Transporter_Km_Trp_Slc7a5)
Transporter_E_T_Slc7a8 = 2226.3728027344; Transporter_Km_Lkynr = 0.032; Transporter_Km_Trp_Slc7a8 = 0.0573; scaling = 1.0; Transporter_kcat_Lkynr = 1.3Reaction: M_Lkynr_c => M_Lkynr_ex; M_trp_DASH_L_c, TRP_ex, M_Lkynr_c, M_Lkynr_ex, Rate Law: Cytosol*scaling*(Transporter_E_T_Slc7a8*Transporter_kcat_Lkynr*M_Lkynr_c/Transporter_Km_Lkynr-Transporter_E_T_Slc7a8*Transporter_kcat_Lkynr*M_Lkynr_ex/Transporter_Km_Lkynr)/(1+M_Lkynr_c/Transporter_Km_Lkynr+M_trp_DASH_L_c/Transporter_Km_Trp_Slc7a8+M_Lkynr_ex/Transporter_Km_Lkynr+TRP_ex/Transporter_Km_Trp_Slc7a8)
Km=0.0065; E_T=48858.2; kcat=1.0; scaling = 1.0Reaction: M_cmusa_c => M_am6sa_c + M_co2_c; M_quln_c, M_kynate_c, M_cmusa_c, Rate Law: Cytosol*E_T*kcat*M_cmusa_c*scaling/(Km+M_cmusa_c)
DDC_E_T = 36074.9140625; DDC_Km_5htrp = 0.049; scaling = 1.0; kcat_B=2.0; DDC_Km_trp_DASH_L = 10.0Reaction: M_5htrp_c => M_srtn_c + M_co2_c; M_trp_DASH_L_c, M_5htrp_c, Rate Law: Cytosol*DDC_E_T*kcat_B*scaling*M_5htrp_c*DDC_Km_trp_DASH_L/(DDC_Km_5htrp*DDC_Km_trp_DASH_L+DDC_Km_5htrp*M_trp_DASH_L_c+DDC_Km_trp_DASH_L*M_5htrp_c)
Kb=0.109; scaling = 1.0; E_T=503.141; kcat=0.57; Ka=0.0228Reaction: M_thbpt + M_trp_DASH_L_c + M_o2_c => M_5htrp_c + M_dhbpt_c + M_h2o_c; M_o2_c, M_thbpt, M_trp_DASH_L_c, Rate Law: Cytosol*kcat*E_T*M_trp_DASH_L_c*M_o2_c*M_thbpt*scaling/(Ka*Kb+Kb*M_trp_DASH_L_c+Ka*M_o2_c+M_trp_DASH_L_c*M_o2_c)
Kb=0.023; E_T=138709.0; scaling = 1.0; Ka=0.022; kcat=0.255Reaction: M_h_c + M_prpp_c + M_quln_c => M_co2_c + M_nicrnt_c + M_ppi_c; M_h_c, M_prpp_c, M_quln_c, Rate Law: Cytosol*kcat*E_T*M_quln_c*M_prpp_c*M_h_c*scaling/(Ka*Kb+Kb*M_quln_c+Ka*M_prpp_c+M_quln_c*M_prpp_c)
AFMID_Km_Lfmkynr = 0.05; AFMID_Km_nformanth = 0.211; kcat_A=13.57; scaling = 1.0; AFMID_Km_5hoxnfky = 0.4; AFMID_E_T = 15820.2158203125Reaction: M_nformanth_c + M_h2o_c => M_for_c + M_anth_c; M_Lfmkynr_c, M_5hoxnfkyn_c, M_nformanth_c, Rate Law: Cytosol*AFMID_E_T*kcat_A*scaling*M_nformanth_c*AFMID_Km_Lfmkynr*AFMID_Km_5hoxnfky/(AFMID_Km_nformanth*AFMID_Km_Lfmkynr*AFMID_Km_5hoxnfky+AFMID_Km_Lfmkynr*AFMID_Km_5hoxnfky*M_nformanth_c+AFMID_Km_nformanth*AFMID_Km_5hoxnfky*M_Lfmkynr_c+AFMID_Km_nformanth*AFMID_Km_Lfmkynr*M_5hoxnfkyn_c)
KYNU_E_T = 56601.7578125; kcat_A=3.5; KYNU_Km_Lkynr = 0.495; KYNU_Km_Lfmkynr = 2.2; scaling = 1.0; KYNU_Km_hLkynr = 0.028Reaction: M_hLkynr_c + M_h2o_c => M_3hanthrn_c + M_ala_DASH_L_c; M_Lkynr_c, M_Lfmkynr_c, M_hLkynr_c, Rate Law: Cytosol*KYNU_E_T*kcat_A*scaling*M_hLkynr_c*KYNU_Km_Lkynr*KYNU_Km_Lfmkynr/(KYNU_Km_hLkynr*KYNU_Km_Lkynr*KYNU_Km_Lfmkynr+KYNU_Km_Lkynr*KYNU_Km_Lfmkynr*M_hLkynr_c+KYNU_Km_hLkynr*KYNU_Km_Lfmkynr*M_Lkynr_c+KYNU_Km_hLkynr*KYNU_Km_Lkynr*M_Lfmkynr_c)
MAO_Km_srtn = 0.43; kcat_B=3.5; MAOA_E_T = 137204.8125; scaling = 1.0; MAO_Km_trypta = 0.033Reaction: M_trypta_c + M_h2o_c + M_o2_c => M_id3acald_c + M_nh4_c + M_h2o2_c; M_srtn_c, M_trypta_c, Rate Law: Cytosol*MAOA_E_T*kcat_B*scaling*M_trypta_c*MAO_Km_srtn/(MAO_Km_trypta*MAO_Km_srtn+MAO_Km_trypta*M_srtn_c+MAO_Km_srtn*M_trypta_c)
IDO_Km_5htrp = 0.02; IDO_Km_O2 = 0.042; IDO_E_T = 453.4833679199; scaling = 1.0; IDO_Km_trp_DASH_L = 0.045; IDO_Km_srtn = 0.1; kcat=0.002Reaction: M_srtn_c + M_o2_c => M_f5hoxkyn_c; M_5htrp_c, M_trp_DASH_L_c, M_o2_c, M_srtn_c, Rate Law: Cytosol*kcat*IDO_E_T*M_srtn_c*M_o2_c*IDO_Km_5htrp*IDO_Km_trp_DASH_L*scaling/(IDO_Km_srtn*IDO_Km_O2*IDO_Km_5htrp*IDO_Km_trp_DASH_L+M_srtn_c*IDO_Km_O2*IDO_Km_5htrp*IDO_Km_trp_DASH_L+M_o2_c*IDO_Km_srtn*IDO_Km_5htrp*IDO_Km_trp_DASH_L+M_5htrp_c*IDO_Km_srtn*IDO_Km_O2*IDO_Km_trp_DASH_L+M_trp_DASH_L_c*IDO_Km_srtn*IDO_Km_O2*IDO_Km_5htrp+M_srtn_c*M_o2_c*IDO_Km_5htrp*IDO_Km_trp_DASH_L+M_5htrp_c*M_o2_c*IDO_Km_srtn*IDO_Km_trp_DASH_L+M_trp_DASH_L_c*M_o2_c*IDO_Km_srtn*IDO_Km_5htrp)
Kb=1.2; Ka=6.5; kcat=1.0; scaling = 1.0; E_T=2046.74Reaction: M_trp_DASH_L_c + M_h2o_c + M_o2_c => M_indpyr_c + M_nh4_c + M_h2o2_c; M_h2o_c, M_o2_c, M_trp_DASH_L_c, Rate Law: Cytosol*kcat*E_T*M_trp_DASH_L_c*M_o2_c*M_h2o_c*scaling/(Ka*Kb+Kb*M_trp_DASH_L_c+Ka*M_o2_c+M_trp_DASH_L_c*M_o2_c)
AANAT_E_T = 2770.9680175781; AANAT_Km_Srtn = 1.35; kcat_B=25.9; scaling = 1.0; AANAT_Km_trypta = 0.88Reaction: M_accoa_c + M_srtn_c => M_Nacsertn_c + M_coa_c + M_h_c; M_trypta_c, M_srtn_c, Rate Law: Cytosol*AANAT_E_T*kcat_B*scaling*M_srtn_c*AANAT_Km_trypta/(AANAT_Km_Srtn*AANAT_Km_trypta+AANAT_Km_Srtn*M_trypta_c+AANAT_Km_trypta*M_srtn_c)
MAO_Km_srtn = 0.43; MAOB_E_T = 294114.875; kcat_B=18.6; scaling = 1.0; MAO_Km_trypta = 0.033Reaction: M_srtn_c + M_h2o_c + M_o2_c => M_5hoxindact_c + M_h2o2_c + M_nh4_c; M_trypta_c, M_5hxkyn_c, M_srtn_c, Rate Law: Cytosol*MAOB_E_T*kcat_B*scaling*M_srtn_c*MAO_Km_trypta/(MAO_Km_srtn*MAO_Km_trypta+MAO_Km_srtn*M_trypta_c+MAO_Km_trypta*M_srtn_c)
IMNT_Km_trypta = 0.27; IMNT_E_T = 4186.5874023438; IMNT_Km_nmtrpta = 0.086; scaling = 1.0; IMNT_Km_srtn = 1.38; kcat_A=0.044Reaction: M_amet_c + M_srtn_c => M_ahcys_c + M_nmsrtn_c; M_nmtrpta_c, M_trypta_c, M_srtn_c, Rate Law: Cytosol*IMNT_E_T*kcat_A*scaling*M_srtn_c*IMNT_Km_nmtrpta*IMNT_Km_trypta/(IMNT_Km_srtn*IMNT_Km_nmtrpta*IMNT_Km_trypta+IMNT_Km_nmtrpta*IMNT_Km_trypta*M_srtn_c+IMNT_Km_srtn*IMNT_Km_trypta*M_nmtrpta_c+IMNT_Km_srtn*IMNT_Km_nmtrpta*M_trypta_c)
MAO_Km_srtn = 0.43; kcat_B=3.5; MAOB_E_T = 294114.875; scaling = 1.0; MAO_Km_trypta = 0.033Reaction: M_trypta_c + M_h2o_c + M_o2_c => M_id3acald_c + M_nh4_c + M_h2o2_c; M_srtn_c, M_trypta_c, Rate Law: Cytosol*MAOB_E_T*kcat_B*scaling*M_trypta_c*MAO_Km_srtn/(MAO_Km_trypta*MAO_Km_srtn+MAO_Km_trypta*M_srtn_c+MAO_Km_srtn*M_trypta_c)
Ka=0.0074; Kb=0.0011; kcat=1.1; E_T=15961.5; scaling = 1.0Reaction: M_atp_c + M_trp_DASH_L_c + M_trna_trp_c => M_amp_c + M_ppi_c + M_trp_L_trna_c; M_atp_c, M_trna_trp_c, M_trp_DASH_L_c, Rate Law: Cytosol*kcat*E_T*M_trp_DASH_L_c*M_trna_trp_c*M_atp_c*scaling/(Ka*Kb+Kb*M_trp_DASH_L_c+Ka*M_trna_trp_c+M_trp_DASH_L_c*M_trna_trp_c)
IDO_Km_5htrp = 0.02; IDO_Km_O2 = 0.042; IDO_E_T = 453.4833679199; scaling = 1.0; kcat=1.65; IDO_Km_trp_DASH_L = 0.045; IDO_Km_srtn = 0.1Reaction: M_trp_DASH_L_c + M_o2_c => M_Lfmkynr_c; M_5htrp_c, M_srtn_c, M_o2_c, M_trp_DASH_L_c, Rate Law: Cytosol*kcat*IDO_E_T*M_trp_DASH_L_c*M_o2_c*IDO_Km_5htrp*IDO_Km_srtn*scaling/(IDO_Km_trp_DASH_L*IDO_Km_O2*IDO_Km_5htrp*IDO_Km_srtn+M_trp_DASH_L_c*IDO_Km_O2*IDO_Km_5htrp*IDO_Km_srtn+M_o2_c*IDO_Km_trp_DASH_L*IDO_Km_5htrp*IDO_Km_srtn+M_5htrp_c*IDO_Km_trp_DASH_L*IDO_Km_O2*IDO_Km_srtn+M_srtn_c*IDO_Km_trp_DASH_L*IDO_Km_O2*IDO_Km_5htrp+M_trp_DASH_L_c*M_o2_c*IDO_Km_5htrp*IDO_Km_srtn+M_5htrp_c*M_o2_c*IDO_Km_trp_DASH_L*IDO_Km_srtn+M_srtn_c*M_o2_c*IDO_Km_trp_DASH_L*IDO_Km_5htrp)
KYNU_E_T = 56601.7578125; kcat_A=0.23; KYNU_Km_Lkynr = 0.495; KYNU_Km_Lfmkynr = 2.2; scaling = 1.0; KYNU_Km_hLkynr = 0.028Reaction: M_Lkynr_c + M_h2o_c => M_anth_c + M_ala_DASH_L_c; M_hLkynr_c, M_Lfmkynr_c, M_Lkynr_c, Rate Law: Cytosol*KYNU_E_T*kcat_A*scaling*M_Lkynr_c*KYNU_Km_hLkynr*KYNU_Km_Lfmkynr/(KYNU_Km_Lkynr*KYNU_Km_hLkynr*KYNU_Km_Lfmkynr+KYNU_Km_hLkynr*KYNU_Km_Lfmkynr*M_Lkynr_c+KYNU_Km_Lkynr*KYNU_Km_Lfmkynr*M_hLkynr_c+KYNU_Km_Lkynr*KYNU_Km_hLkynr*M_Lfmkynr_c)
Ka=0.1; E_T=9766.18; Kb=0.071; scaling = 1.0; Kc=0.153; kcat=2.2Reaction: M_Lkynr_c + M_o2_c + M_nadph_c + M_h_c => M_hLkynr_c + M_nadp_c + M_h2o_c; M_Lkynr_c, M_h_c, M_nadph_c, M_o2_c, Rate Law: Cytosol*kcat*E_T*M_Lkynr_c*M_o2_c*M_nadph_c*M_h_c*scaling/(Ka*Kb*Kc+M_Lkynr_c*Kb*Kc+M_o2_c*Ka*Kc+M_nadph_c*Ka*Kb+M_Lkynr_c*M_o2_c*Kc+M_o2_c*M_nadph_c*Ka+M_Lkynr_c*M_nadph_c*Kb+M_Lkynr_c*M_o2_c*M_nadph_c)
DDC_E_T = 36074.9140625; DDC_Km_5htrp = 0.049; kcat_B=0.38; scaling = 1.0; DDC_Km_trp_DASH_L = 10.0Reaction: M_trp_DASH_L_c => M_trypta_c + M_co2_c; M_5htrp_c, M_trp_DASH_L_c, Rate Law: Cytosol*DDC_E_T*kcat_B*scaling*M_trp_DASH_L_c*DDC_Km_5htrp/(DDC_Km_trp_DASH_L*DDC_Km_5htrp+DDC_Km_trp_DASH_L*M_5htrp_c+DDC_Km_5htrp*M_trp_DASH_L_c)

States:

NameDescription
M quln c[quinolinic acid]
M nmtrpta c[N-methyltryptamine]
M o2 c[dioxygen]
M glu DASH L c[L-glutamate(1-)]
M nndmtrpta c[N,N-dimethyltryptamine]
M h2o2 c[hydrogen peroxide]
M nformanth c[N-formylanthranilic acid]
M amp c[AMP]
M f5hoxkyn c[formyl-5-hydroxykynurenamine]
M trp DASH L c[tryptophan]
M co2 c[carbon dioxide]
M thbpt[5,6,7,8-tetrahydrobiopterin]
M dnad c[deamido-NAD(+)]
M nicrnt c[nicotinic acid D-ribonucleotide]
M trypta c[tryptamine]
M nadp c[NADP(+)]
M cmusa c[cis,cis-2-amino-3-(3-oxoprop-1-enyl)but-2-enedioic acid]
M indpyr c[3-(indol-3-yl)pyruvic acid]
M for c[formate]
M coa c[coenzyme A]
M ppi c[diphosphate(4-)]
M 5hoxindact c[(5-hydroxyindol-3-yl)acetaldehyde]
M h c[hydrogen(.)]
M Xanthurenate[xanthurenic acid]
M Nactrypta c[N-acetyl-2-arylethylamine]
M nh4 c[ammonium]
M anth c[anthranilic acid]
M 5hxkyn c[5-hydroxy-L-kynurenine]
M o2s c[sulfur dioxide]
M dhbpt c[(6R)-L-erythro-6,7-dihydrobiopterin; Dihydrobiopterin]
M id3acald c[indol-3-ylacetaldehyde oxime]
M prpp c[5-O-phosphono-alpha-D-ribofuranosyl diphosphate]
M nadph c[NADPH]
TRP ex[tryptophan]
M 5htrp c[5-hydroxytryptophan]
M trna trp c[transfer RNA]
M srtn c[serotonin]
M Lkynr c[L-kynurenine; L-Kynurenine]
M Lkynr ex[L-kynurenine]
M hLkynr c[5-hydroxy-L-kynurenine]
M nmsrtn c[N-methylserotonin]
M h2o c[water]
M Lfmkynr c[N-formylkynurenine]
M accoa c[acetyl-CoA]

Steckmann2012 - Amyloid beta-protein fibrillogenesis (kinetics of secondary structure conversion): BIOMD0000000533v0.0.1

Steckmann2012 - Amyloid beta-protein fibrillogenesis (kinetics of secondary structure conversion)This model is described…

Details

Amyloid fibrils are a common component in many debilitating human neurological diseases such as Alzheimer's (AD), Parkinson's, and Creutzfeldt-Jakob, and in animal diseases such as BSE. The role of fibrillar Αβ proteins in AD has stimulated interest in the kinetics of Αβ fibril formation. Kinetic models that include reaction pathways and rate parameters for the various stages of the process can be helpful towards understanding the dynamics on a molecular level. Based upon experimental data, we have developed a mathematical model for the reaction pathways and determined rate parameters for peptide secondary structural conversion and aggregation during the entire fibrillogenesis process from random coil to mature fibrils, including the molecular species that accelerate the conversions. The model and the rate parameters include different molecular structural stages in the nucleation and polymerization processes and the numerical solutions yield graphs of concentrations of different molecular species versus time that are in close agreement with experimental results. The model also allows for the calculation of the time-dependent increase in aggregate size. The calculated results agree well with experimental results, and allow differences in experimental conditions to be included in the calculations. The specific steps of the model and the rate constants that are determined by fitting to experimental data provide insight on the molecular species involved in the fibril formation process. link: http://identifiers.org/pubmed/22586726

Parameters:

NameDescription
k1 = 0.672; q = 2.0; k2 = 0.678Reaction: alpha = k1*BTX*RCT0-k2*BTX^q*alpha, Rate Law: k1*BTX*RCT0-k2*BTX^q*alpha
epsilon = 0.0; k1 = 0.672; k0 = 0.59Reaction: RCT0 = (-k0)*(epsilon+BM)*RCT0-k1*BTX*RCT0, Rate Law: (-k0)*(epsilon+BM)*RCT0-k1*BTX*RCT0
epsilon = 0.0; k0 = 0.59Reaction: RCT1 = k0*(epsilon+BM)*RCT0, Rate Law: k0*(epsilon+BM)*RCT0
k4 = 0.554Reaction: BM = k4*BTX, Rate Law: k4*BTX
k4 = 0.554; k3 = 0.0392Reaction: BTX = 4*k3*BN4-k4*BTX, Rate Law: 4*k3*BN4-k4*BTX
k3 = 0.0392Reaction: BN2 = 4*k3*BN1-4*k3*BN2, Rate Law: 4*k3*BN1-4*k3*BN2
q = 2.0; k3 = 0.0392; k2 = 0.678Reaction: BN1 = k2*BTX^q*alpha-4*k3*BN1, Rate Law: k2*BTX^q*alpha-4*k3*BN1

States:

NameDescription
RCT1[Amyloid beta A4 protein]
alpha[Amyloid beta A4 protein]
BN1[Amyloid beta A4 protein]
BTX[Amyloid beta A4 protein]
beta[Amyloid beta A4 protein]
BN3[Amyloid beta A4 protein]
RCT0[Amyloid beta A4 protein]
BN2[Amyloid beta A4 protein]
BN4[Amyloid beta A4 protein]
BM[Amyloid beta A4 protein]
RC[Amyloid beta A4 protein]

Stefan2008 - calmodulin allostery: BIOMD0000000183v0.0.1

Stefan2008 - calmodulin allostery An allosteric model for calmodulin activation, in which binding to calcium facilitate…

Details

Calmodulin plays a vital role in mediating bidirectional synaptic plasticity by activating either calcium/calmodulin-dependent protein kinase II (CaMKII) or protein phosphatase 2B (PP2B) at different calcium concentrations. We propose an allosteric model for calmodulin activation, in which binding to calcium facilitates the transition between a low-affinity [tense (T)] and a high-affinity [relaxed (R)] state. The four calcium-binding sites are assumed to be nonidentical. The model is consistent with previously reported experimental data for calcium binding to calmodulin. It also accounts for known properties of calmodulin that have been difficult to model so far, including the activity of nonsaturated forms of calmodulin (we predict the existence of open conformations in the absence of calcium), an increase in calcium affinity once calmodulin is bound to a target, and the differential activation of CaMKII and PP2B depending on calcium concentration. link: http://identifiers.org/pubmed/18669651

Parameters:

NameDescription
parameter_5 = 2101.0101010101Reaction: species_29 => species_26 + species_1, Rate Law: compartment_0*parameter_5*species_29
parameter_18 = 0.0013Reaction: species_54 => species_4 + species_50, Rate Law: compartment_0*parameter_18*species_54
parameter_3 = 17.4Reaction: species_15 => species_10 + species_1, Rate Law: compartment_0*parameter_3*species_15
parameter_10 = 48.3792936623125; parameter_9 = 1000000.0Reaction: species_2 => species_18, Rate Law: compartment_0*species_2*parameter_9*parameter_10^(1/2)
parameter_4 = 0.0145Reaction: species_13 => species_6 + species_1, Rate Law: compartment_0*parameter_4*species_13
parameter_1 = 8.32Reaction: species_14 => species_11 + species_1, Rate Law: compartment_0*parameter_1*species_14
parameter_15 = 3200000.0Reaction: species_5 + species_33 => species_38, Rate Law: compartment_0*parameter_15*species_5*species_33
parameter_7 = 4393.93939393939Reaction: species_30 => species_24 + species_1, Rate Law: compartment_0*parameter_7*species_30
parameter_16 = 0.343Reaction: species_37 => species_4 + species_33, Rate Law: compartment_0*parameter_16*species_37
parameter_6 = 4.19191919191919Reaction: species_31 => species_27 + species_1, Rate Law: compartment_0*parameter_6*species_31
parameter_0 = 1000000.0Reaction: species_13 + species_1 => species_16, Rate Law: compartment_0*parameter_0*species_13*species_1
parameter_10 = 48.3792936623125Reaction: species_17 => species_0, Rate Law: compartment_0*parameter_10*species_17
parameter_8 = 3.66161616161616Reaction: species_29 => species_22 + species_1, Rate Law: compartment_0*parameter_8*species_29
parameter_2 = 0.0166Reaction: species_13 => species_8 + species_1, Rate Law: compartment_0*parameter_2*species_13

States:

NameDescription
species 19[calcium(2+); Calmodulin]
species 33[calmodulin-dependent protein kinase activity; Calcium/calmodulin-dependent protein kinase type II subunit alpha; IPR015742; calcium- and calmodulin-dependent protein kinase complex]
species 1[calcium(2+); Calcium cation]
species 18[calcium(2+); Calmodulin]
species 4[calcium(2+); Calmodulin]
species 17[Calmodulin-3Calmodulin-1Calmodulin-2; Calmodulin]
species 24[calcium(2+); Calmodulin]
species 5[calcium(2+); Calmodulin]
species 26[calcium(2+); Calmodulin]
species 28[calcium(2+); Calmodulin]

Stephanou2019 - pH as a potential therapeutic target to improve temozolomide antitumor efficacy: MODEL1909300003v0.0.1

Abstract: Despite intensive treatments including temozolomide (TMZ) administration, glioblastoma patient prognosis rema…

Details

Despite intensive treatments including temozolomide (TMZ) administration, glioblastoma patient prognosis remains dismal and innovative therapeutic strategies are urgently needed. A systems pharmacology approach was undertaken to investigate TMZ pharmacokinetics-pharmacodynamics (PK-PD) incorporating the effect of local pH, tumor spatial configuration and micro-environment. A hybrid mathematical framework was designed coupling ordinary differential equations describing the intracellular reactions, with a spatial cellular automaton to individualize the cells. A differential drug impact on tumor and healthy cells at constant extracellular pH was computationally demonstrated as TMZ-induced DNA damage was larger in tumor cells as compared to normal cells due to less acidic intracellular pH in cancer cells. Optimality of TMZ efficacy defined as maximum difference between damage in tumor and healthy cells was reached for extracellular pH between 6.8 and 7.5. Next, TMZ PK-PD in a solid tumor was demonstrated to highly depend on its spatial configuration as spread cancer cells or fragmented tumors presented higher TMZ-induced damage as compared to compact tumor spheroid. Simulations highlighted that smaller tumors were less acidic than bigger ones allowing for faster TMZ activation and their closer distance to blood capillaries allowed for better drug penetration. For model parameters corresponding to U87 glioma cells, inter-cell variability in TMZ uptake play no role regarding the mean drug-induced damage in the whole cell population whereas this quantity was increased by inter-cell variability in TMZ efflux which was thus a disadvantage in terms of drug resistance. Overall, this study revealed pH as a new potential target to significantly improve TMZ antitumor efficacy. link: http://identifiers.org/pubmed/30705757

Stewart2009_ActionPotential_PurkinjeFibreCells: MODEL1006230012v0.0.1

This a model from the article: Mathematical models of the electrical action potential of Purkinje fibre cells. Phili…

Details

Early development of ionic models for cardiac myocytes, from the pioneering modification of the Hodgkin-Huxley giant squid axon model by Noble to the iconic DiFrancesco-Noble model integrating voltage-gated ionic currents, ion pumps and exchangers, Ca(2+) sequestration and Ca(2+)-induced Ca(2+) release, provided a general description for a mammalian Purkinje fibre (PF) and the framework for modern cardiac models. In the past two decades, development has focused on tissue-specific models with an emphasis on the sino-atrial (SA) node, atria and ventricles, while the PFs have largely been neglected. However, achieving the ultimate goal of creating a virtual human heart will require detailed models of all distinctive regions of the cardiac conduction system, including the PFs, which play an important role in conducting cardiac excitation and ensuring the synchronized timing and sequencing of ventricular contraction. In this paper, we present details of our newly developed model for the human PF cell including validation against experimental data. Ionic mechanisms underlying the heterogeneity between the PF and ventricular action potentials in humans and other species are analysed. The newly developed PF cell model adds a new member to the family of human cardiac cell models developed previously for the SA node, atrial and ventricular cells, which can be incorporated into an anatomical model of the human heart with details of its electrophysiological heterogeneity and anatomical complexity. link: http://identifiers.org/pubmed/19414454

Stone1996 - activation of soluble guanylate cyclase by nitric oxide: BIOMD0000000198v0.0.1

Stone1996 - activation of soluble guanylate cyclase by nitric oxideThis features the two step binding of NO to soluble G…

Details

The soluble form of guanylate cyclase (sGC) is the only definitive receptor for the signaling agent nitric oxide (.NO). The enzyme is a heterodimer of homologous subunits in which each subunit binds 1 equiv of 5-coordinate high-spin heme. .NO increases the Vmax of sGC up to 400-fold and has previously been shown to bind to the heme to form a 5-coordinate complex. Using stopped-flow spectrophotometry, it is demonstrated that the binding of .NO to the heme of sGC is a complex process. .NO first binds to the heme to form a 6-coordinate nitrosyl complex, which then converts to a 5-coordinate nitrosyl complex through one of two ways. For 28 +/- 4% of the heme, the 6-coordinate nitrosyl complex rapidly (approximately 20 s-1) converts to the 5-coordinate complex. For the remaining 72 +/- 4% of the heme, the conversion of the 6-coordinate nitrosyl complex to a 5-coordinate nitrosyl complex is slow (0.1-1.0 s-1) and is dependent upon the interaction of .NO with an unidentified non-heme site on the protein. The heme (200 nM) was completely converted to the 5-coordinate state with as little as 500 nM .NO, and the equilibrium dissociation constant of .NO for activating the enzyme was determined to be < or = 250 nM. Gel-filtration analysis indicates that the binding of .NO to the heme has no effect on the native molecular mass of the protein. Correlation of electronic absorption spectra with activity measurements indicates that the 5-coordinate nitrosyl form of the enzyme is activated relative to the resting 5-coordinate ferrous form of the enzyme. link: http://identifiers.org/pubmed/8573563

Parameters:

NameDescription
k6 = 700.0 l*μmol^(-1)*s^(-1); k7 = 800.0 s^(-1)Reaction: NO + sGCslow => NO_sGCslow, Rate Law: cytosol*(k6*NO*sGCslow-k7*NO_sGCslow)
k2 = 800.0 s^(-1); k1 = 700.0 l*μmol^(-1)*s^(-1)Reaction: NO + sGCfast => NO_sGCfast, Rate Law: cytosol*(k1*NO*sGCfast-k2*NO_sGCfast)
k8 = 850.0 s^(-1)Reaction: NO_sGCslow => NO_sGCslow_6coord, Rate Law: k8*cytosol*NO_sGCslow
k11 = 1.6 s^(-1); k12 = 0.02 s^(-1)Reaction: NO_sGCslow_6coord_NO_int => NO_sGCslow_5coord, Rate Law: cytosol*(k11*NO_sGCslow_6coord_NO_int-k12*NO_sGCslow_5coord)
k3 = 850.0 s^(-1)Reaction: NO_sGCfast => NO_sGCfast_6coord, Rate Law: k3*cytosol*NO_sGCfast
k10 = 25.0 s^(-1); k9 = 5.0 l*μmol^(-1)*s^(-1)Reaction: NO + NO_sGCslow_6coord => NO_sGCslow_6coord_NO_int, Rate Law: cytosol*(k9*NO*NO_sGCslow_6coord-k10*NO_sGCslow_6coord_NO_int)
k4 = 20.0 s^(-1); k5 = 0.2 s^(-1)Reaction: NO_sGCfast_6coord => NO_sGCfast_5coord, Rate Law: cytosol*(k4*NO_sGCfast_6coord-k5*NO_sGCfast_5coord)

States:

NameDescription
NO sGCslow[nitric oxide; Guanylate cyclase soluble subunit alpha-1; Guanylate cyclase soluble subunit beta-1]
NO sGCfast 6coord[nitric oxide; Guanylate cyclase soluble subunit alpha-1; Guanylate cyclase soluble subunit beta-1]
NO[nitric oxide; Nitric oxide]
sGC inact tot[nitric oxide; Guanylate cyclase soluble subunit beta-1; Guanylate cyclase soluble subunit alpha-1]
sGCfast[Guanylate cyclase soluble subunit alpha-1; Guanylate cyclase soluble subunit beta-1]
NO sGCslow 5coord[nitric oxide; Guanylate cyclase soluble subunit alpha-1; Guanylate cyclase soluble subunit beta-1]
sGCslow[Guanylate cyclase soluble subunit alpha-1; Guanylate cyclase soluble subunit beta-1]
NO sGCslow 6coord NO int[nitric oxide; Guanylate cyclase soluble subunit alpha-1; Guanylate cyclase soluble subunit beta-1]
NO sGC 5coord tot[nitric oxide; Guanylate cyclase soluble subunit beta-1; Guanylate cyclase soluble subunit alpha-1]
NO sGCfast[nitric oxide; Guanylate cyclase soluble subunit alpha-1; Guanylate cyclase soluble subunit beta-1]
NO sGCfast 5coord[nitric oxide; Guanylate cyclase soluble subunit alpha-1; Guanylate cyclase soluble subunit beta-1]
NO sGCslow 6coord[nitric oxide; Guanylate cyclase soluble subunit alpha-1; Guanylate cyclase soluble subunit beta-1]

Stortelder1997 - Thrombin Generation Amidolytic Activity: BIOMD0000000358v0.0.1

Stortelder1997 - Thrombin Generation Amidolytic ActivityMathematical modelling of a part of the blood coagulation mechan…

Details

This paper describes the mathematical modelling of a part of the blood coagulation mechanism. The model includes the activation of factor X by a purified enzyme from Russel's Viper Venom (RVV), factor V and prothrombin, and also comprises the inactivation of the products formed. In this study we assume that in principle the mechanism of the process is known. However, the exact structure of the mechanism is unknown, and the process still can be described by different mathematical models. These models are put to test by measuring their capacity to explain the course of thrombin generation as observed in plasma after recalcification in presence of RVV. The mechanism studied is mathematically modelled as a system of differential-algebraic equations (DAEs). Each candidate model contains some freedom, which is expressed in the model equations by the presence of unknown parameters. For example, reaction constants or initial concentrations are unknown. The goal of parameter estimation is to determine these unknown parameters in such a way that the theoretical (i.e., computed) results fit the experimental data within measurement accuracy and to judge which modifications of the chemical reaction scheme allow the best fit. We present results on model discrimination and estimation of reaction constants, which are hard to obtain in another way. link: http://www.narcis.nl/publication/RecordID/oai:cwi.nl:4725

Parameters:

NameDescription
k_PL = 801.4; k_PT = 122.9Reaction: Va + Xa + PL => PT, Rate Law: compartment_1*(k_PT*Va*Xa*PL-k_PL*PT)
kcat_2 = 12.4; km_2 = 0.06148Reaction: II => IIa; Xa, Rate Law: compartment_1*kcat_2*Xa*II/(km_2+II)
ki_Xa = 4.531Reaction: Xa => Xa_ATIII, Rate Law: compartment_1*ki_Xa*Xa
km_II = 62.25; kcat_II = 43.87Reaction: II => IIa; PT, Rate Law: compartment_1*kcat_II*PT*II/(km_II+II)
ki_IIaATIII = 0.7859Reaction: IIa => IIa_ATIII, Rate Law: compartment_1*ki_IIaATIII*IIa
kcat_X = 239.1; km_X = 23.65Reaction: X => Xa; RVV, Rate Law: compartment_1*kcat_X*RVV*X/(km_X+X)
km_V = 149.7; kcat_V = 7.844Reaction: V => Va; IIa, Rate Law: compartment_1*kcat_V*IIa*V/(km_V+V)
ki_IIaAlpha2M = 0.1762Reaction: IIa => IIa_alpha2M, Rate Law: compartment_1*ki_IIaAlpha2M*IIa

States:

NameDescription
IIa ATIII[Prothrombin; Antithrombin-III]
Xa ATIII[Antithrombin-III; Coagulation factor X]
X[Coagulation factor X]
PTPT
V[Coagulation factor V]
Xa[Coagulation factor X]
Va[Coagulation factor V]
IIa[Prothrombin]
IIa alpha2M[Alpha-2-macroglobulin; Prothrombin]
PLPL
II[Prothrombin]

Strasen2018 - TGFb SMAD Signalling - Degradation of 25pM ligand (TGFb): BIOMD0000000990v0.0.1

MOdel simulates 25pM ligand degradation kinetics as shown in Figure 4D

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling - Dose dependent dynamics upon TGFb stimulation: BIOMD0000000989v0.0.1

This model simulates TGFb dose dependent kinetics of The SMADs. TGFb ligand dose applied are 1pM, 2.5pM, 5pM, 25pM, and…

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling - DRB treatment: BIOMD0000000997v0.0.1

Simulates figure 4H. DRB = 1 simulates the wild type condition and DRB = 0.2 simulates the DRB treated condition.

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling - Restimulation with 5pM TGFb at 3hr: BIOMD0000000994v0.0.1

Restimulation with 5pM TGFβ at 3hr - Figure 4E

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling - Restimulation with 5pM TGFb at 8hr: BIOMD0000000995v0.0.1

Restimulation with 5pM TGFβ at 8 hrs, shown in Figure 4F

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling - Restimulation with 100pM TGFb at 6hr: BIOMD0000000996v0.0.1

Restimulation with 100pM TGFβ at 6hr- figure 4G

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling Class 1: BIOMD0000000998v0.0.1

Model depicting response Class 1 defined by a minimal response to stimulation which is considered as non‐responders.

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling Class 2: BIOMD0000000999v0.0.1

Model depicting class 2 signaling class observed upon stimulation with 100 pM TGFβ1.

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling Class 3: BIOMD0000001000v0.0.1

Simulates signaling class 3

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling Class 4: BIOMD0000001001v0.0.1

Simulating Class 4 signalling

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling Class 5: BIOMD0000001002v0.0.1

simulating class 5 signalling

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Strasen2018 - TGFb SMAD Signalling Class 6: BIOMD0000001003v0.0.1

simulating class 6 signalling

Details

The cytokine TGFb provides important information during embryonic development, adult tissue homeostasis, and regeneration. Alterations in the cellular response to TGFb are involved in severe human diseases. To understand how cells encode the extracellular input and transmit its information to elicit appropriate responses, we acquired quantitative time-resolved measurements of pathway activation at the single-cell level. We established dynamic time warping to quantitatively compare signaling dynamics of thousands of individual cells and described heterogeneous single-cell responses by mathematical modeling. Our combined experimental and theoretical study revealed that the response to a given dose of TGFb is determined cell specifically by the levels of defined signaling proteins. This heterogeneity in signaling protein expression leads to decomposition of cells into classes with qualitatively distinct signaling dynamics and phenotypic outcome. Negative feedback regulators promote heterogeneous signaling, as a SMAD7 knock-out specifically affected the signal duration in a subpopulation of cells. Taken together, we propose a quantitative framework that allows predicting and testing sources of cellular signaling heterogeneity. link: http://identifiers.org/pubmed/29371237

Sturis1991_InsulinGlucoseModel_UltradianOscillation: BIOMD0000000382v0.0.1

This a model from the article: Computer model for mechanisms underlying ultradian oscillations of insulin and gluco…

Details

Oscillations in human insulin secretion have been observed in two distinct period ranges, 10-15 min (i.e. rapid) and 100-150 min (i.e., ultradian). The cause of the ultradian oscillations remains to be elucidated. To determine whether the oscillations could result from the feedback loops between insulin and glucose, a parsimonious mathematical model including the major mechanisms involved in glucose regulation was developed. This model comprises two major negative feedback loops describing the effects of insulin on glucose utilization and glucose production, respectively, and both loops include the stimulatory effect of glucose on insulin secretion. Model formulations and parameters are representative of results from published clinical investigations. The occurrence of sustained insulin and glucose oscillations was found to be dependent on two essential features: 1) a time delay of 30-45 min for the effect of insulin on glucose production and 2) a sluggish effect of insulin on glucose utilization, because insulin acts from a compartment remote from plasma. When these characteristics were incorporated in the model, numerical simulations mimicked all experimental findings so far observed for these ultradian oscillations, including 1) self-sustained oscillations during constant glucose infusion at various rates; 2) damped oscillations after meal or oral glucose ingestion; 3) increased amplitude of oscillation after increased stimulation of insulin secretion, without change in frequency; and 4) slight advance of the glucose oscillation compared with the insulin oscillation.(ABSTRACT TRUNCATED AT 250 WORDS) link: http://identifiers.org/pubmed/2035636

Parameters:

NameDescription
E = 0.21; t2 = 100.0; v2 = 11.0; v1 = 3.0Reaction: y = E*(x/v1-y/v2)-y/t2, Rate Law: E*(x/v1-y/v2)-y/t2
E = 0.21; v2 = 11.0; f1 = NaN; t1 = 6.0; v1 = 3.0Reaction: x = (f1-E*(x/v1-y/v2))-x/t1, Rate Law: (f1-E*(x/v1-y/v2))-x/t1
t3 = 36.0Reaction: h1 = 3*(x-h1)/t3, Rate Law: 3*(x-h1)/t3
f2 = NaN; f4 = NaN; I = 216.0; f3 = NaN; f5 = NaNReaction: z = ((f5+I)-f2)-f3*f4, Rate Law: ((f5+I)-f2)-f3*f4

States:

NameDescription
h1h1
h3h3
h2h2
x[Insulin]
z[glucose]
y[Insulin]

Sturrock2015 - glioma growth: BIOMD0000000801v0.0.1

The paper describes a model of glioma. Created by COPASI 4.26 (Build 213) This model is described in the article:…

Details

Due to their location, the malignant gliomas of the brain in humans are very difficult to treat in advanced stages. Blood-based biomarkers for glioma are needed for more accurate evaluation of treatment response as well as early diagnosis. However, biomarker research in primary brain tumors is challenging given their relative rarity and genetic diversity. It is further complicated by variations in the permeability of the blood brain barrier that affects the amount of marker released into the bloodstream. Inspired by recent temporal data indicating a possible decrease in serum glucose levels in patients with gliomas yet to be diagnosed, we present an ordinary differential equation model to capture early stage glioma growth. The model contains glioma-glucose-immune interactions and poses a potential mechanism by which this glucose drop can be explained. We present numerical simulations, parameter sensitivity analysis, linear stability analysis and a numerical experiment whereby we show how a dormant glioma can become malignant. link: http://identifiers.org/pubmed/26073722

Parameters:

NameDescription
as = 0.7 1/d; v = 0.7 1Reaction: => I; B, Rate Law: tme*as*(v+I)*B
at = 1.575 1/d; kt = 2.0 1Reaction: => T; B, Rate Law: tme*at*B*T*(1-T/kt)
F = 8.0E-4 1/dReaction: => S, Rate Law: tme*F
dti = 0.072 1/dReaction: T => ; I, Rate Law: tme*dti*I*T
as = 0.7 1/d; do = 0.01 1/d; v = 0.7 1Reaction: B => ; I, Rate Law: tme*(do+as*(v+I))*B
ao = 20.0 1/dReaction: B => S, Rate Law: tme*ao*B
do = 0.01 1/dReaction: S =>, Rate Law: tme*do*S
dto = 1.0 1/dReaction: B => ; T, Rate Law: tme*dto*T*B
di = 0.01 1/dReaction: I =>, Rate Law: tme*di*I
dtt = 0.72 1/dReaction: I => ; T, Rate Law: tme*dtt*T*I
dt = 1.0E-4 1/dReaction: T =>, Rate Law: tme*dt*T
ati = 3.0E-4 1/dReaction: => I; T, Rate Law: tme*ati*T*I

States:

NameDescription
B[glucose]
S[glucose]
I[leukocyte]
T[glioma cell]

Subramanian2017 - Metabolic adaptations of Leishmania parasite, Genome-scale constraint-based model of Leishmania infantum: MODEL2010130001v0.0.1

The uploaded model is linked to the Scientific Reports article: Subramanian, A., Sarkar, R.R. Revealing the mystery of m…

Details

Human macrophage phagolysosome and sandfly midgut provide antagonistic ecological niches for Leishmania parasites to survive and proliferate. Parasites optimize their metabolism to utilize the available inadequate resources by adapting to those environments. Lately, a number of metabolomics studies have revived the interest to understand metabolic strategies utilized by the Leishmania parasite for optimal survival within its hosts. For the first time, we propose a reconstructed genome-scale metabolic model for Leishmania infantum JPCM5, the analyses of which not only captures observations reported by metabolomics studies in other Leishmania species but also divulges novel features of the L. infantum metabolome. Our results indicate that Leishmania metabolism is organized in such a way that the parasite can select appropriate alternatives to compensate for limited external substrates. A dynamic non-essential amino acid motif exists within the network that promotes a restricted redistribution of resources to yield required essential metabolites. Further, subcellular compartments regulate this metabolic re-routing by reinforcing the physiological coupling of specific reactions. This unique metabolic organization is robust against accidental errors and provides a wide array of choices for the parasite to achieve optimal survival. link: http://identifiers.org/doi/10.1038/s41598-017-10743-x

Subrmanian2015 - Energy metabolism of Leishmania infantum, constrain-based metabolic model: MODEL2010130002v0.0.1

The uploaded model is linked to the PLoS ONE article: Subramanian A, Jhawar J, Sarkar RR (2015) Dissecting Leishmania in…

Details

Leishmania infantum, causative agent of visceral leishmaniasis in humans, illustrates a complex lifecycle pertaining to two extreme environments, namely, the gut of the sandfly vector and human macrophages. Leishmania is capable of dynamically adapting and tactically switching between these critically hostile situations. The possible metabolic routes ventured by the parasite to achieve this exceptional adaptation to its varying environments are still poorly understood. In this study, we present an extensively reconstructed energy metabolism network of Leishmania infantum as an attempt to identify certain strategic metabolic routes preferred by the parasite to optimize its survival in such dynamic environments. The reconstructed network consists of 142 genes encoding for enzymes performing 237 reactions distributed across five distinct model compartments. We annotated the subcellular locations of different enzymes and their reactions on the basis of strong literature evidence and sequence-based detection of cellular localization signal within a protein sequence. To explore the diverse features of parasite metabolism the metabolic network was implemented and analyzed as a constraint-based model. Using a systems-based approach, we also put forth an extensive set of lethal reaction knockouts; some of which were validated using published data on Leishmania species. Performing a robustness analysis, the model was rigorously validated and tested for the secretion of overflow metabolites specific to Leishmania under varying extracellular oxygen uptake rate. Further, the fate of important non-essential amino acids in L. infantum metabolism was investigated. Stage-specific scenarios of L. infantum energy metabolism were incorporated in the model and key metabolic differences were outlined. Analysis of the model revealed the essentiality of glucose uptake, succinate fermentation, glutamate biosynthesis and an active TCA cycle as driving forces for parasite energy metabolism and its optimal growth. Finally, through our in silico knockout analysis, we could identify possible therapeutic targets that provide experimentally testable hypotheses. link: http://identifiers.org/doi/10.1371/journal.pone.0137976

Suh2004_KCNQ_Regulation: BIOMD0000000081v0.0.1

The model reproduces FIG 11A and FIG 11B of the paper. However, please note that FIG 11B is a plot of normalised amounts…

Details

Receptor-mediated modulation of KCNQ channels regulates neuronal excitability. This study concerns the kinetics and mechanism of M1 muscarinic receptor-mediated regulation of the cloned neuronal M channel, KCNQ2/KCNQ3 (Kv7.2/Kv7.3). Receptors, channels, various mutated G-protein subunits, and an optical probe for phosphatidylinositol 4,5-bisphosphate (PIP2) were coexpressed by transfection in tsA-201 cells, and the cells were studied by whole-cell patch clamp and by confocal microscopy. Constitutively active forms of Galphaq and Galpha11, but not Galpha13, caused a loss of the plasma membrane PIP2 and a total tonic inhibition of the KCNQ current. There were no further changes upon addition of the muscarinic agonist oxotremorine-M (oxo-M). Expression of the regulator of G-protein signaling, RGS2, blocked PIP2 hydrolysis and current suppression by muscarinic stimulation, confirming that the Gq family of G-proteins is necessary. Dialysis with the competitive inhibitor GDPbetaS (1 mM) lengthened the time constant of inhibition sixfold, decreased the suppression of current, and decreased agonist sensitivity. Removal of intracellular Mg2+ slowed both the development and the recovery from muscarinic suppression. When combined with GDPbetaS, low intracellular Mg2+ nearly eliminated muscarinic inhibition. With nonhydrolyzable GTP analogs, current suppression developed spontaneously and muscarinic inhibition was enhanced. Such spontaneous suppression was antagonized by GDPbetaS or GTP or by expression of RGS2. These observations were successfully described by a kinetic model representing biochemical steps of the signaling cascade using published rate constants where available. The model supports the following sequence of events for this Gq-coupled signaling: A classical G-protein cycle, including competition for nucleotide-free G-protein by all nucleotide forms and an activation step requiring Mg2+, followed by G-protein-stimulated phospholipase C and hydrolysis of PIP2, and finally PIP2 dissociation from binding sites for inositol lipid on the channels so that KCNQ current was suppressed. Further experiments will be needed to refine some untested assumptions. link: http://identifiers.org/pubmed/15173220

Parameters:

NameDescription
kPIP2on=2.5E-4; kPIP2off=0.25Reaction: KCNQsites_M + PIP2_M => PIP2xKCNQ_M, Rate Law: M*(kPIP2on*KCNQsites_M*PIP2_M+(-kPIP2off*PIP2xKCNQ_M))
kGTPon=0.45; kGTPoff=0.08Reaction: G_M => GGTP_M; GTP_C, Rate Law: (kGTPon*G_M*GTP_C+(-kGTPoff*GGTP_M))*M
kAlF4off=0.005; kAlF4on=7.0E-6Reaction: AlF4_C + GGDP_M => GGDPAlF4_M, Rate Law: (kAlF4on*AlF4_C*GGDP_M+(-kAlF4off*GGDPAlF4_M))*M
MgSat20 = 0.990566037735849 1; kPI4Kinase=1.0E-4; ATPSat1000 = 0.750031246094238 1Reaction: ATP_C + PI_M => PIP_M, Rate Law: (0.2+0.8*MgSat20)*kPI4Kinase*PI_M*ATPSat1000*M
MgSat20 = 0.990566037735849 1; kPIP5Kinase=0.06; ATPSat300 = 0.909104681108923 1Reaction: ATP_C + PIP_M => PIP2_M, Rate Law: (0.2+0.8*MgSat20)*kPIP5Kinase*PIP_M*ATPSat300
kIP3ase=0.3; NA_micro = 6.022E17 1/MAvogadroReaction: ip3_C =>, Rate Law: Cytoplasm*ip3_C*kIP3ase*NA_micro
kMg2onGTP=0.003Reaction: GGTP_M => GGTPMg_M; Mg2_C, Rate Law: kMg2onGTP*GGTP_M*Mg2_C*M
kPLC=4.8; PLCspont=7.5E-4; fGactive = 5.0E-4 1Reaction: PIP2_M => ip3_C, Rate Law: kPLC*(fGactive+PLCspont)*PIP2_M*M
MgSat20 = 0.990566037735849 1; kPIP2Pase=0.005Reaction: PIP2_M => PIP_M, Rate Law: M*MgSat20*kPIP2Pase*PIP2_M
MgSat10 = 0.996208530805687 1; kGGTPase=1.8Reaction: GGTPMg_M => GGDP_M, Rate Law: M*kGGTPase*MgSat10*GGTPMg_M
TonicAct=0.002; kGDPon=0.003; kGDPoff=0.5; OxoSat = 1.24999843750195E-6 1Reaction: GGDP_M => GDP_C + G_M; oxoM_EX, Rate Law: (kGDPoff*GGDP_M*(OxoSat+TonicAct)+(-kGDPon*GDP_C*G_M))*M
kGTPgSon=0.006; kGTPgSoff=0.005Reaction: GTPgS_C + G_M => GGTPgS_M, Rate Law: (kGTPgSon*G_M*GTPgS_C+(-kGTPgSoff*GGTPgS_M))*M
kGDPbSon=0.28; kGGDPbSoff=0.1; OxoSat = 1.24999843750195E-6 1Reaction: G_M + GDPbS_C => GGDPbS_M, Rate Law: (kGDPbSon*G_M*GDPbS_C+(-kGGDPbSoff*(1+20*OxoSat)*GGDPbS_M))*M
kMg2onGTPgS=0.002Reaction: GGTPgS_M => GGTPgSMg_M; Mg2_C, Rate Law: kMg2onGTPgS*GGTPgS_M*Mg2_C*M
kMgonAlF4=0.002Reaction: GGDPAlF4_M => GGDPAlF4Mg_M; Mg2_C, Rate Law: kMgonAlF4*GGDPAlF4_M*Mg2_C*M

States:

NameDescription
GGDP M[heterotrimeric G-protein complex; GDP; GDP]
ATP C[ATP; ATP]
PI M[1-phosphatidyl-1D-myo-inositol; 1-Phosphatidyl-D-myo-inositol]
GGDPAlF4 M[GDP; heterotrimeric G-protein complex; GDP; tetrafluoroaluminate(1-)]
GGTPgS M[heterotrimeric G-protein complex; guanosine 5'-[gamma-thio]triphosphate; CHEBI_5235; GTP-gamma-S]
ip3 C[1D-myo-inositol 1,4,5-trisphosphate; D-myo-Inositol 1,4,5-trisphosphate]
PIP M[1-phosphatidyl-1D-myo-inositol 4-phosphate; 1-Phosphatidyl-1D-myo-inositol 4-phosphate]
AlF4 C[tetrafluoroaluminate(1-)]
GGTPgSMg M[Magnesium cation; CHEBI_5235; magnesium atom; GTP-gamma-S; heterotrimeric G-protein complex; magnesium(2+)]
GGDPAlF4Mg M[Guanine nucleotide-binding protein subunit alpha-11; Magnesium cation; GDP; tetrafluoroaluminate(1-); magnesium atom; GDP]
G M[heterotrimeric G-protein complex]
GGDPbS M[GDP-beta-S; heterotrimeric G-protein complex]
GDPbS C[GDP-beta-S]
PIP2 M[1-phosphatidyl-1D-myo-inositol 4,5-bisphosphate; 1-Phosphatidyl-D-myo-inositol 4,5-bisphosphate]
GDP C[GDP; GDP]
GTP C[GTP; GTP]
GGTP M[heterotrimeric G-protein complex; GTP; GTP]
GTPgS C[GTP-gamma-S; CHEBI_5235]
GGTPMg M[GTP; Magnesium cation; heterotrimeric G-protein complex; magnesium(2+); GTP; magnesium atom]
PIP2xKCNQ M[Potassium voltage-gated channel subfamily KQT member 3; 1-phosphatidyl-1D-myo-inositol 4,5-bisphosphate; 1-Phosphatidyl-D-myo-inositol 4,5-bisphosphate; Potassium voltage-gated channel subfamily KQT member 2]
KCNQsites M[Potassium voltage-gated channel subfamily KQT member 3; Potassium voltage-gated channel subfamily KQT member 2]

Sumana2018 - Mathematical modeling of cancer-immune system, considering the role of antibodies.: BIOMD0000000885v0.0.1

Mathematical modeling of cancer-immune system, considering the role of antibodies. Ghosh S1, Banerjee S2. Author informa…

Details

A mathematical model for the quantitative analysis of cancer-immune interaction, considering the role of antibodies has been proposed in this paper. The model is based on the clinical evidence, which states that antibodies can directly kill cancerous cells (Ivano et al. in J Clin Investig 119(8):2143-2159, 2009). The existence of transcritical bifurcation, which has been proved using Sotomayor theorem, provides strong biological implications. Through numerical simulations, it has been illustrated that under certain therapy (like monoclonal antibody therapy), which is capable of altering the parameters of the system, cancer-free state can be obtained. link: http://identifiers.org/pubmed/29572780

Parameters:

NameDescription
r = 0.431; k_2 = 9.8E8Reaction: => T, Rate Law: compartment*r*T*(1-T/k_2)
u = 0.1; b = 0.01Reaction: B =>, Rate Law: compartment*b*(1-u)*B
mu_1 = 0.01Reaction: P =>, Rate Law: compartment*mu_1*P
mu_2 = 6.884Reaction: A =>, Rate Law: compartment*mu_2*A
a = 0.1; k_1 = 1000000.0; u = 0.1Reaction: => B, Rate Law: compartment*a*u*B*(1-B/k_1)
r_1 = 100.0; r_2 = 1000.0Reaction: => A; B, P, Rate Law: compartment*(r_1*B+r_2*P)
beta_1 = 3.0218E7Reaction: T => ; A, Rate Law: compartment*beta_1*A*T

States:

NameDescription
BB
AA
T[6754]
P[Plasma]

Sun2009 - Genome-scale metabolic network of Geobacter metallireducens (iJS747): MODEL1507180002v0.0.1

Sun2009 - Genome-scale metabolic network of Geobacter metallireducens (iJS747)This model is described in the article: […

Details

BACKGROUND: Geobacter metallireducens was the first organism that can be grown in pure culture to completely oxidize organic compounds with Fe(III) oxide serving as electron acceptor. Geobacter species, including G. sulfurreducens and G. metallireducens, are used for bioremediation and electricity generation from waste organic matter and renewable biomass. The constraint-based modeling approach enables the development of genome-scale in silico models that can predict the behavior of complex biological systems and their responses to the environments. Such a modeling approach was applied to provide physiological and ecological insights on the metabolism of G. metallireducens. RESULTS: The genome-scale metabolic model of G. metallireducens was constructed to include 747 genes and 697 reactions. Compared to the G. sulfurreducens model, the G. metallireducens metabolic model contains 118 unique reactions that reflect many of G. metallireducens' specific metabolic capabilities. Detailed examination of the G. metallireducens model suggests that its central metabolism contains several energy-inefficient reactions that are not present in the G. sulfurreducens model. Experimental biomass yield of G. metallireducens growing on pyruvate was lower than the predicted optimal biomass yield. Microarray data of G. metallireducens growing with benzoate and acetate indicated that genes encoding these energy-inefficient reactions were up-regulated by benzoate. These results suggested that the energy-inefficient reactions were likely turned off during G. metallireducens growth with acetate for optimal biomass yield, but were up-regulated during growth with complex electron donors such as benzoate for rapid energy generation. Furthermore, several computational modeling approaches were applied to accelerate G. metallireducens research. For example, growth of G. metallireducens with different electron donors and electron acceptors were studied using the genome-scale metabolic model, which provided a fast and cost-effective way to understand the metabolism of G. metallireducens. CONCLUSION: We have developed a genome-scale metabolic model for G. metallireducens that features both metabolic similarities and differences to the published model for its close relative, G. sulfurreducens. Together these metabolic models provide an important resource for improving strategies on bioremediation and bioenergy generation. link: http://identifiers.org/pubmed/19175927

Sun2018 - Instantaneous mutation rate in cancer initiation and progression: BIOMD0000000915v0.0.1

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

Details

BACKGROUND:Cancer is one of the leading causes for the morbidity and mortality worldwide. Although substantial studies have been conducted theoretically and experimentally in recent years, it is still a challenge to explore the mechanisms of cancer initiation and progression. The investigation for these problems is very important for the diagnosis of cancer diseases and development of treatment schemes. RESULTS:To accurately describe the process of cancer initiation, we propose a new concept of gene initial mutation rate based on our recently designed mathematical model using the non-constant mutation rate. Unlike the widely-used average gene mutation rate that depends on the number of mutations, the gene initial mutation rate can be used to describe the initiation process of a single patient. In addition, we propose the instantaneous tumour doubling time that is a continuous function of time based on the non-constant mutation rate. Our proposed concepts are supported by the clinic data of seven patients with advanced pancreatic cancer. The regression results suggest that, compared with the average mutation rate, the estimated initial mutation rate has a larger value of correlation coefficient with the patient survival time. We also provide the estimated tumour size of these seven patients over time. CONCLUSIONS:The proposed concepts can be used to describe the cancer initiation and progression for different patients more accurately. Since a quantitative understanding of cancer progression is important for clinical treatment, our proposed model and calculated results may provide insights into the development of treatment schemes and also have other clinic implications. link: http://identifiers.org/pubmed/30463617

Parameters:

NameDescription
myu = 0.001Reaction: p_0 => p_1, Rate Law: compartment*myu*p_0

States:

NameDescription
p 3[C19700; pancreatic carcinoma]
p 1[pancreatic carcinoma; C19700]
p 6[C19700; pancreatic carcinoma]
p 8[C19700; pancreatic carcinoma]
p 7[C19700; pancreatic carcinoma]
p 0[pancreatic carcinoma; C19700]
p 4[C19700; pancreatic carcinoma]
p 2[C19700; pancreatic carcinoma]
p 5[C19700; pancreatic carcinoma]

Susree2017 - Role of platelet count and inhibitors in blood coagulation: MODEL1808130001v0.0.1

Mathematical model of blood coagulation that incorporates platelet binding sites.

Details

A mechanistic model including the role of platelets is proposed for clot formation and growth in plasma in vitro. Initiation of clot formation is by the addition of tissue factor, and initiation via the intrinsic pathway is neglected. Activation of zymogens follows the extrinsic pathway cascade and reactions on platelet membranes are included. Platelet activation occurs due to thrombin and also due to other activated platelets. Inhibition of the active clotting factors is by ATIII and TFPI, whereas inhibition due to APC is not relevant in the conditions modeled. The model predictions matched existing data for thrombin production in synthetic plasma. The model predicts that inhibition of platelet-driven activation of platelets has a major effect on concentration of activated platelets in PRP, normal plasma and PPP. Inhibition of platelet activation by (other activated) platelets significantly delays thrombin production in PRP and normal plasma as compared to that by thrombin. Further, sensitivity analysis shows that the model is most sensitive to the activation of platelet membrane-bound factor X by the intrinsic tenase complex. link: http://identifiers.org/doi/10.1007/s12046-017-0602-3

Susree2018 - Effect of coated platelets on thrombin generation: MODEL1808080001v0.0.1

Mathematical model of blood coagulation that simulates the effect of coated platelets on thrombin generation.

Details

Platelets play a crucial role in the initiation, progress, termination as well as regulation of blood coagulation. Recent studies have confirmed that not all but only a small percentage of thrombin-activated platelets ("coated" platelets) exhibit procoagulant properties (namely the expression of phosphatidylserine binding sites) required for the acceleration and progress of coagulation. A mechanistic model is developed for in vitro coagulation whose key features are distinct equations for coated platelets, thrombin dose-dependence for coated platelets, and competitive binding of coagulation factors to platelet membrane. Model predictions show significant delay in the onset of peak Va production, and peak thrombin production when dose-dependence is incorporated instead of a fixed theoretical maximum percentage of coated platelets. Further, peak thrombin concentration is significantly overestimated when either fractional presence of coated platelets is ignored (by 299.4%) or when dose-dependence on thrombin is ignored (by 24.7%). link: http://identifiers.org/pubmed/29782929

Suthers2009 - Genome-scale metabolic network of Mycoplasma genitalium (iPS189): MODEL1507180052v0.0.1

Suthers2009 - Genome-scale metabolic network of Mycoplasma genitalium (iPS189)This model is described in the article: […

Details

With a genome size of approximately 580 kb and approximately 480 protein coding regions, Mycoplasma genitalium is one of the smallest known self-replicating organisms and, additionally, has extremely fastidious nutrient requirements. The reduced genomic content of M. genitalium has led researchers to suggest that the molecular assembly contained in this organism may be a close approximation to the minimal set of genes required for bacterial growth. Here, we introduce a systematic approach for the construction and curation of a genome-scale in silico metabolic model for M. genitalium. Key challenges included estimation of biomass composition, handling of enzymes with broad specificities, and the lack of a defined medium. Computational tools were subsequently employed to identify and resolve connectivity gaps in the model as well as growth prediction inconsistencies with gene essentiality experimental data. The curated model, M. genitalium iPS189 (262 reactions, 274 metabolites), is 87% accurate in recapitulating in vivo gene essentiality results for M. genitalium. Approaches and tools described herein provide a roadmap for the automated construction of in silico metabolic models of other organisms. link: http://identifiers.org/pubmed/19214212

Swat2004_Mammalian_G1_S_Transition: BIOMD0000000228v0.0.1

This is the extended model described the article: Bifurcation analysis of the regulatory modules of the mammalian G1/S…

Details

MOTIVATION: Mathematical models of the cell cycle can contribute to an understanding of its basic mechanisms. Modern simulation tools make the analysis of key components and their interactions very effective. This paper focuses on the role of small modules and feedbacks in the gene-protein network governing the G1/S transition in mammalian cells. Mutations in this network may lead to uncontrolled cell proliferation. Bifurcation analysis helps to identify the key components of this extremely complex interaction network. RESULTS: We identify various positive and negative feedback loops in the network controlling the G1/S transition. It is shown that the positive feedback regulation of E2F1 and a double activator-inhibitor module can lead to bistability. Extensions of the core module preserve the essential features such as bistability. The complete model exhibits a transcritical bifurcation in addition to bistability. We relate these bifurcations to the cell cycle checkpoint and the G1/S phase transition point. Thus, core modules can explain major features of the complex G1/S network and have a robust decision taking function. link: http://identifiers.org/pubmed/15231543

Parameters:

NameDescription
Km1 = 0.5; k1 = 1.0; J11 = 0.5; J61 = 5.0Reaction: => pRB; pRB, pRBp, E2F1, Rate Law: cell*k1*E2F1/(Km1+E2F1)*J11/(J11+pRB)*J61/(J61+pRBp)
phi_pRBp = 0.06Reaction: pRBp =>, Rate Law: cell*phi_pRBp*pRBp
phi_CycEa = 0.05Reaction: CycEa =>, Rate Law: cell*phi_CycEa*CycEa
k98 = 0.01Reaction: CycEa => CycEi, Rate Law: cell*k98*CycEa
J15 = 0.001; k25 = 0.9; Fm = 0.005; J65 = 6.0Reaction: => AP1; E2F1, pRB, pRBp, Rate Law: cell*(Fm+k25*E2F1*J15/(J15+pRB)*J65/(J65+pRBp))
k67 = 0.7Reaction: pRBp => pRBpp; pRBp, CycEa, Rate Law: cell*k67*pRBp*CycEa
k34 = 0.04; Km4 = 0.3Reaction: CycDi => CycDa; CycDi, CycDa, Rate Law: cell*k34*CycDi*CycDa/(Km4+CycDa)
k3 = 0.05; J63 = 2.0; k23 = 0.3; J13 = 0.002Reaction: => CycDi; E2F1, pRB, pRBp, AP1, Rate Law: cell*(k3*AP1+k23*E2F1*J13/(J13+pRB)*J63/(J63+pRBp))
k61 = 0.3Reaction: pRBp => pRB, Rate Law: cell*k61*pRBp
phi_CycEi = 0.06Reaction: CycEi =>, Rate Law: cell*phi_CycEi*CycEi
phi_pRBpp = 0.04Reaction: pRBpp =>, Rate Law: cell*phi_pRBpp*pRBpp
phi_E2F1 = 0.1Reaction: E2F1 =>, Rate Law: cell*phi_E2F1*E2F1
k43 = 0.01Reaction: CycDa => CycDi, Rate Law: cell*k43*CycDa
phi_AP1 = 0.01Reaction: AP1 =>, Rate Law: cell*phi_AP1*AP1
J18 = 0.6; J68 = 7.0; k28 = 0.06Reaction: => CycEi; E2F1, pRB, pRBp, Rate Law: cell*k28*E2F1*J18/(J18+pRB)*J68/(J68+pRBp)
J12 = 5.0; k2 = 1.6; kp = 0.05; J62 = 8.0; Km2 = 4.0; a = 0.04Reaction: => E2F1; E2F1, pRB, pRBp, Rate Law: cell*(kp+k2*(a^2+E2F1^2)/(Km2^2+E2F1^2)*J12/(J12+pRB)*J62/(J62+pRBp))
k16 = 0.4Reaction: pRB => pRBp; pRB, CycDa, Rate Law: cell*k16*pRB*CycDa
phi_pRB = 0.005Reaction: pRB =>, Rate Law: cell*phi_pRB*pRB
phi_CycDa = 0.03Reaction: CycDa =>, Rate Law: cell*phi_CycDa*CycDa
k76 = 0.1Reaction: pRBpp => pRBp, Rate Law: cell*k76*pRBpp
phi_CycDi = 0.023Reaction: CycDi =>, Rate Law: cell*phi_CycDi*CycDi
Km9 = 0.005; k89 = 0.07Reaction: CycEi => CycEa; CycEi, CycEa, Rate Law: cell*k89*CycEi*CycEa/(Km9+CycEa)

States:

NameDescription
AP1[Proto-oncogene c-Fos]
E2F1[Transcription factor E2F1]
CycEi[G1/S-specific cyclin-E1; G1/S-specific cyclin-E2; Cyclin-dependent kinase 2]
CycDa[Cyclin-dependent kinase 6; Cyclin-dependent kinase 4; G1/S-specific cyclin-D3]
pRBpp[Retinoblastoma-associated protein]
CycDi[G1/S-specific cyclin-D3; Cyclin-dependent kinase 4; Cyclin-dependent kinase 6]
CycEa[G1/S-specific cyclin-E1; G1/S-specific cyclin-E2; Cyclin-dependent kinase 2]
pRBp[Retinoblastoma-associated protein]
pRB[Retinoblastoma-associated protein]

Szappanos2011_GeneticInteractionNetwork_YeastMetabolism: MODEL1107190000v0.0.1

This model is from the article: An integrated approach to characterize genetic interaction networks in yeast metabolis…

Details

Although experimental and theoretical efforts have been applied to globally map genetic interactions, we still do not understand how gene-gene interactions arise from the operation of biomolecular networks. To bridge the gap between empirical and computational studies, we i, quantitatively measured genetic interactions between ∼185,000 metabolic gene pairs in Saccharomyces cerevisiae, ii, superposed the data on a detailed systems biology model of metabolism and iii, introduced a machine-learning method to reconcile empirical interaction data with model predictions. We systematically investigated the relative impacts of functional modularity and metabolic flux coupling on the distribution of negative and positive genetic interactions. We also provide a mechanistic explanation for the link between the degree of genetic interaction, pleiotropy and gene dispensability. Last, we show the feasibility of automated metabolic model refinement by correcting misannotations in NAD biosynthesis and confirming them by in vivo experiments. link: http://identifiers.org/pubmed/21623372

Szeliova 2020 - biomass specific iCHO model (iCHO_DGpar-8mMCD): MODEL1907260012v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_DXepo-0mMCD): MODEL1907260010v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_DXepo-8mMCD): MODEL1907260008v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_DXpar-8mMCD): MODEL1907260011v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_GScd4-8mMCD): MODEL1907260006v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_GSher-8mMCD): MODEL1907260007v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_GSpar-8mMCD): MODEL1907260009v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_HYher-8mMCD): MODEL1907260004v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_HYpar-8mMCD): MODEL1907260005v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_K1par-0mMCD): MODEL1907260015v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_K1par-8mMAP): MODEL1907260014v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_K1par-8mMCD): MODEL1907260016v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szeliova 2020 - biomass specific iCHO model (iCHO_S-par-8mMCD): MODEL1907260013v0.0.1

iCHO1766 model from Hefzi et al. (2016) with cell line specific biomass composition and exchange rates

Details

Background Cell line-specific, genome-scale metabolic models enable rigorous and systematic in silico investigation of cellular metabolism. Such models have recently become available for Chinese hamster ovary (CHO) cells. However, a key ingredient, namely an experimentally validated biomass function that summarizes the cellular composition, was so far missing. Here, we close this gap by providing extensive experimental data on the biomass composition of 13 parental and producer CHO cell lines under various conditions.

Results We report total protein, lipid, DNA, RNA and carbohydrate content, cell dry mass, and detailed protein and lipid composition. Furthermore, we present meticulous data on exchange rates between cells and environment and provide detailed experimental protocols on how to determine all of the above. The biomass composition is converted into cell line- and condition-specific biomass functions for use in cell line-specific, genome-scale metabolic models of CHO. Finally, flux-balance analysis (FBA) is used to demonstrate consistency between in silico predictions and experimental analysis.

Conclusions Our study reveals a strong variability of the total protein content and cell dry mass across cell lines. However, the relative amino acid composition is independent of the cell line and condition and thus needs not be explicitly measured for each new cell line. In contrast, the lipid composition is strongly influenced by the growth media and thus will have to be determined in each case. These cell line-specific variations in biomass composition have a small impact on growth rate predictions with FBA, as inaccuracies in the predictions are rather dominated by inaccuracies in the exchange rate spectra. Cell-specific biomass variations only become important if the experimental errors in the exchange rate spectra drop below twenty percent.

Keywords Chinese hamster ovary, biomass composition, metabolic modelling, flux balance analysis, uptake rates, secretion rates link: http://identifiers.org/doi/10.1016/j.ymben.2020.06.002

Szymanska2009 - Mathematical modeling of heat shock protein synthesis in response to temperature change: BIOMD0000000896v0.0.1

This is a mathematical model of heat shock protein synthesis induced by an external temperature stimulus. The model cons…

Details

One of the most important questions in cell biology is how cells cope with rapid changes in their environment. The range of common molecular responses includes a dramatic change in the pattern of gene expression and the elevated synthesis of so-called heat shock (or stress) proteins (HSPs). Induction of HSPs increases cell survival under stress conditions [Morimoto, R.I., 1993. Cells in stress: transcriptional activation of heat shock genes. Science 259, 1409-1410]. In this paper we propose a mathematical model of heat shock protein synthesis induced by an external temperature stimulus. Our model consists of a system of nine nonlinear ordinary differential equations describing the temporal evolution of the key variables involved in the regulation of HSP synthesis. Computational simulations of our model are carried out for different external temperature stimuli. We compare our model predictions with experimental data for three different cases-one corresponding to heat shock, the second corresponding to slow heating conditions and the third corresponding to a short heat shock (lasting about 40 min). We also present our model predictions for heat shocks carried out up to different final temperatures and finally we present a new hypothesis concerning the molecular response to stress that explains some phenomena observed in experiments. link: http://identifiers.org/pubmed/19327370

Parameters:

NameDescription
k_2 = 0.42Reaction: Hsp70 + S => Hsp70_S, Rate Law: compartment*k_2*Hsp70*S
k_3 = 0.023Reaction: HSF => HSF_3, Rate Law: compartment*k_3*HSF^3
k_7 = 0.035Reaction: HSF_3 + HSE => HSF_3_HSE, Rate Law: compartment*k_7*HSF_3*HSE
k_10 = 0.014Reaction: Hsp70_S => Hsp70, Rate Law: compartment*k_10*Hsp70_S
k_4 = 0.035Reaction: mRNA => Hsp70, Rate Law: compartment*k_4*mRNA
F_T = 0.099Reaction: => S, Rate Law: compartment*F_T
l_1 = 0.005Reaction: Hsp70_HSF => Hsp70 + HSF, Rate Law: compartment*l_1*Hsp70_HSF
l_6 = 3.6E-4Reaction: Hsp70_S + HSF => Hsp70_HSF + S, Rate Law: compartment*l_6*Hsp70_S*HSF
l_7 = 0.035Reaction: HSF_3_HSE => HSE + HSF_3, Rate Law: compartment*l_7*HSF_3_HSE
k_8 = 0.035Reaction: => mRNA; HSF_3_HSE, Rate Law: compartment*k_8*HSF_3_HSE
l_3 = 0.00575Reaction: HSF_3 + Hsp70 => HSF + Hsp70_HSF, Rate Law: compartment*l_3*HSF_3*Hsp70
l_10 = 0.013Reaction: Hsp70 =>, Rate Law: compartment*l_10*Hsp70
l_2 = 0.005Reaction: Hsp70_S => Hsp70 + S, Rate Law: compartment*l_2*Hsp70_S
k_6 = 0.023Reaction: S + Hsp70_HSF => Hsp70_S + HSF, Rate Law: compartment*k_6*S*Hsp70_HSF
k_1 = 0.42Reaction: Hsp70 + HSF => Hsp70_HSF, Rate Law: compartment*k_1*Hsp70*HSF

States:

NameDescription
S[C120264; MI:0908]
Hsp70 HSF[C71446; C17765]
Hsp70 S[C17765; C120264]
HSF[C71446]
HSF 3 HSE[SO:0001850; C71446]
HSE[SO:0001850]
mRNA[C17765; Messenger RNA]
HSF 3[C71446]
Hsp70[C17765]

T


Tabak2007_dopamine: BIOMD0000000138v0.0.1

The model is encoded according to the paper *Low dose of dopamine may stimulate prolactin secretion by increasing fast p…

Details

Dopamine (DA) released from the hypothalamus tonically inhibits pituitary lactotrophs. DA (at micromolar concentration) opens potassium channels, hyperpolarizing the lactotrophs and thus preventing the calcium influx that triggers prolactin hormone release. Surprisingly, at concentrations approximately 1000 lower, DA can stimulate prolactin secretion. Here, we investigated whether an increase in a K+ current could mediate this stimulatory effect. We considered the fast K+ currents flowing through large-conductance BK channels and through A-type channels. We developed a minimal lactotroph model to investigate the effects of these two currents. Both IBK and IA could transform the electrical pattern of activity from spiking to bursting, but through distinct mechanisms. IBK always increased the intracellular Ca2+ concentration, while IA could either increase or decrease it. Thus, the stimulatory effects of DA could be mediated by a fast K+ conductance which converts tonically spiking cells to bursters. In addition, the study illustrates that link: http://identifiers.org/pubmed/17058022

Parameters:

NameDescription
ff = 0.01; ica = NaN; kc = 0.16; alpha = 0.0015Reaction: => c, Rate Law: (-ff)*(alpha*ica+kc*c)*cell

States:

NameDescription
c[calcium(2+); Calcium cation]

Tabak2010_NeuronalNetworks: MODEL1006230028v0.0.1

This a model from the article: Mechanism for the universal pattern of activity in developing neuronal networks. Taba…

Details

Spontaneous episodic activity is a fundamental mode of operation of developing networks. Surprisingly, the duration of an episode of activity correlates with the length of the silent interval that precedes it, but not with the interval that follows. Here we use a modeling approach to explain this characteristic, but thus far unexplained, feature of developing networks. Because the correlation pattern is observed in networks with different structures and components, a satisfactory model needs to generate the right pattern of activity regardless of the details of network architecture or individual cell properties. We thus developed simple models incorporating excitatory coupling between heterogeneous neurons and activity-dependent synaptic depression. These models robustly generated episodic activity with the correct correlation pattern. The correlation pattern resulted from episodes being triggered at random levels of recovery from depression while they terminated around the same level of depression. To explain this fundamental difference between episode onset and termination, we used a mean field model, where only average activity and average level of recovery from synaptic depression are considered. In this model, episode onset is highly sensitive to inputs. Thus noise resulting from random coincidences in the spike times of individual neurons led to the high variability at episode onset and to the observed correlation pattern. This work further shows that networks with widely different architectures, different cell types, and different functions all operate according to the same general mechanism early in their development. link: http://identifiers.org/pubmed/20164396

Takahashi2015 - Zinc regulation E.coli: MODEL1502180000v0.0.1

This a model from the article: The dynamic balance of import and export of zinc in Escherichia coli suggests a heter…

Details

Zinc is essential for life, but toxic in excess. Thus all cells must control their internal zinc concentration. We used a systems approach, alternating rounds of experiments and models, to further elucidate the zinc control systems in Escherichia coli. We measured the response to zinc of the main specific zinc import and export systems in the wild-type, and a series of deletion mutant strains. We interpreted these data with a detailed mathematical model and Bayesian model fitting routines. There are three key findings: first, that alternate, non-inducible importers and exporters are important. Second, that an internal zinc reservoir is essential for maintaining the internal zinc concentration. Third, our data fitting led us to propose that the cells mount a heterogeneous response to zinc: some respond effectively, while others die or stop growing. In a further round of experiments, we demonstrated lower viable cell counts in the mutant strain tested exposed to excess zinc, consistent with this hypothesis. A stochastic model simulation demonstrated considerable fluctuations in the cellular levels of the ZntA exporter protein, reinforcing this proposal. We hypothesize that maintaining population heterogeneity could be a bet-hedging response allowing a population of cells to survive in varied and fluctuating environments. link: http://identifiers.org/pubmed/25808337

Talemi2014 - Arsenic toxicity and detoxification mechanisms in yeast: BIOMD0000000547v0.0.1

Talemi2014 - Arsenic toxicity and detoxification mechanisms in yeastThe model implements arsenite (AsIII) transport regu…

Details

Arsenic has a dual role as causative and curative agent of human disease. Therefore, there is considerable interest in elucidating arsenic toxicity and detoxification mechanisms. By an ensemble modelling approach, we identified a best parsimonious mathematical model which recapitulates and predicts intracellular arsenic dynamics for different conditions and mutants, thereby providing novel insights into arsenic toxicity and detoxification mechanisms in yeast, which could partly be confirmed experimentally by dedicated experiments. Specifically, our analyses suggest that: (i) arsenic is mainly protein-bound during short-term (acute) exposure, whereas glutathione-conjugated arsenic dominates during long-term (chronic) exposure, (ii) arsenic is not stably retained, but can leave the vacuole via an export mechanism, and (iii) Fps1 is controlled by Hog1-dependent and Hog1-independent mechanisms during arsenite stress. Our results challenge glutathione depletion as a key mechanism for arsenic toxicity and instead suggest that (iv) increased glutathione biosynthesis protects the proteome against the damaging effects of arsenic and that (v) widespread protein inactivation contributes to the toxicity of this metalloid. Our work in yeast may prove useful to elucidate similar mechanisms in higher eukaryotes and have implications for the use of arsenic in medical therapy. link: http://identifiers.org/pubmed/24798644

Parameters:

NameDescription
parameter_42 = 6.56918E-4Reaction: species_2 => species_1; species_2, Rate Law: parameter_42*species_2
parameter_32 = 0.0719168Reaction: species_15 => species_11; species_15, Rate Law: parameter_32*species_15
parameter_30 = 1102.15; parameter_31 = 0.0730991; parameter_29 = 2.57134E-4Reaction: species_11 => species_15; species_1, species_10, species_11, species_1, species_10, Rate Law: compartment_3*species_11/compartment_3*(parameter_29*species_1/compartment_4+parameter_30*species_10/compartment_3+parameter_31)
parameter_38 = 1.0; parameter_36 = 3.49703E-6Reaction: species_3 => species_4; species_5, species_5, species_3, Rate Law: parameter_38*species_5/compartment_3*parameter_36*(36*pi)^(1/3)*compartment_1^(2/3)*species_3/compartment_4
parameter_35 = 5.16159E-6; parameter_34 = 1.0Reaction: species_1 => species_6; species_14, species_14, species_1, Rate Law: (36*pi)^(1/3)*compartment_3^(2/3)*species_14/compartment_3*parameter_34*species_1/compartment_4/(parameter_35+species_1/compartment_4)
parameter_43 = 9.01422E-13Reaction: species_14 => ; species_14, Rate Law: parameter_43*species_14
parameter_33 = 0.00215551Reaction: species_6 => species_1; species_11, species_11, species_6, species_1, Rate Law: (36*pi)^(1/3)*compartment_3^(2/3)*species_11/compartment_3*parameter_33*(species_6/compartment_2-species_1/compartment_4)
parameter_28 = 1.0; parameter_26 = 0.0757274Reaction: species_9 => species_10; species_1, species_9, species_1, Rate Law: compartment_3*parameter_26*parameter_28*species_1/compartment_4*species_9/compartment_3
parameter_40 = 6.1432Reaction: species_3 => species_1 + species_7; species_3, Rate Law: parameter_40*species_3
parameter_41 = 0.00880734Reaction: species_1 => species_2; species_1, Rate Law: parameter_41*species_1
parameter_37 = 1.92773E-7Reaction: species_4 => species_3; species_4, Rate Law: (36*pi)^(1/3)*compartment_1^(2/3)*parameter_37*species_4/compartment_1
parameter_27 = 161.334Reaction: species_10 => species_9; species_10, Rate Law: parameter_27*species_10
parameter_39 = 0.202797; parameter_22 = 1.0Reaction: species_1 + species_7 => species_3; species_1, species_7, Rate Law: compartment_4*parameter_39*parameter_22*species_1/compartment_4*species_7/compartment_4
parameter_7 = 0.0; parameter_9 = 30.0; parameter_5 = 100.0; parameter_8 = 3600.0; parameter_6 = 1000.0Reaction: species_6 = piecewise(parameter_5, time < parameter_7, piecewise((parameter_5+parameter_6)*exp((parameter_8-time)/parameter_9), time > parameter_8, parameter_5+parameter_6*(1-exp((parameter_7-time)/parameter_9))))*compartment_2, Rate Law: missing
parameter_2 = 2.16561157822054E-17; parameter_25 = 1.0Reaction: => species_14; species_1, species_1, Rate Law: compartment_3*parameter_2*parameter_25*species_1/compartment_4

States:

NameDescription
species 9[Mitogen-activated protein kinase HOG1]
species 2[arsenite(1-); protein]
species 6[arsenite(1-); extracellular region]
species 10[Mitogen-activated protein kinase HOG1; phosphorylated]
species 11[Glycerol uptake/efflux facilitator protein]
species 1[arsenite(1-); intracellular]
species 4[arsenite(1-); vacuolar part]
species 14[Arsenical-resistance protein 3]
species 3[arsenite(1-)]
species 7[glutathione]
species 15[Glycerol uptake/efflux facilitator protein; phosphorylated]

Talemi2015 - Persistent telomere-associated DNA damage foci (TAF), a measure to predict cancer risks: MODEL1412200000v0.0.1

A Robust Model of DNA Damage Dynamics. Rasgou Talemi and Schaber, 12.20.2014.

Details

Mathematical modelling has been instrumental to understand kinetics of radiation-induced DNA damage repair and associated secondary cancer risk. The widely accepted two-lesion kinetic (TLK) model assumes two kinds of double strand breaks, simple and complex ones, with different repair rates. Recently, persistent DNA damage associated with telomeres was reported as a new kind of DNA damage. We therefore extended existing versions of the TLK model by new categories of DNA damage and re-evaluated those models using extensive data. We subjected different versions of the TLK model to a rigorous model discrimination approach. This enabled us to robustly select a best approximating parsimonious model that can both recapitulate and predict transient and persistent DNA damage after ionizing radiation. Models and data argue for i) nonlinear dose-damage relationships, and ii) negligible saturation of repair kinetics even for high doses. Additionally, we show that simulated radiation-induced persistent telomere-associated DNA damage foci (TAF) can be used to predict excess relative risk (ERR) of developing secondary leukemia after fractionated radiotherapy. We suggest that TAF may serve as an additional measure to predict cancer risk after radiotherapy using high dose rates. This may improve predicting risk-dose dependency of ionizing radiation especially for long-term therapies. link: http://identifiers.org/pubmed/26359627

Talemi2016 - Yeast osmo-homoestasis: MODEL1606100000v0.0.1

Talemi2016 - Yeast osmo-homoestasisThis model is described in the article: [Systems Level Analysis of the Yeast Osmo-St…

Details

Adaptation is an important property of living organisms enabling them to cope with environmental stress and maintaining homeostasis. Adaptation is mediated by signaling pathways responding to different stimuli. Those signaling pathways might communicate in order to orchestrate the cellular response to multiple simultaneous stimuli, a phenomenon called crosstalk. Here, we investigate possible mechanisms of crosstalk between the High Osmolarity Glycerol (HOG) and the Cell Wall Integrity (CWI) pathways in yeast, which mediate adaptation to hyper- and hypo-osmotic challenges, respectively. We combine ensemble modeling with experimental investigations to test in quantitative terms different hypotheses about the crosstalk of the HOG and the CWI pathways. Our analyses indicate that for the conditions studied i) the CWI pathway activation employs an adaptive mechanism with a variable volume-dependent threshold, in contrast to the HOG pathway, whose activation relies on a fixed volume-dependent threshold, ii) there is no or little direct crosstalk between the HOG and CWI pathways, and iii) its mainly the HOG alone mediating adaptation of cellular osmotic pressure for both hyper- as well as hypo-osmotic stress. Thus, by iteratively combining mathematical modeling with experimentation we achieved a better understanding of regulatory mechanisms of yeast osmo-homeostasis and formulated new hypotheses about osmo-sensing. link: http://identifiers.org/pubmed/27515486

Tan2012 - Antibiotic Treatment, Inoculum Effect: BIOMD0000000425v0.0.1

Tan2012 - Antibiotic Treatment, Inoculum EffectThe efficacy of many antibiotics decreases with increasing bacterial dens…

Details

The inoculum effect (IE) refers to the decreasing efficacy of an antibiotic with increasing bacterial density. It represents a unique strategy of antibiotic tolerance and it can complicate design of effective antibiotic treatment of bacterial infections. To gain insight into this phenomenon, we have analyzed responses of a lab strain of Escherichia coli to antibiotics that target the ribosome. We show that the IE can be explained by bistable inhibition of bacterial growth. A critical requirement for this bistability is sufficiently fast degradation of ribosomes, which can result from antibiotic-induced heat-shock response. Furthermore, antibiotics that elicit the IE can lead to 'band-pass' response of bacterial growth to periodic antibiotic treatment: the treatment efficacy drastically diminishes at intermediate frequencies of treatment. Our proposed mechanism for the IE may be generally applicable to other bacterial species treated with antibiotics targeting the ribosomes. link: http://identifiers.org/pubmed/23047527

Parameters:

NameDescription
kd=1.0Reaction: c => ; c, Rate Law: kd*c
kappa=0.5Reaction: => c; c, Rate Law: c/(kappa+c)
gamma=1.0E-5; phi=5.0E-6; delta=1.0E-5Reaction: c => ; c, Rate Law: phi*c/(delta+gamma*c)
alpha=0.001Reaction: => c, Rate Law: alpha

States:

NameDescription
c[30S ribosomal protein S1; 50S ribosomal protein L1]

Tang2010_PolyGlutamate: BIOMD0000000285v0.0.1

This a model from the article: Experimental and computational analysis of polyglutamine-mediated cytotoxicity. Tang…

Details

Expanded polyglutamine (polyQ) proteins are known to be the causative agents of a number of human neurodegenerative diseases but the molecular basis of their cytoxicity is still poorly understood. PolyQ tracts may impede the activity of the proteasome, and evidence from single cell imaging suggests that the sequestration of polyQ into inclusion bodies can reduce the proteasomal burden and promote cell survival, at least in the short term. The presence of misfolded protein also leads to activation of stress kinases such as p38MAPK, which can be cytotoxic. The relationships of these systems are not well understood. We have used fluorescent reporter systems imaged in living cells, and stochastic computer modeling to explore the relationships of polyQ, p38MAPK activation, generation of reactive oxygen species (ROS), proteasome inhibition, and inclusion body formation. In cells expressing a polyQ protein inclusion, body formation was preceded by proteasome inhibition but cytotoxicity was greatly reduced by administration of a p38MAPK inhibitor. Computer simulations suggested that without the generation of ROS, the proteasome inhibition and activation of p38MAPK would have significantly reduced toxicity. Our data suggest a vicious cycle of stress kinase activation and proteasome inhibition that is ultimately lethal to cells. There was close agreement between experimental data and the predictions of a stochastic computer model, supporting a central role for proteasome inhibition and p38MAPK activation in inclusion body formation and ROS-mediated cell death. link: http://identifiers.org/pubmed/20885783

Parameters:

NameDescription
kproteff = 1.0; kalive = 1.0; kdegMisP = 0.01Reaction: MisP_Proteasome => Proteasome, Rate Law: kdegMisP*MisP_Proteasome*kalive*kproteff
kalive = 1.0; kgenROSAggP = 5.0E-6Reaction: AggP3 => AggP3 + ROS, Rate Law: kgenROSAggP*AggP3*kalive
kdegPolyQ = 0.0025; kproteff = 1.0; kalive = 1.0Reaction: PolyQ_Proteasome => Proteasome, Rate Law: kdegPolyQ*PolyQ_Proteasome*kalive*kproteff
kalive = 1.0; kgenROSp38 = 7.0E-4; kp38act = 1.0Reaction: p38_P => p38_P + ROS, Rate Law: kgenROSp38*p38_P*kp38act*kalive
kalive = 1.0; ksynNatP = 2.4Reaction: Source => NatP, Rate Law: ksynNatP*Source*kalive
kalive = 1.0; kdisaggMisP5 = 1.0E-7Reaction: AggP5 => MisP + AggP4, Rate Law: kdisaggMisP5*AggP5*kalive
kalive = 1.0; kseqPolyQProt = 5.0E-7Reaction: PolyQ_Proteasome + SeqAggP => SeqAggP, Rate Law: kseqPolyQProt*PolyQ_Proteasome*SeqAggP*kalive
kalive = 1.0; kPIdeath = 2.5E-8Reaction: AggP_Proteasome => AggP_Proteasome + PIdeath, Rate Law: kPIdeath*AggP_Proteasome*kalive
kalive = 1.0; kbinmRFPu = 5.0E-7Reaction: mRFPu + Proteasome => mRFPu_Proteasome, Rate Law: kbinmRFPu*mRFPu*Proteasome*kalive
kproteff = 1.0; kalive = 1.0; kdegmRFPu = 0.005Reaction: mRFPu_Proteasome => Proteasome, Rate Law: kdegmRFPu*mRFPu_Proteasome*kalive*kproteff
kalive = 1.0; kdisaggMisP2 = 4.0E-7Reaction: AggP2 => MisP + AggP1, Rate Law: kdisaggMisP2*AggP2*kalive
kalive = 1.0; kbinPolyQ = 5.0E-8Reaction: PolyQ + Proteasome => PolyQ_Proteasome, Rate Law: kbinPolyQ*PolyQ*Proteasome*kalive
kalive = 1.0; kgenROS = 0.0017Reaction: Source => ROS, Rate Law: kgenROS*Source*kalive
krelMisPProt = 1.0E-8; kalive = 1.0Reaction: MisP_Proteasome => MisP + Proteasome, Rate Law: krelMisPProt*MisP_Proteasome*kalive
kdisaggPolyQ3 = 3.0E-7; kalive = 1.0Reaction: AggPolyQ3 => PolyQ + AggPolyQ2, Rate Law: kdisaggPolyQ3*AggPolyQ3*kalive
kactp38 = 5.0E-6; kalive = 1.0Reaction: ROS + p38 => ROS + p38_P, Rate Law: kactp38*ROS*p38*kalive
kalive = 1.0; kagg2MisP = 1.0E-10Reaction: MisP + AggP1 => AggP2, Rate Law: kagg2MisP*MisP*AggP1*kalive
kdisaggPolyQ1 = 5.0E-7; kalive = 1.0Reaction: AggPolyQ1 => PolyQ, Rate Law: kdisaggPolyQ1*AggPolyQ1*kalive
kalive = 1.0; kdisaggPolyQ4 = 2.0E-7Reaction: AggPolyQ4 => PolyQ + AggPolyQ3, Rate Law: kdisaggPolyQ4*AggPolyQ4*kalive
kalive = 1.0; ksynmRFPu = 0.138Reaction: Source => mRFPu, Rate Law: ksynmRFPu*Source*kalive
kalive = 1.0; krelmRFPu = 1.0E-8Reaction: mRFPu_Proteasome => mRFPu + Proteasome, Rate Law: krelmRFPu*mRFPu_Proteasome*kalive
kalive = 1.0; kbinMisPProt = 5.0E-8Reaction: MisP + Proteasome => MisP_Proteasome, Rate Law: kbinMisPProt*MisP*Proteasome*kalive
kalive = 1.0; kseqmRFPuProt = 5.0E-7Reaction: mRFPu_Proteasome + SeqAggP => SeqAggP, Rate Law: kseqmRFPuProt*mRFPu_Proteasome*SeqAggP*kalive
kalive = 1.0; kinhprot = 5.0E-9Reaction: AggP3 + Proteasome => AggP_Proteasome, Rate Law: kinhprot*AggP3*Proteasome*kalive
kdisaggMisP3 = 3.0E-7; kalive = 1.0Reaction: AggP3 => MisP + AggP2, Rate Law: kdisaggMisP3*AggP3*kalive
kalive = 1.0; kgenROSSeqAggP = 1.0E-7Reaction: SeqAggP => SeqAggP + ROS, Rate Law: kgenROSSeqAggP*SeqAggP*kalive
kalive = 1.0; kaggPolyQ = 5.0E-8Reaction: PolyQ + ROS => AggPolyQ1 + ROS, Rate Law: kaggPolyQ*PolyQ*(PolyQ-1)*0.5*ROS^2/(10^2+ROS^2)*kalive
kseqMisPProt = 5.0E-7; kalive = 1.0Reaction: MisP_Proteasome + SeqAggP => SeqAggP, Rate Law: kseqMisPProt*MisP_Proteasome*SeqAggP*kalive
kmisfold = 2.0E-6; kalive = 1.0Reaction: NatP + ROS => MisP + ROS, Rate Law: kmisfold*NatP*ROS*kalive
kalive = 1.0; kremROS = 2.0E-4Reaction: ROS => Sink, Rate Law: kremROS*ROS*kalive
kalive = 1.0; kdisaggMisP1 = 5.0E-7Reaction: AggP1 => MisP, Rate Law: kdisaggMisP1*AggP1*kalive
kseqmRFPu = 1.0E-10; kalive = 1.0Reaction: mRFPu + SeqAggP => SeqAggP, Rate Law: kseqmRFPu*mRFPu*SeqAggP*kalive
kdisaggPolyQ2 = 4.0E-7; kalive = 1.0Reaction: AggPolyQ2 => PolyQ + AggPolyQ1, Rate Law: kdisaggPolyQ2*AggPolyQ2*kalive
krelPolyQ = 1.0E-9; kalive = 1.0Reaction: PolyQ_Proteasome => PolyQ + Proteasome, Rate Law: krelPolyQ*PolyQ_Proteasome*kalive
kseqPolyQ = 8.0E-7; kalive = 1.0Reaction: PolyQ + SeqAggP => SeqAggP, Rate Law: kseqPolyQ*PolyQ*SeqAggP*kalive
kaggMisP = 1.0E-11; kalive = 1.0Reaction: MisP => AggP1, Rate Law: kaggMisP*MisP*(MisP-1)*0.5*kalive
kseqAggPProt = 5.0E-7; kalive = 1.0Reaction: AggP_Proteasome + SeqAggP => SeqAggP, Rate Law: kseqAggPProt*AggP_Proteasome*SeqAggP*kalive
kseqMisP = 1.0E-9; kalive = 1.0Reaction: MisP + SeqAggP => SeqAggP, Rate Law: kseqMisP*MisP*SeqAggP*kalive
kalive = 1.0; kinactp38 = 0.002Reaction: p38_P => p38, Rate Law: kinactp38*p38_P*kalive
kdisaggMisP4 = 2.0E-7; kalive = 1.0Reaction: AggP4 => MisP + AggP3, Rate Law: kdisaggMisP4*AggP4*kalive
kdisaggPolyQ5 = 1.0E-7; kalive = 1.0Reaction: AggPolyQ5 => PolyQ + AggPolyQ4, Rate Law: kdisaggPolyQ5*AggPolyQ5*kalive

States:

NameDescription
AggP3AggP3
Proteasome[proteasome complex]
ROSROS
AggP4AggP4
mRFPumRFPu
AggP ProteasomeAggP_Proteasome
p38 Pp38_P
AggPolyQ4AggPolyQ4
PolyQ[Huntingtin]
AggP1AggP1
SeqAggPSeqAggP
AggP2AggP2
mRFPu ProteasomemRFPu_Proteasome
PolyQ ProteasomePolyQ_Proteasome
AggPolyQ3AggPolyQ3
SourceSource
MisP ProteasomeMisP_Proteasome
AggPolyQ5AggPolyQ5

Tang2019 - Pharmacology modelling of AURKB and ZAK interaction in TNBC: BIOMD0000000940v0.0.1

# Aurora Kinase B and ZAK interaction model Equivalent of the stochastic model used in "Network pharmacology model pred…

Details

Cancer cells with heterogeneous mutation landscapes and extensive functional redundancy easily develop resistance to monotherapies by emerging activation of compensating or bypassing pathways. To achieve more effective and sustained clinical responses, synergistic interactions of multiple druggable targets that inhibit redundant cancer survival pathways are often required. Here, we report a systematic polypharmacology strategy to predict, test, and understand the selective drug combinations for MDA-MB-231 triple-negative breast cancer cells. We started by applying our network pharmacology model to predict synergistic drug combinations. Next, by utilizing kinome-wide drug-target profiles and gene expression data, we pinpointed a synergistic target interaction between Aurora B and ZAK kinase inhibition that led to enhanced growth inhibition and cytotoxicity, as validated by combinatorial siRNA, CRISPR/Cas9, and drug combination experiments. The mechanism of such a context-specific target interaction was elucidated using a dynamic simulation of MDA-MB-231 signaling network, suggesting a cross-talk between p53 and p38 pathways. Our results demonstrate the potential of polypharmacological modeling to systematically interrogate target interactions that may lead to clinically actionable and personalized treatment options. link: http://identifiers.org/pubmed/31312514

Parameters:

NameDescription
k_pten = 0.2Reaction: => PTEN; TP53, Rate Law: Cell*k_pten*TP53
kd_mapk13 = 1.4Reaction: MAPK13 =>, Rate Law: Cell*kd_mapk13*MAPK13
kd_tp53 = 2.0Reaction: TP53 =>, Rate Law: Cell*kd_tp53*TP53
k_prkaca = 2.0Reaction: => PRKACA; AURKB, Rate Law: Cell*k_prkaca*AURKB
k_tp53 = 0.6Reaction: => TP53; SRC, Rate Law: Cell*k_tp53*SRC
k_src = 0.2Reaction: => SRC; CSF1R, Rate Law: Cell*k_src*CSF1R
k_mapk13 = 2.0Reaction: => MAPK13; MAP2K4, Rate Law: Cell*k_mapk13*MAP2K4
kd_src = 1.0Reaction: SRC =>, Rate Law: Cell*kd_src*SRC
kd_parp1 = 0.005Reaction: PARP1 =>, Rate Law: Cell*kd_parp1*PARP1
kd_csf1r = 30.0Reaction: CSF1R =>, Rate Law: Cell*kd_csf1r*CSF1R
k_shc1 = 2.0Reaction: => SHC1; TGFBR1, Rate Law: Cell*k_shc1*TGFBR1
kd_pik3r1 = 10.0Reaction: PIK3R1 => ; PTEN, Rate Law: Cell*kd_pik3r1*PIK3R1*PTEN
kd_aurkb = 4.5Reaction: AURKB =>, Rate Law: Cell*kd_aurkb*AURKB
k_tgfbr1 = 0.5Reaction: => TGFBR1; ZAK, Rate Law: Cell*k_tgfbr1*ZAK
kd_ywhaz = 0.072Reaction: YWHAZ =>, Rate Law: Cell*kd_ywhaz*YWHAZ
kd_brca1 = 20.0Reaction: BRCA1 =>, Rate Law: Cell*kd_brca1*BRCA1
k_ywhaz = 0.9Reaction: => YWHAZ; ZAK, Rate Law: Cell*k_ywhaz*ZAK
kd_bad = 0.04; const=0.0133Reaction: BAD => ; YWHAZ, Rate Law: Cell*kd_bad*BAD*YWHAZ*const
kd_prkaca = 6.0Reaction: PRKACA =>, Rate Law: Cell*kd_prkaca*PRKACA
kd_pten = 0.5Reaction: PTEN =>, Rate Law: Cell*kd_pten*PTEN
k_brca1 = 2.0Reaction: => BRCA1; PARP1, Rate Law: Cell*k_brca1*PARP1
k_atm = 0.1Reaction: => ATM; MAPK14, Rate Law: Cell*k_atm*MAPK14
k_aurkb = 3.0Reaction: => AURKB; PARP1, Rate Law: Cell*k_aurkb*PARP1
kd_map2k4 = 0.6Reaction: MAP2K4 =>, Rate Law: Cell*kd_map2k4*MAP2K4
k_pkn1 = 0.5Reaction: => PKN1, Rate Law: Cell*k_pkn1
k_map2k4 = 0.2Reaction: => MAP2K4; MAP2K3, Rate Law: Cell*k_map2k4*MAP2K3
k_pik3r1 = 2.0Reaction: => PIK3R1; SHC1, Rate Law: Cell*k_pik3r1*SHC1
kd_atm = 3.0Reaction: ATM =>, Rate Law: Cell*kd_atm*ATM
kd_zak = 0.5Reaction: ZAK =>, Rate Law: Cell*kd_zak*ZAK
k_map2k3 = 0.2Reaction: => MAP2K3; PKN1, Rate Law: Cell*k_map2k3*PKN1
kd_tgfbr1 = 0.45Reaction: TGFBR1 =>, Rate Law: Cell*kd_tgfbr1*TGFBR1
k_mapk14 = 2.0Reaction: => MAPK14; MAP2K3, Rate Law: Cell*k_mapk14*MAP2K3
kd_tp53 = 2.0; const=0.0067Reaction: TP53 => ; AURKB, Rate Law: Cell*kd_tp53*TP53*AURKB*const
kd_mapk14 = 5.0Reaction: MAPK14 =>, Rate Law: Cell*kd_mapk14*MAPK14
kd_shc1 = 0.06Reaction: SHC1 => ; PTEN, Rate Law: Cell*kd_shc1*SHC1*PTEN
k_csf1r = 2.0Reaction: => CSF1R; PIK3R1, Rate Law: Cell*k_csf1r*PIK3R1
kd_pkn1 = 0.005Reaction: PKN1 =>, Rate Law: Cell*kd_pkn1*PKN1
k_bad = 5.0Reaction: => BAD, Rate Law: Cell*k_bad
k_zak = 0.1Reaction: => ZAK; PKN1, Rate Law: Cell*k_zak*PKN1
k_parp1 = 0.5Reaction: => PARP1, Rate Law: Cell*k_parp1
kd_map2k3 = 0.6Reaction: MAP2K3 =>, Rate Law: Cell*kd_map2k3*MAP2K3
kd_bad = 0.04Reaction: BAD =>, Rate Law: Cell*kd_bad*BAD

States:

NameDescription
ATM[Serine-protein kinase ATM]
MAPK13[Mitogen-activated protein kinase 13]
SHC1[SHC-transforming protein 1]
TP53[Cellular tumor antigen p53]
SRC[Proto-oncogene tyrosine-protein kinase Src]
PARP1[Poly [ADP-ribose] polymerase 1]
PTEN[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN]
CSF1R[P07333]
BRCA1[P38398]
PKN1[Q16512]
BAD[Bcl2-associated agonist of cell death]
MAP2K3[Dual specificity mitogen-activated protein kinase kinase 3]
MAPK14[Mitogen-activated protein kinase 14]
TGFBR1[TGF-beta receptor type-1]
MAP2K4[Dual specificity mitogen-activated protein kinase kinase 4]
PRKACA[cAMP-dependent protein kinase catalytic subunit alpha]
PIK3R1[Phosphatidylinositol 3-kinase regulatory subunit alpha]
AURKB[Q96GD4]
ZAK[Q75JK0]
YWHAZ[P63104]

Tang2020 - Estimation of transmission risk of COVID-19 and impact of public health interventions: BIOMD0000000971v0.0.1

Since the emergence of the first cases in Wuhan, China, the novel coronavirus (2019-nCoV) infection has been quickly spr…

Details

Since the emergence of the first cases in Wuhan, China, the novel coronavirus (2019-nCoV) infection has been quickly spreading out to other provinces and neighboring countries. Estimation of the basic reproduction number by means of mathematical modeling can be helpful for determining the potential and severity of an outbreak and providing critical information for identifying the type of disease interventions and intensity. A deterministic compartmental model was devised based on the clinical progression of the disease, epidemiological status of the individuals, and intervention measures. The estimations based on likelihood and model analysis show that the control reproduction number may be as high as 6.47 (95% CI 5.71-7.23). Sensitivity analyses show that interventions, such as intensive contact tracing followed by quarantine and isolation, can effectively reduce the control reproduction number and transmission risk, with the effect of travel restriction adopted by Wuhan on 2019-nCoV infection in Beijing being almost equivalent to increasing quarantine by a 100 thousand baseline value. It is essential to assess how the expensive, resource-intensive measures implemented by the Chinese authorities can contribute to the prevention and control of the 2019-nCoV infection, and how long they should be maintained. Under the most restrictive measures, the outbreak is expected to peak within two weeks (since 23 January 2020) with a significant low peak value. With travel restriction (no imported exposed individuals to Beijing), the number of infected individuals in seven days will decrease by 91.14% in Beijing, compared with the scenario of no travel restriction. link: http://identifiers.org/pubmed/32046137

Tang2020 - Estimation of transmission risk of COVID-19 and impact of public health interventions - update: BIOMD0000000972v0.0.1

The basic reproduction number of an infectious agent is the average number of infections one case can generate over the…

Details

The basic reproduction number of an infectious agent is the average number of infections one case can generate over the course of the infectious period, in a naïve, uninfected population. It is well-known that the estimation of this number may vary due to several methodological issues, including different assumptions and choice of parameters, utilized models, used datasets and estimation period. With the spreading of the novel coronavirus (2019-nCoV) infection, the reproduction number has been found to vary, reflecting the dynamics of transmission of the coronavirus outbreak as well as the case reporting rate. Due to significant variations in the control strategies, which have been changing over time, and thanks to the introduction of detection technologies that have been rapidly improved, enabling to shorten the time from infection/symptoms onset to diagnosis, leading to faster confirmation of the new coronavirus cases, our previous estimations on the transmission risk of the 2019-nCoV need to be revised. By using time-dependent contact and diagnose rates, we refit our previously proposed dynamics transmission model to the data available until January 29th, 2020 and re-estimated the effective daily reproduction ratio that better quantifies the evolution of the interventions. We estimated when the effective daily reproduction ratio has fallen below 1 and when the epidemics will peak. Our updated findings suggest that the best measure is persistent and strict self-isolation. The epidemics will continue to grow, and can peak soon with the peak time depending highly on the public health interventions practically implemented. link: http://identifiers.org/pubmed/32099934

Tanos2008 - Blood coagulation and Snake Venom: MODEL1806150001v0.0.1

Mathematical model of the blood coagulation cascade including interaction of snake venom and antivenom.

Details

Many snake venoms contain procoagulant toxins that activate the coagulation cascade and cause venom-induced consumptive coagulopathy (VICC). We developed a semi-mechanistic model of the clotting cascade in order to explore the effects of the procoagulant toxin from taipan venom on this system as well as the effects of antivenom. Simulations of the time course in the change of clotting factors were compared to data collected from taipan envenomed patients. The model accurately predicted the observed concentration of clotting factors over time following taipan envenomation. Investigations from the model indicated that the upper limit of the half-life of the procoagulant toxin was 1h. Simulations from the model also suggest that antivenom for Australasian elapids has negligible effect on reducing the recovery time of the coagulation profile unless administered almost immediately after envenomation. The model has generality to be expanded to describe the effects of other venoms and drugs on the clotting cascade. link: http://identifiers.org/pubmed/18831981

Telesco2011_HER3-ErbB3-RTK_SignalingNetwork: MODEL1102210001v0.0.1

This model is from the article: A multiscale modeling approach to investigate molecular mechanisms of pseudokinase act…

Details

Multiscale modeling provides a powerful and quantitative platform for investigating the complexity inherent in intracellular signaling pathways and rationalizing the effects of molecular perturbations on downstream signaling events and ultimately, on the cell phenotype. Here we describe the application of a multiscale modeling scheme to the HER3/ErbB3 receptor tyrosine kinase (RTK) signaling network, which regulates critical cellular processes including proliferation, migration and differentiation. The HER3 kinase is a topic of current interest and investigation, as it has been implicated in mechanisms of resistance to tyrosine kinase inhibition (TKI) of EGFR and HER2 in the treatment of many human malignancies. Moreover, the commonly regarded status of HER3 as a catalytically inactive 'pseudokinase' has recently been challenged by our previous study, which demonstrated robust residual kinase activity for HER3. Through our multiscale model, we investigate the most significant molecular interactions that contribute to potential mechanisms of HER3 activity and the physiological relevance of this activity to mechanisms of drug resistance in an ErbB-driven tumor cell in silico. The results of our molecular-scale simulations support the characterization of HER3 as a weakly active kinase that, in contrast to its fully-active ErbB family members, depends upon a unique hydrophobic interface to coordinate the alignment of specific catalytic residues required for its activity. Translating our molecular simulation results of the uniquely active behavior of the HER3 kinase into a physiologically relevant environment, our HER3 signaling model demonstrates that even a weak level of HER3 activity may be sufficient to induce AKT signaling and TKI resistance in the context of an ErbB signaling-dependent tumor cell, and therefore therapeutic targeting of HER3 may represent a superior treatment strategy for specific ErbB-driven cancers. link: http://identifiers.org/pubmed/21509365

tenTusscher2004_CardiacArrhythmias: MODEL0393108880v0.0.1

This a model from the article: A model for human ventricular tissue. ten Tusscher KH, Noble D, Noble PJ, Panfilov AV…

Details

The experimental and clinical possibilities for studying cardiac arrhythmias in human ventricular myocardium are very limited. Therefore, the use of alternative methods such as computer simulations is of great importance. In this article we introduce a mathematical model of the action potential of human ventricular cells that, while including a high level of electrophysiological detail, is computationally cost-effective enough to be applied in large-scale spatial simulations for the study of reentrant arrhythmias. The model is based on recent experimental data on most of the major ionic currents: the fast sodium, L-type calcium, transient outward, rapid and slow delayed rectifier, and inward rectifier currents. The model includes a basic calcium dynamics, allowing for the realistic modeling of calcium transients, calcium current inactivation, and the contraction staircase. We are able to reproduce human epicardial, endocardial, and M cell action potentials and show that differences can be explained by differences in the transient outward and slow delayed rectifier currents. Our model reproduces the experimentally observed data on action potential duration restitution, which is an important characteristic for reentrant arrhythmias. The conduction velocity restitution of our model is broader than in other models and agrees better with available data. Finally, we model the dynamics of spiral wave rotation in a two-dimensional sheet of human ventricular tissue and show that the spiral wave follows a complex meandering pattern and has a period of 265 ms. We conclude that the proposed model reproduces a variety of electrophysiological behaviors and provides a basis for studies of reentrant arrhythmias in human ventricular tissue. link: http://identifiers.org/pubmed/14656705

TenTusscher2006_VentricularCellModel: MODEL7910499126v0.0.1

This a model from the article: Alternans and spiral breakup in a human ventricular tissue model. ten Tusscher KH, Pa…

Details

Ventricular fibrillation (VF) is one of the main causes of death in the Western world. According to one hypothesis, the chaotic excitation dynamics during VF are the result of dynamical instabilities in action potential duration (APD) the occurrence of which requires that the slope of the APD restitution curve exceeds 1. Other factors such as electrotonic coupling and cardiac memory also determine whether these instabilities can develop. In this paper we study the conditions for alternans and spiral breakup in human cardiac tissue. Therefore, we develop a new version of our human ventricular cell model, which is based on recent experimental measurements of human APD restitution and includes a more extensive description of intracellular calcium dynamics. We apply this model to study the conditions for electrical instability in single cells, for reentrant waves in a ring of cells, and for reentry in two-dimensional sheets of ventricular tissue. We show that an important determinant for the onset of instability is the recovery dynamics of the fast sodium current. Slower sodium current recovery leads to longer periods of spiral wave rotation and more gradual conduction velocity restitution, both of which suppress restitution-mediated instability. As a result, maximum restitution slopes considerably exceeding 1 (up to 1.5) may be necessary for electrical instability to occur. Although slopes necessary for the onset of instabilities found in our study exceed 1, they are within the range of experimentally measured slopes. Therefore, we conclude that steep APD restitution-mediated instability is a potential mechanism for VF in the human heart. link: http://identifiers.org/pubmed/16565318

Terentyeva2015 - Mechanisms of XII activation: MODEL1808160001v0.0.1

Mathematical model of the mechanism and kinetics of blood coagulation factor XII.

Details

Surface-induced activation of factor XII is critical part of the intrinsic pathway of blood coagulation. The mechanism of this process remains unclear: in particular, it is not known whether the initial amounts of factor XIIa, an active form of factor XII, are produced purely by factor XII contacting a surface or if traces of factor XIIa pre-exist. Furthermore, it is not known whether factor XII first has to bind to a surface before it can interact with the surface-bound factor XIIa in a two-dimensional process to become activated ("bound-substrate model") or if surface-bound factor XIIa activates a fluid-delivered form of factor XII ("free-substrate model"). To investigate these possibilities, we used mathematical modeling to implement various hypotheses. Time courses of factor XII production were generated under different initial conditions and matched with experimental data. We established that only the "bound-substrate model" fits with the majority of experimental data, whereas the "free-substrate model" does not. We also addressed the question of spontaneous activation and found that measurable differences between the models with and without spontaneous activation appear only under limiting conditions (deficit or excess of surface). As there are insufficient data regarding the system's behavior upon such variations of surface concentration in the literature, we designed new experiments to answer this question. link: http://identifiers.org/pubmed/26187095

Terkildsen2008_CardiomyocyteFunction: MODEL7893871775v0.0.1

This a model from the article: Using Physiome standards to couple cellular functions for rat cardiac excitation-contra…

Details

Scientific endeavour is reliant upon the extension and reuse of previous knowledge. The formalization of this process for computational modelling is facilitated by the use of accepted standards with which to describe and simulate models, ensuring consistency between the models and thus reducing the development and propagation of errors. CellML 1.1, an XML-based programming language, has been designed as a modelling standard which, by virtue of its import and grouping functions, facilitates model combination and reuse. Using CellML 1.1, we demonstrate the process of formalized model reuse by combining three separate models of rat cardiomyocyte function (an electrophysiology model, a model of cellular calcium dynamics and a mechanics model) which together make up the Pandit-Hinch-Niederer et al. cell model. Not only is this integrative model of rat electromechanics a useful tool for cardiac modelling but it is also an ideal framework with which to demonstrate both the power of model reuse and the challenges associated with this process. We highlight and classify a number of these issues associated with combining models and provide some suggested solutions. link: http://identifiers.org/pubmed/18344258

Teusink1998_Glycolysis_TurboDesign: BIOMD0000000253v0.0.1

This is the model described in the article: The danger of metabolic pathways with turbo design Teusink B, Walsh MC,…

Details

Many catabolic pathways begin with an ATP-requiring activation step, after which further metabolism yields a surplus of ATP. Such a 'turbo' principle is useful but also contains an inherent risk. This is illustrated by a detailed kinetic analysis of a paradoxical Saccharomyces cerevisiae mutant; the mutant fails to grow on glucose because of overactive initial enzymes of glycolysis, but is defective only in an enzyme (trehalose 6-phosphate synthase) that appears to have little relevance to glycolysis. The ubiquity of pathways that possess an initial activation step, suggests that there might be many more genes that, when deleted, cause rather paradoxical regulation phenotypes (i.e. growth defects caused by enhanced utilization of growth substrate). link: http://identifiers.org/pubmed/9612078

Parameters:

NameDescription
KFru16P2=1.0 mM; KADP=0.1 mM; Vlower=20.0 mM per minReaction: Fru16P2 + ADP => ATP, Rate Law: cell*Vlower*Fru16P2*ADP/(KFru16P2*KADP)/((1+Fru16P2/KFru16P2)*(1+ADP/KADP))
VATPase=68.0 mM per min; KATP=3.0 mMReaction: ATP => ADP, Rate Law: cell*VATPase*ATP/(KATP+ATP)
wild_type=1.0 dimensionless; VHK=68.0 mM per min; KATP=0.15 mM; KGlc=1.0 mM; KiTre6P=4.422 mMReaction: Glc + ATP => HMP; Tre6P, Rate Law: cell*VHK*Glc*ATP/(KGlc*KATP)/((1+Glc/KGlc+wild_type*Tre6P/KiTre6P)*(1+ATP/KATP))
L = NaN; gR = 10.0; lambda1 = NaN; lambda2 = NaN; T = NaN; VPFK=30.0 mM per min; R = NaNReaction: HMP + ATP => Fru16P2, Rate Law: cell*VPFK*gR*lambda1*lambda2*R/(R^2+L*T^2)

States:

NameDescription
ATP[ATP]
Tre6P[alpha,alpha-trehalose 6-phosphate]
HMP[beta-D-fructofuranose 6-phosphate; alpha-D-glucose 6-phosphate]
Fru16P2[keto-D-fructose 1,6-bisphosphate]
Glc[alpha-D-glucose]
ADP[ADP]

Teusink2000_Glycolysis: BIOMD0000000064v0.0.1

**Can yeast glycolysis be understood in terms of in vitro kinetics of the constituent enzymes? Testing biochemistry.**…

Details

This paper examines whether the in vivo behavior of yeast glycolysis can be understood in terms of the in vitro kinetic properties of the constituent enzymes. In nongrowing, anaerobic, compressed Saccharomyces cerevisiae the values of the kinetic parameters of most glycolytic enzymes were determined. For the other enzymes appropriate literature values were collected. By inserting these values into a kinetic model for glycolysis, fluxes and metabolites were calculated. Under the same conditions fluxes and metabolite levels were measured. In our first model, branch reactions were ignored. This model failed to reach the stable steady state that was observed in the experimental flux measurements. Introduction of branches towards trehalose, glycogen, glycerol and succinate did allow such a steady state. The predictions of this branched model were compared with the empirical behavior. Half of the enzymes matched their predicted flux in vivo within a factor of 2. For the other enzymes it was calculated what deviation between in vivo and in vitro kinetic characteristics could explain the discrepancy between in vitro rate and in vivo flux. link: http://identifiers.org/pubmed/10951190

Parameters:

NameDescription
KTREHALOSE=2.4 mMperminReaction: G6P + P => Trh, Rate Law: cytosol*KTREHALOSE
KeqGLT=1.0 mM; KmGLTGLCo=1.1918 mM; VmGLT=97.264 mmolepermin; KmGLTGLCi=1.1918 mMReaction: GLCo => GLCi, Rate Law: VmGLT/KmGLTGLCo*(GLCo-GLCi/KeqGLT)/(1+GLCo/KmGLTGLCo+GLCi/KmGLTGLCi+0.91*GLCo*GLCi/(KmGLTGLCo*KmGLTGLCi))
KSUCC=21.4Reaction: ACE + NAD + P => NADH + SUCC, Rate Law: cytosol*KSUCC*ACE
KmG3PDHGLY=1.0 mM; KeqG3PDH=4300.0 dimensionless; KmG3PDHDHAP=0.4 mM; KmG3PDHNADH=0.023 mM; KeqTPI = 0.045 dimensionless; KmG3PDHNAD=0.93 mM; VmG3PDH=70.15 mMperminReaction: TRIO + NADH => NAD + GLY, Rate Law: cytosol*VmG3PDH/(KmG3PDHDHAP*KmG3PDHNADH)*(1/(1+KeqTPI)*TRIO*NADH-GLY*NAD/KeqG3PDH)/((1+1/(1+KeqTPI)*TRIO/KmG3PDHDHAP+GLY/KmG3PDHGLY)*(1+NADH/KmG3PDHNADH+NAD/KmG3PDHNAD))
KmPGIG6P_2=1.4 mM; VmPGI_2=339.677 mMpermin; KmPGIF6P_2=0.3 mM; KeqPGI_2=0.314 dimensionlessReaction: G6P => F6P, Rate Law: cytosol*VmPGI_2/KmPGIG6P_2*(G6P-F6P/KeqPGI_2)/(1+G6P/KmPGIG6P_2+F6P/KmPGIF6P_2)
KmGLKADP=0.23 mM; KmGLKGLCi=0.08 mM; VmGLK=226.452 mMpermin; KmGLKG6P=30.0 mM; KeqGLK=3800.0 dimensionless; KmGLKATP=0.15 mMReaction: GLCi + P => G6P; ATP, ADP, Rate Law: cytosol*VmGLK/(KmGLKGLCi*KmGLKATP)*(GLCi*ATP-G6P*ADP/KeqGLK)/((1+GLCi/KmGLKGLCi+G6P/KmGLKG6P)*(1+ATP/KmGLKATP+ADP/KmGLKADP))
KATPASE=33.7 perminReaction: P => ; ATP, Rate Law: cytosol*KATPASE*ATP
KPFKF26BP = 6.82E-4 mM; KmPFKF6P = 0.1 mM; CiPFKATP = 100.0 dimensionless; CPFKATP = 3.0 dimensionless; CPFKAMP = 0.0845 dimensionless; KPFKF16BP = 0.111 mM; Lzero = 0.66 dimensionless; VmPFK=182.903 mMpermin; CPFKF26BP = 0.0174 dimensionless; CPFKF16BP = 0.397 dimensionless; KPFKAMP = 0.0995 mM; gR = 5.12 dimensionless; KiPFKATP = 0.65 mM; KmPFKATP = 0.71 mMReaction: F6P + P => F16P; AMP, ATP, F26BP, Rate Law: cytosol*VmPFK*gR*F6P/KmPFKF6P*ATP/KmPFKATP*R_PFK(KmPFKF6P, KmPFKATP, gR, ATP, F6P)/(R_PFK(KmPFKF6P, KmPFKATP, gR, ATP, F6P)^2+L_PFK(Lzero, CiPFKATP, KiPFKATP, CPFKAMP, KPFKAMP, CPFKF26BP, KPFKF26BP, CPFKF16BP, KPFKF16BP, ATP, AMP, F16P, F26BP)*T_PFK(CPFKATP, KmPFKATP, ATP)^2)
VmGAPDHf=1184.52 mMpermin; VmGAPDHr=6549.8 mMpermin; KmGAPDHNAD=0.09 mM; KmGAPDHBPG=0.0098 mM; KmGAPDHGAP=0.21 mM; KeqTPI = 0.045 dimensionless; KmGAPDHNADH=0.06 mMReaction: TRIO + NAD => BPG + NADH, Rate Law: cytosol*(VmGAPDHf*KeqTPI/(1+KeqTPI)*TRIO*NAD/(KmGAPDHGAP*KmGAPDHNAD)-VmGAPDHr*BPG*NADH/(KmGAPDHBPG*KmGAPDHNADH))/((1+KeqTPI/(1+KeqTPI)*TRIO/KmGAPDHGAP+BPG/KmGAPDHBPG)*(1+NAD/KmGAPDHNAD+NADH/KmGAPDHNADH))
KeqENO=6.7 dimensionless; KmENOP2G=0.04 mM; KmENOPEP=0.5 mM; VmENO=365.806 mMperminReaction: P2G => PEP, Rate Law: cytosol*VmENO/KmENOP2G*(P2G-PEP/KeqENO)/(1+P2G/KmENOP2G+PEP/KmENOPEP)
KmPGMP3G=1.2 mM; KeqPGM=0.19 dimensionless; VmPGM=2525.81 mMpermin; KmPGMP2G=0.08 mMReaction: P3G => P2G, Rate Law: cytosol*VmPGM/KmPGMP3G*(P3G-P2G/KeqPGM)/(1+P3G/KmPGMP3G+P2G/KmPGMP2G)
KGLYCOGEN_3=6.0 mMperminReaction: G6P + P => Glyc, Rate Law: cytosol*KGLYCOGEN_3
KmPGKBPG=0.003 mM; KmPGKATP=0.3 mM; KeqPGK=3200.0 dimensionless; VmPGK=1306.45 mMpermin; KmPGKP3G=0.53 mM; KmPGKADP=0.2 mMReaction: BPG => P3G + P; ATP, ADP, Rate Law: cytosol*VmPGK/(KmPGKP3G*KmPGKATP)*(KeqPGK*BPG*ADP-P3G*ATP)/((1+BPG/KmPGKBPG+P3G/KmPGKP3G)*(1+ATP/KmPGKATP+ADP/KmPGKADP))
KmALDGAP=2.0 mM; VmALD=322.258 mMpermin; KeqALD=0.069 dimensionless; KmALDDHAP=2.4 mM; KeqTPI = 0.045 dimensionless; KmALDGAPi=10.0 mM; KmALDF16P=0.3 mMReaction: F16P => TRIO, Rate Law: cytosol*VmALD/KmALDF16P*(F16P-KeqTPI/(1+KeqTPI)*TRIO*1/(1+KeqTPI)*TRIO/KeqALD)/(1+F16P/KmALDF16P+KeqTPI/(1+KeqTPI)*TRIO/KmALDGAP+1/(1+KeqTPI)*TRIO/KmALDDHAP+KeqTPI/(1+KeqTPI)*TRIO*1/(1+KeqTPI)*TRIO/(KmALDGAP*KmALDDHAP)+F16P*KeqTPI/(1+KeqTPI)*TRIO/(KmALDGAPi*KmALDF16P))
KeqAK = 0.45 dimensionlessReaction: ADP = (SUM_P-(P^2*(1-4*KeqAK)+2*SUM_P*P*(4*KeqAK-1)+SUM_P^2)^0.5)/(1-4*KeqAK), Rate Law: missing
VmPDC=174.194 mMpermin; KmPDCPYR=4.33 mM; nPDC=1.9 dimensionlessReaction: PYR => ACE + CO2, Rate Law: cytosol*VmPDC*PYR^nPDC/KmPDCPYR^nPDC/(1+PYR^nPDC/KmPDCPYR^nPDC)
KmADHNAD=0.17 mM; KiADHETOH=90.0 mM; KiADHNADH=0.031 mM; KiADHACE=1.1 mM; KmADHETOH=17.0 mM; KeqADH=6.9E-5 dimensionless; KmADHNADH=0.11 mM; KiADHNAD=0.92 mM; VmADH=810.0 mMpermin; KmADHACE=1.11 mMReaction: ACE + NADH => NAD + ETOH, Rate Law: (-cytosol)*VmADH/(KiADHNAD*KmADHETOH)*(NAD*ETOH-NADH*ACE/KeqADH)/(1+NAD/KiADHNAD+KmADHNAD*ETOH/(KiADHNAD*KmADHETOH)+KmADHNADH*ACE/(KiADHNADH*KmADHACE)+NADH/KiADHNADH+NAD*ETOH/(KiADHNAD*KmADHETOH)+KmADHNADH*NAD*ACE/(KiADHNAD*KiADHNADH*KmADHACE)+KmADHNAD*ETOH*NADH/(KiADHNAD*KmADHETOH*KiADHNADH)+NADH*ACE/(KiADHNADH*KmADHACE)+NAD*ETOH*ACE/(KiADHNAD*KmADHETOH*KiADHACE)+ETOH*NADH*ACE/(KiADHETOH*KiADHNADH*KmADHACE))
VmPYK=1088.71 mMpermin; KmPYKATP=1.5 mM; KeqPYK=6500.0 dimensionless; KmPYKPYR=21.0 mM; KmPYKADP=0.53 mM; KmPYKPEP=0.14 mMReaction: PEP => PYR + P; ATP, ADP, Rate Law: cytosol*VmPYK/(KmPYKPEP*KmPYKADP)*(PEP*ADP-PYR*ATP/KeqPYK)/((1+PEP/KmPYKPEP+PYR/KmPYKPYR)*(1+ATP/KmPYKATP+ADP/KmPYKADP))

States:

NameDescription
ATP[ATP; ATP]
P[ADP; ATP; ADP; ADP; ATP]
Trh[trehalose; alpha,alpha-Trehalose]
GLY[glycerol; Glycerol]
AMP[AMP; AMP]
F16P[keto-D-fructose 1,6-bisphosphate; D-Fructose 1,6-bisphosphate]
GLCi[glucose; C00293]
P2G[2-phospho-D-glyceric acid; 2-Phospho-D-glycerate]
P3G[3-phospho-D-glyceric acid; 3-Phospho-D-glycerate]
GLCo[glucose; C00293]
NADH[NADH; NADH]
SUCC[succinate(2-)]
PYR[pyruvate; Pyruvate; pyruvic acid]
TRIO[dihydroxyacetone phosphate; D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate; Glycerone phosphate; D-glyceraldehyde 3-phosphate]
Glyc[glycogen; Glycogen]
F6P[keto-D-fructose 6-phosphate; beta-D-Fructose 6-phosphate]
CO2[carbon dioxide; CO2]
BPG[3-phospho-D-glyceroyl dihydrogen phosphate; 3-Phospho-D-glyceroyl phosphate]
G6P[alpha-D-glucose 6-phosphate; alpha-D-Glucose 6-phosphate]
PEP[Phosphoenolpyruvate; phosphoenolpyruvate; phosphoenolpyruvate]
NAD[NAD(+); NAD+]
ETOH[ethanol; Ethanol]
ADP[ADP; ADP]
ACE[acetaldehyde; Acetaldehyde]

Teusink2006 - Genome-scale metabolic network of Lactobacillus plantarum (iBT721): MODEL1507180045v0.0.1

Teusink2006 - Genome-scale metabolic network of Lactobacillus plantarum (iBT721)This model is described in the article:…

Details

A genome-scale metabolic model of the lactic acid bacterium Lactobacillus plantarum WCFS1 was constructed based on genomic content and experimental data. The complete model includes 721 genes, 643 reactions, and 531 metabolites. Different stoichiometric modeling techniques were used for interpretation of complex fermentation data, as L. plantarum is adapted to nutrient-rich environments and only grows in media supplemented with vitamins and amino acids. (i) Based on experimental input and output fluxes, maximal ATP production was estimated and related to growth rate. (ii) Optimization of ATP production further identified amino acid catabolic pathways that were not previously associated with free-energy metabolism. (iii) Genome-scale elementary flux mode analysis identified 28 potential futile cycles. (iv) Flux variability analysis supplemented the elementary mode analysis in identifying parallel pathways, e.g. pathways with identical end products but different co-factor usage. Strongly increased flexibility in the metabolic network was observed when strict coupling between catabolic ATP production and anabolic consumption was relaxed. These results illustrate how a genome-scale metabolic model and associated constraint-based modeling techniques can be used to analyze the physiology of growth on a complex medium rather than a minimal salts medium. However, optimization of biomass formation using the Flux Balance Analysis approach, reported to successfully predict growth rate and by product formation in Escherichia coli and Saccharomyces cerevisiae, predicted too high biomass yields that were incompatible with the observed lactate production. The reason is that this approach assumes optimal efficiency of substrate to biomass conversion, and can therefore not predict the metabolically inefficient lactate formation. link: http://identifiers.org/pubmed/17062565

Tham2008 - PDmodel, Tumour shrinkage by gemcitabine and carboplatin: BIOMD0000000234v0.0.1

Tham2008 - PDmodel, Tumour shrinkage by gemcitabine and carboplatin This model is described in the article: [A pharmaco…

Details

PURPOSE: This tumor response pharmacodynamic model aims to describe primary lesion shrinkage in non-small cell lung cancer over time and determine if concentration-based exposure metrics for gemcitabine or that of its metabolites, 2',2'-difluorodeoxyuridine or gemcitabine triphosphate, are better than gemcitabine dose for prediction of individual response. EXPERIMENTAL DESIGN: Gemcitabine was given thrice weekly on days 1 and 8 in combination with carboplatin, which was given only on day 1 of every cycle. Gemcitabine amount in the body and area under the concentration-time curves of plasma gemcitabine, 2',2'-difluorodeoxyuridine, and intracellular gemcitabine triphosphate in white cells were compared to determine which best describes tumor shrinkage over time. Tumor growth kinetics were described using a Gompertz-like model. RESULTS: The apparent half-life for the effect of gemcitabine was 7.67 weeks. The tumor turnover time constant was 21.8 week.cm. Baseline tumor size and gemcitabine amount in the body to attain 50% of tumor shrinkage were estimated to be 6.66 cm and 10,600 mg. There was no evidence of relapse during treatment. CONCLUSIONS: Concentration-based exposure metrics for gemcitabine and its metabolites were no better than gemcitabine amount in predicting tumor shrinkage in primary lung cancer lesions. Gemcitabine dose-based models did marginally better than treatment-based models that ignored doses of drug administered to patients. Modeling tumor shrinkage in primary lesions can be used to quantify individual sensitivity and response to antitumor effects of anticancer drugs. link: http://identifiers.org/pubmed/18594002

Parameters:

NameDescription
Keq = NaN per_week; Exposure = NaN mgReaction: Ce = Exposure/1-Ce*Keq, Rate Law: Exposure/1-Ce*Keq

States:

NameDescription
CeCe

Theinmozhi2018 - Mechanism of PD1 inhibiting TCR signaling in Tumor immune regulation: BIOMD0000000724v0.0.1

Its a Deterministic ODE model showcasing mechanism of PDL1 induced TCR and CD38 signalling inhibition. The model also co…

Details

Programmed cell death-1 (PD-1) is an inhibitory immune checkpoint receptor that negatively regulates the functioning of T cell. Although the direct targets of PD-1 were not identified, its inhibitory action on the TCR signaling pathway was known much earlier. Recent experiments suggest that the PD-1 inhibits the TCR and CD28 signaling pathways at a very early stage ─ at the level of phosphorylation of the cytoplasmic domain of TCR and CD28 receptors. Here, we develop a mathematical model to investigate the influence of inhibitory effect of PD-1 on the activation of early TCR and CD28 signaling molecules. Proposed model recaptures several quantitative experimental observations of PD-1 mediated inhibition. Model simulations show that PD-1 imposes a net inhibitory effect on the Lck kinase. Further, the inhibitory effect of PD-1 on the activation of TCR signaling molecules such as Zap70 and SLP76 is significantly enhanced by the PD-1 mediated inhibition of Lck. These results suggest a critical role for Lck as a mediator for PD-1 induced inhibition of TCR signaling network. Multi parametric sensitivity analysis explores the effect of parameter uncertainty on model simulations. link: http://identifiers.org/pubmed/30356330

Parameters:

NameDescription
Kpa_i = 1.0E-6 1/sReaction: LCKi => LCKya, Rate Law: Cell*Kpa_i*LCKi
Kpi_i = 6.0E-7 1/sReaction: LCKi => LCKyi, Rate Law: Cell*Kpi_i*LCKi
Kdpi_yi = 0.0 l/(nmol*s)Reaction: LCKyi => LCKi; CPactive, Rate Law: Cell*Kdpi_yi*CPactive*LCKyi
Kd_slp = 0.12 1/s; Ka_slp = 0.015 l/(nmol*s)Reaction: GADSa + SLP76 => SLP76i, Rate Law: Cell*(Ka_slp*GADSa*SLP76-Kd_slp*SLP76i)
Ka_shp = 0.0065 l/(nmol*s); Kd1_shp = 10.0 1/sReaction: PD1p1 + SHP2 => CP1, Rate Law: Cell*(Ka_shp*PD1p1*SHP2-Kd1_shp*CP1)
Ka_zap = 7.0E-5 l/(nmol*s); Kd_zap = 0.001 1/sReaction: CD3a + ZAP70 => ZAP70i, Rate Law: Cell*(Ka_zap*CD3a*ZAP70-Kd_zap*ZAP70i)
Ka_gads = 5.0E-4 l/(nmol*s); Kd_gads = 1.5 1/sReaction: LATa + GADS => GADSa, Rate Law: Cell*(Ka_gads*LATa*GADS-Kd_gads*GADSa)
Kd2_shp = 1.0 1/sReaction: CP2 => SHP2 + PD1p1, Rate Law: Cell*Kd2_shp*CP2
Kdp_cd28 = 5.0 1/s; KMdp_cd28 = 500.0 nmol/lReaction: CD28a => CD28i; CPactive, Rate Law: Cell*Kdp_cd28*CPactive*CD28a/(KMdp_cd28+CD28a)
Kdpi_yiya = 0.0 l/(nmol*s)Reaction: LCKyiya => LCKya; CPactive, Rate Law: Cell*Kdpi_yiya*CPactive*LCKyiya
Kdpa_pi = 0.0 l/(nmol*s)Reaction: LCKpi => LCKyi; CPactive, Rate Law: Cell*Kdpa_pi*CPactive*LCKpi
KMp_pd1 = 1000.0 nmol/l; Kp_pd1 = 7.5 1/sReaction: PD1p1 => PD1p2; LCKactive, Rate Law: Cell*Kp_pd1*LCKactive*PD1p1/(KMp_pd1+PD1p1)
Kp_slp = 0.003 l/(nmol*s)Reaction: SLP76i => SLP76a; ZAP70a2, Rate Law: Cell*Kp_slp*ZAP70a2*SLP76i
Kp2_zap = 3.0E-5 l/(nmol*s)Reaction: ZAP70a1 => ZAP70a2; LCKactive, Rate Law: Cell*Kp2_zap*LCKactive*ZAP70a1
Kp_cd28 = 1.0 1/s; KMp_cd28 = 1000.0 nmol/lReaction: CD28i => CD28a; LCKactive, Rate Law: Cell*Kp_cd28*LCKactive*CD28i/(KMp_cd28+CD28i)
Kpa_yi = 7.5E-4 1/sReaction: LCKyi => LCKpi, Rate Law: Cell*Kpa_yi*LCKyi
KMp_cd3 = 80.0 nmol/l; Kp_cd3 = 3.29 1/sReaction: CD3i => CD3a; LCKactive, Rate Law: Cell*Kp_cd3*LCKactive*CD3i/(KMp_cd3+CD3i)
KMdp_cd3 = 150.0 nmol/l; Kdp_cd3 = 5.0 1/sReaction: CD3a => CD3i; CPactive, Rate Law: Cell*Kdp_cd3*CPactive*CD3a/(KMdp_cd3+CD3a)
Kdp_cp2 = 5.0E-8 1/sReaction: CP2 => CP1, Rate Law: Cell*Kdp_cp2*CP2
Kpi_ya = 6.0E-5 1/sReaction: LCKya => LCKyiya, Rate Law: Cell*Kpi_ya*LCKya
KMp_pd1 = 1000.0 nmol/l; Kp_pd1 = 7.5 1/s; k = 41.0Reaction: PD1 => PD1p1; LCKactive, PD1p1, PD1p2, LCKt, Rate Law: Cell*Kp_pd1*LCKactive*PD1/(KMp_pd1+PD1)*(1-(PD1p1+PD1p2)/(LCKt*k))
Kp1_zap = 2.0E-6 l/(nmol*s)Reaction: ZAP70i => ZAP70a1; LCKactive, Rate Law: Cell*Kp1_zap*LCKactive*ZAP70i
Kdpa_ya = 0.0 l/(nmol*s)Reaction: LCKya => LCKi; CPactive, Rate Law: Cell*Kdpa_ya*CPactive*LCKya
Ka_pi3k = 1.4E-6 l/(nmol*s); Kd_pi3k = 9.0E-4 1/sReaction: CD28a + PI3K => PI3Kb, Rate Law: Cell*(Ka_pi3k*CD28a*PI3K-Kd_pi3k*PI3Kb)
Kp_lat = 0.001 l/(nmol*s)Reaction: LATi => LATa; ZAP70a2, Rate Law: Cell*Kp_lat*ZAP70a2*LATi

States:

NameDescription
CD3a[T-cell surface glycoprotein CD3 zeta chain; 0002220]
SHP2[Tyrosine-protein phosphatase non-receptor type 11]
PD1p1[Programmed cell death 1 ligand 1; 0002220]
LCKinactive[Tyrosine-protein kinase Lck; 0002355]
CP1[Tyrosine-protein phosphatase non-receptor type 11; Programmed cell death 1 ligand 1; 0002220]
SLP76[Lymphocyte cytosolic protein 2]
GADSt[GRB2-related adapter protein; Growth factor receptor-bound protein 2]
CD3t[T-cell surface glycoprotein CD3 zeta chain]
CD28a[T-cell-specific surface glycoprotein CD28; 0002220]
CD28i[T-cell-specific surface glycoprotein CD28; 0002355]
GADSa[Linker for activation of T-cells family member 1; Growth factor receptor-bound protein 2; GRB2-related adapter protein]
ZAP70[Tyrosine-protein kinase ZAP-70]
PI3Kt[Phosphatidylinositol 3-kinase regulatory subunit alpha; Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform]
ZAP70t[Tyrosine-protein kinase ZAP-70]
SLP76t[Lymphocyte cytosolic protein 2]
LCKpi[Tyrosine-protein kinase Lck; 0002220]
ZAP70a1[Tyrosine-protein kinase ZAP-70; 0002220]
LATa[Linker for activation of T-cells family member 1; 0002220]
LCKya[Tyrosine-protein kinase Lck; 0002220]
GADS[Growth factor receptor-bound protein 2; GRB2-related adapter protein]
PI3K[Phosphatidylinositol 3-kinase regulatory subunit alpha; Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform]
SLP76i[Lymphocyte cytosolic protein 2]
CD28t[T-cell-specific surface glycoprotein CD28]
LCKactive[Tyrosine-protein kinase Lck; 0002220]
LCKyiya[Tyrosine-protein kinase Lck; 0002220]
CPactive[Tyrosine-protein phosphatase non-receptor type 11; Programmed cell death 1 ligand 1; 0002220]
LCKyi[Tyrosine-protein kinase Lck; 0002355]
SLP76a[Lymphocyte cytosolic protein 2]
LCKt[Tyrosine-protein kinase Lck]
PD1p2[Programmed cell death 1 ligand 1; 0002220]
LATt[Linker for activation of T-cells family member 1]
ZAP70i[T-cell surface glycoprotein CD3 zeta chain; Tyrosine-protein kinase ZAP-70; 0002355]
ZAP70a2[Tyrosine-protein kinase ZAP-70; 0002220]
CD3i[T-cell surface glycoprotein CD3 zeta chain; 0002355]
LATi[Linker for activation of T-cells family member 1; 0002355]
PI3Kb[Phosphatidylinositol 3-kinase regulatory subunit alpha; Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform; 0002220]
CP2[Programmed cell death 1 ligand 1; Tyrosine-protein phosphatase non-receptor type 11; 0002220]
PD1[Programmed cell death 1 ligand 1; 0002355]
LCKi[Tyrosine-protein kinase Lck; 0002355]

Thiaville2016 - Folate pathway model (PanB overexpression and THF regulation): BIOMD0000000690v0.0.1

Henry2016 Folate pathway model with induced PanB reactionThis model is described in the article: [Experimental and Meta…

Details

Tetrahydrofolate (THF) and its one-carbon derivatives, collectively termed folates, are essential cofactors, but are inherently unstable. While it is clear that chemical oxidation can cleave folates or damage their pterin precursors, very little is known about enzymatic damage to these molecules or about whether the folate biosynthesis pathway responds adaptively to damage to its end-products. The presence of a duplication of the gene encoding the folate biosynthesis enzyme 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase (FolK) in many sequenced bacterial genomes combined with a strong chromosomal clustering of the folK gene with panB, encoding the 5,10-methylene-THF-dependent enzyme ketopantoate hydroxymethyltransferase, led us to infer that PanB has a side activity that cleaves 5,10-methylene-THF, yielding a pterin product that is recycled by FolK. Genetic and metabolic analyses of Escherichia coli strains showed that overexpression of PanB leads to accumulation of the likely folate cleavage product 6-hydroxymethylpterin and other pterins in cells and medium, and-unexpectedly-to a 46% increase in total folate content. In silico modeling of the folate biosynthesis pathway showed that these observations are consistent with the in vivo cleavage of 5,10-methylene-THF by a side-activity of PanB, with FolK-mediated recycling of the pterin cleavage product, and with regulation of folate biosynthesis by folates or their damage products. link: http://identifiers.org/pubmed/27065985

Parameters:

NameDescription
k1=10029.0Reaction: L_Glutamate + ATP + H2_Pteroate => DHF + ADP + Phosphate, Rate Law: compartment*k1*L_Glutamate*ATP*H2_Pteroate
k1=8000.0Reaction: p_ABA + H2_HMPterinPP => PPi + H2_Pteroate, Rate Law: compartment*k1*p_ABA*H2_HMPterinPP
v=2.35E-7Reaction: => H2_HMPt, Rate Law: compartment*v
k1=0.008Reaction: CH2_THF => H2_HMPt + p_ABA, Rate Law: compartment*k1*CH2_THF
k1=86170.0Reaction: DHF + NADPH => NADP + THF, Rate Law: compartment*k1*DHF*NADPH
k1=4080.0; k2=2000.0Reaction: THF + L_serine => CH2_THF + Glycine, Rate Law: compartment*(k1*THF*L_serine-k2*CH2_THF*Glycine)
Km=5.921E-5; V=1.726E-7Reaction: CH2_THF =>, Rate Law: compartment*V*CH2_THF/(Km+CH2_THF)
k1=24.8Reaction: ATP + H2_HMPt => AMP + H2_HMPterinPP, Rate Law: compartment*k1*ATP*H2_HMPt
V=1.243E-7; Km=1.571E-4Reaction: THF =>, Rate Law: compartment*V*THF/(Km+THF)

States:

NameDescription
H2 Pteroate[Dihydropteroate; pteroate]
ATP[ATP; ATP]
NADP[NADP; NADP+]
THF[tetrahydrofolate; Tetrahydrofolate]
AMP[AMP; AMP]
DHF[Dihydrofolate; dihydrofolate(2-)]
Phosphate[phosphate]
H2 HMPterinPP[6-(Hydroxymethyl)-7,8-dihydropterin; phosphorylation; dihydropterin]
NADPH[NADPH; NADPH]
L Glutamate[L-Glutamate; glutamate(2-)]
Glycine[glycine; Glycine]
ADP[ADP; ADP]
PPi[Diphosphate]
CH2 THF[5,10-methylenetetrahydrofolate(2-); 5,10-Methylenetetrahydrofolate]
L serine[serine; L-Serine]
H2 HMPt[dihydropterin; 6-(Hydroxymethyl)-7,8-dihydropterin]
p ABA[4-Aminobenzoate; 4-aminobenzoate]

Thiaville2016 - Folate pathway model (PanB overexpression): BIOMD0000000689v0.0.1

Henry2016 Folate pathway model with induced PanB reactionThis model is described in the article: [Experimental and Meta…

Details

Tetrahydrofolate (THF) and its one-carbon derivatives, collectively termed folates, are essential cofactors, but are inherently unstable. While it is clear that chemical oxidation can cleave folates or damage their pterin precursors, very little is known about enzymatic damage to these molecules or about whether the folate biosynthesis pathway responds adaptively to damage to its end-products. The presence of a duplication of the gene encoding the folate biosynthesis enzyme 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase (FolK) in many sequenced bacterial genomes combined with a strong chromosomal clustering of the folK gene with panB, encoding the 5,10-methylene-THF-dependent enzyme ketopantoate hydroxymethyltransferase, led us to infer that PanB has a side activity that cleaves 5,10-methylene-THF, yielding a pterin product that is recycled by FolK. Genetic and metabolic analyses of Escherichia coli strains showed that overexpression of PanB leads to accumulation of the likely folate cleavage product 6-hydroxymethylpterin and other pterins in cells and medium, and-unexpectedly-to a 46% increase in total folate content. In silico modeling of the folate biosynthesis pathway showed that these observations are consistent with the in vivo cleavage of 5,10-methylene-THF by a side-activity of PanB, with FolK-mediated recycling of the pterin cleavage product, and with regulation of folate biosynthesis by folates or their damage products. link: http://identifiers.org/pubmed/27065985

Parameters:

NameDescription
k1=0.0121Reaction: CH2_THF => H2_HMPt + p_ABA, Rate Law: compartment*k1*CH2_THF
k1=3602.18Reaction: L_Glutamate + ATP + H2_Pteroate => DHF + ADP + Phosphate, Rate Law: compartment*k1*L_Glutamate*ATP*H2_Pteroate
k1=4000.0Reaction: p_ABA + H2_HMPterinPP => PPi + H2_Pteroate, Rate Law: compartment*k1*p_ABA*H2_HMPterinPP
k1=31170.0Reaction: DHF + NADPH => NADP + THF, Rate Law: compartment*k1*DHF*NADPH
k1=4080.0; k2=2000.0Reaction: THF + L_serine => CH2_THF + Glycine, Rate Law: compartment*(k1*THF*L_serine-k2*CH2_THF*Glycine)
v=1.66E-7Reaction: => p_ABA, Rate Law: compartment*v
Km=5.921E-5; V=1.726E-7Reaction: CH2_THF =>, Rate Law: compartment*V*CH2_THF/(Km+CH2_THF)
k1=15.8Reaction: ATP + H2_HMPt => AMP + H2_HMPterinPP, Rate Law: compartment*k1*ATP*H2_HMPt
V=1.243E-7; Km=1.571E-4Reaction: THF =>, Rate Law: compartment*V*THF/(Km+THF)

States:

NameDescription
H2 Pteroate[pteroate; Dihydropteroate]
ATP[ATP; ATP]
NADP[NADP+; NADP]
AMP[AMP; AMP]
THF[Tetrahydrofolate; tetrahydrofolate]
DHF[dihydrofolate(2-); Dihydrofolate]
Phosphate[phosphate]
H2 HMPterinPP[6-(Hydroxymethyl)-7,8-dihydropterin; phosphorylation; dihydropterin]
NADPH[NADPH; NADPH]
L Glutamate[L-Glutamate; glutamate(2-)]
Glycine[Glycine; glycine]
ADP[ADP; ADP]
PPi[Diphosphate]
CH2 THF[5,10-Methylenetetrahydrofolate; 5,10-methylenetetrahydrofolate(2-)]
L serine[serine; L-Serine]
H2 HMPt[6-(Hydroxymethyl)-7,8-dihydropterin; dihydropterin]
p ABA[4-aminobenzoate; 4-Aminobenzoate]

Thiaville2016 - Wild type folate pathway model with proposed PanB reaction: BIOMD0000000639v0.0.1

Thiaville2016 - Wild type folate pathway model with proposed PanB reactionThis is a wild type E. coli model, and is one…

Details

Tetrahydrofolate (THF) and its one-carbon derivatives, collectively termed folates, are essential cofactors, but are inherently unstable. While it is clear that chemical oxidation can cleave folates or damage their pterin precursors, very little is known about enzymatic damage to these molecules or about whether the folate biosynthesis pathway responds adaptively to damage to its end-products. The presence of a duplication of the gene encoding the folate biosynthesis enzyme 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase (FolK) in many sequenced bacterial genomes combined with a strong chromosomal clustering of the folK gene with panB, encoding the 5,10-methylene-THF-dependent enzyme ketopantoate hydroxymethyltransferase, led us to infer that PanB has a side activity that cleaves 5,10-methylene-THF, yielding a pterin product that is recycled by FolK. Genetic and metabolic analyses of Escherichia coli strains showed that overexpression of PanB leads to accumulation of the likely folate cleavage product 6-hydroxymethylpterin and other pterins in cells and medium, and-unexpectedly-to a 46% increase in total folate content. In silico modeling of the folate biosynthesis pathway showed that these observations are consistent with the in vivo cleavage of 5,10-methylene-THF by a side-activity of PanB, with FolK-mediated recycling of the pterin cleavage product, and with regulation of folate biosynthesis by folates or their damage products. link: http://identifiers.org/pubmed/27065985

Parameters:

NameDescription
k1=15.8Reaction: ATP + H2_HMPt => AMP + H2_HMPterinPP, Rate Law: compartment*k1*ATP*H2_HMPt
k1=0.004Reaction: CH2_THF => H2_HMPt + p_ABA, Rate Law: compartment*k1*CH2_THF
k1=4080.0; k2=2000.0Reaction: THF + L_serine => CH2_THF + Glycine, Rate Law: compartment*(k1*THF*L_serine-k2*CH2_THF*Glycine)
k1=4000.0Reaction: p_ABA + H2_HMPterinPP => PPi + H2_Pteroate, Rate Law: compartment*k1*p_ABA*H2_HMPterinPP
Km=5.921E-5; V=1.726E-7Reaction: CH2_THF =>, Rate Law: compartment*V*CH2_THF/(Km+CH2_THF)
k1=6184.0Reaction: L_Glutamate + ATP + H2_Pteroate => DHF + ADP + Phosphate, Rate Law: compartment*k1*L_Glutamate*ATP*H2_Pteroate
k1=31170.0Reaction: DHF + NADPH => NADP + THF, Rate Law: compartment*k1*DHF*NADPH
v=1.66E-7Reaction: => H2_HMPt, Rate Law: compartment*v
V=1.243E-7; Km=1.571E-4Reaction: THF =>, Rate Law: compartment*V*THF/(Km+THF)

States:

NameDescription
H2 PteroateH2-Pteroate
ATPATP
NADPNADP
AMPAMP
THFTHF
DHFDHF
PhosphatePhosphate
H2 HMPterinPPH2-HMPterinPP
NADPHNADPH
L GlutamateL-Glutamate
GlycineGlycine
ADPADP
PPiPPi
CH2 THFCH2-THF
p ABAp-ABA
H2 HMPtH2-HMPt
L serineL-serine

Thiele2005 - Genome-scale metabolic network of Helicobacter pylori (iIT341): MODEL1507180007v0.0.1

Thiele2005 - Genome-scale metabolic network of Helicobacter pylori (iIT341)This model is described in the article: [Exp…

Details

Helicobacter pylori is a human gastric pathogen infecting almost half of the world population. Herein, we present an updated version of the metabolic reconstruction of H. pylori strain 26695 based on the revised genome annotation and new experimental data. This reconstruction, iIT341 GSM/GPR, represents a detailed review of the current literature about H. pylori as it integrates biochemical and genomic data in a comprehensive framework. In total, it accounts for 341 metabolic genes, 476 intracellular reactions, 78 exchange reactions, and 485 metabolites. Novel features of iIT341 GSM/GPR include (i) gene-protein-reaction associations, (ii) elementally and charge-balanced reactions, (iii) more accurate descriptions of isoprenoid and lipopolysaccharide metabolism, and (iv) quantitative assessments of the supporting data for each reaction. This metabolic reconstruction was used to carry out in silico deletion studies to identify essential and conditionally essential genes in H. pylori. A total of 128 essential and 75 conditionally essential metabolic genes were identified. Predicted growth phenotypes of single knockouts were validated using published experimental data. In addition, in silico double-deletion studies identified a total of 47 synthetic lethal mutants involving 67 different metabolic genes in rich medium. link: http://identifiers.org/pubmed/16077130

Thiele2011 - Genome-scale metabolic network of Salmonella Typhimurium (STM_v1_0): MODEL1507180017v0.0.1

Thiele2011 - Genome-scale metabolic network of Salmonella Typhimurium (STM_v1_0)This model is described in the article:…

Details

Metabolic reconstructions (MRs) are common denominators in systems biology and represent biochemical, genetic, and genomic (BiGG) knowledge-bases for target organisms by capturing currently available information in a consistent, structured manner. Salmonella enterica subspecies I serovar Typhimurium is a human pathogen, causes various diseases and its increasing antibiotic resistance poses a public health problem.Here, we describe a community-driven effort, in which more than 20 experts in S. Typhimurium biology and systems biology collaborated to reconcile and expand the S. Typhimurium BiGG knowledge-base. The consensus MR was obtained starting from two independently developed MRs for S. Typhimurium. Key results of this reconstruction jamboree include i) development and implementation of a community-based workflow for MR annotation and reconciliation; ii) incorporation of thermodynamic information; and iii) use of the consensus MR to identify potential multi-target drug therapy approaches.Taken together, with the growing number of parallel MRs a structured, community-driven approach will be necessary to maximize quality while increasing adoption of MRs in experimental design and interpretation. link: http://identifiers.org/pubmed/21244678

Thiele2013 - Adrenal gland glandular cells: MODEL1310110001v0.0.1

Thiele2013 - Adrenal gland glandular cellsThe model of adrenal gland glandular cells metabolism is derived from the comm…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Appendix glandular cells: MODEL1310110038v0.0.1

Thiele2013 - Appendix glandular cellsThe model of appendix glandular cells metabolism is derived from the community-driv…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Appendix lymphoid tissue: MODEL1310110032v0.0.1

Thiele2013 - Appendix lymphoid tissueThe model of appendix lymphoid tissue metabolism is derived from the community-driv…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Bone marrow hematopoietic cells: MODEL1310110030v0.0.1

Thiele2013 - Bone marrow hematopoietic cellsThe model of bone marrow hematopoietic cells metabolism is derived from the…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Breast glandular cells: MODEL1310110023v0.0.1

Thiele2013 - Breast glandular cellsThe model of breast glandular cells metabolism is derived from the community-driven g…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Bronchus respiratory epithelial cells: MODEL1310110063v0.0.1

Thiele2013 - Bronchus respiratory epithelial cellsThe model of bronchus respiratory epithelial cells metabolism is deriv…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Cerebellum cells in granular layer: MODEL1310110000v0.0.1

Thiele2013 - Cerebellum cells in granular layerThe model of cerebellum cells in granular layer metabolism is derived fro…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Cerebellum cells in molecular layer: MODEL1310110016v0.0.1

Thiele2013 - Cerebellum cells in molecular layerThe model of cerebellum cells in molecular layer metabolism is derived f…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Cerebellum Purkinje cells: MODEL1310110050v0.0.1

Thiele2013 - Cerebellum Purkinje cellsThe model of cerebellum Purkinje cells metabolism is derived from the community-dr…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Cerebral cortex glial cells: MODEL1310110064v0.0.1

Thiele2013 - Cerebral cortex glial cellsThe model of cerebral cortex glial cells metabolism is derived from the communit…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Cerebral cortex neuronal cells: MODEL1310110033v0.0.1

Thiele2013 - Cerebral cortex neuronal cellsThe model of cerebral cortex neuronal cells metabolism is derived from the co…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Cervix uterine glandular cells: MODEL1310110026v0.0.1

Thiele2013 - Cervix uterine glandular cellsThe model of cervix uterine glandular cells metabolism is derived from the co…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Cervix uterine squamous epithelial cells: MODEL1310110044v0.0.1

Thiele2013 - Cervix uterine squamous epithelial cellsThe model of cervix uterine squamous epithelial cells metabolism is…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Colon glandular cells: MODEL1310110043v0.0.1

Thiele2013 - Colon glandular cellsThe model of colon glandular cells metabolism is derived from the community-driven glo…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Duodenum glandular cells: MODEL1310110012v0.0.1

Thiele2013 - Duodenum glandular cellsThe model of duodenum glandular cells metabolism is derived from the community-driv…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Epididymis glandular cells: MODEL1310110054v0.0.1

Thiele2013 - Epididymis glandular cellsThe model of epididymis glandular cells metabolism is derived from the community-…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Esophagus squamous epithelial cells: MODEL1310110048v0.0.1

Thiele2013 - Esophagus squamous epithelial cellsThe model of esophagus squamous epithelial cells metabolism is derived f…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Fallopian tube glandular cells: MODEL1310110009v0.0.1

Thiele2013 - Fallopian tube glandular cellsThe model of fallopian tube glandular cells metabolism is derived from the co…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Gall bladder glandular cells: MODEL1310110042v0.0.1

Thiele2013 - Gall bladder glandular cellsThe model of gall bladder glandular cells metabolism is derived from the commun…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Heart muscle myocytes: MODEL1310110056v0.0.1

Thiele2013 - Heart muscle myocytesThe model of heart muscle myocytes metabolism is derived from the community-driven glo…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Hippocampus glial cells: MODEL1310110005v0.0.1

Thiele2013 - Hippocampus glial cellsThe model of hippocampus glial cells metabolism is derived from the community-driven…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Hippocampus neuronal cells: MODEL1310110034v0.0.1

Thiele2013 - Hippocampus neuronal cellsThe model of hippocampus neuronal cells metabolism is derived from the community-…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Human metabolism global reconstruction (Recon 2): MODEL1109130000v0.0.1

Thiele2013 - Human metabolism global reconstruction (Recon 2)Community-driven global reconstruction of human metabolism…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Kidney cells in glomeruli: MODEL1310110053v0.0.1

Thiele2013 - Kidney cells in glomeruliThe model of kidney cells in glomeruli metabolism is derived from the community-dr…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Kidney cells in tubules: MODEL1310110047v0.0.1

Thiele2013 - Kidney cells in tubulesThe model of kidney cells in tubules metabolism is derived from the community-driven…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Lateral ventricle glial cells: MODEL1310110037v0.0.1

Thiele2013 - Lateral ventricle glial cellsThe model of lateral ventricle glial cells metabolism is derived from the comm…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439

Thiele2013 - Lateral ventricle neuronal cells: MODEL1310110040v0.0.1

Thiele2013 - Lateral ventricle neuronal cellsThe model of lateral ventricle neuronal cells metabolism is derived from th…

Details

Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ∼2× more reactions and ∼1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type-specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/. link: http://identifiers.org/pubmed/23455439