SBMLBioModels: U - W

U


Uhlén2015 - Human tissue-based proteome metabolic network - pancreas: MODEL1411240025v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - pancreas Human pancreas specific proteome metabolic network…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - placenta: MODEL1411240030v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - placenta Human placenta specific proteome metabolic network…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - prostate: MODEL1411240028v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - prostate Human prostate specific proteome metabolic network…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - rectum: MODEL1411240007v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - rectum Human rectum specific proteome metabolic network Th…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - salivary gland: MODEL1411240013v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - salivary gland Human salivary gland specific proteome metabo…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - skeletal: MODEL1411240009v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - skeletal Human skeletal tissue specific proteome metabolic n…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - skin: MODEL1411240022v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - skin Human skin specific proteome metabolic network This m…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - small intestine: MODEL1411240024v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - small intestine Human small intestine specific proteome meta…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - smooth muscle: MODEL1411240005v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - smooth muscle Human smooth muscle tissue specific proteome m…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - spleen: MODEL1411240014v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - spleen Human spleen specific proteome metabolic network Th…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - stomach: MODEL1411240017v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - stomach Human stomach specific proteome metabolic network…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - testis: MODEL1411240018v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - testis Human testis specific proteome metabolic network Th…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - thyroid: MODEL1411240008v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - thyroid Human thyroid tissue specific proteome metabolic net…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - tonsil: MODEL1411240023v0.0.1

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

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Uhlén2015 - Human tissue-based proteome metabolic network - urinary: MODEL1411240031v0.0.1

Uhlén2015 - Human tissue-based proteome metabolic network - urinary Human urinary track specific proteome metabolic netw…

Details

Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body. link: http://identifiers.org/pubmed/25613900

Ung2008_EGFR_Endocytosis: BIOMD0000000205v0.0.1

Model reproduces the various plots in the publication for "Control" concentrations. Model successfully tested on MathSBM…

Details

Deregulations of EGFR endocytosis in EGFR-ERK signaling are known to cause cancers and developmental disorders. Mutations that impaired c-Cbl-EGFR association delay EGFR endocytosis and produce higher mitogenic signals in lung cancer. ROCK, an effector of small GTPase RhoA was shown to negatively regulate EGFR endocytosis via endophilin A1. A mathematical model was developed to study how RhoA and ROCK regulate EGFR endocytosis. Our study suggested that over-expressing RhoA as well as ROCK prolonged ERK activation partly by reducing EGFR endocytosis. Overall, our study hypothesized an alternative role of RhoA in tumorigenesis in addition to its regulation of cytoskeleton and cell motility. link: http://identifiers.org/pubmed/18505685

Parameters:

NameDescription
k1=0.001 sec_1; k2=10.0 uM_1_s_1Reaction: species_62 => species_63 + species_57, Rate Law: compartment_0*(k1*species_62-k2*species_63*species_57)
k1=0.2661 sec_1Reaction: species_6 => species_3 + species_5, Rate Law: compartment_0*k1*species_6
k2=1.67 sec_1; k1=5.0 uM_1_s_1Reaction: species_166 + species_26 => species_167, Rate Law: compartment_0*(k1*species_166*species_26-k2*species_167)
k1=2.9 sec_1Reaction: species_29 => species_25 + species_30, Rate Law: compartment_0*k1*species_29
k1=0.27 sec_1Reaction: species_42 => species_33 + species_41, Rate Law: compartment_0*k1*species_42
k1=4.481 sec_1; k2=0.3 uM_1_s_1Reaction: species_9 => species_4 + species_10, Rate Law: compartment_0*(k1*species_9-k2*species_4*species_10)
k2=0.1 sec_1; k1=8.898 uM_1_s_1Reaction: species_35 + species_15 => species_46, Rate Law: compartment_0*(k1*species_35*species_15-k2*species_46)
k1=100.0 uM_1_s_1; k2=0.0038 sec_1Reaction: species_0 + species_1 => species_2, Rate Law: compartment_0*(k1*species_0*species_1-k2*species_2)
k2=0.9356 sec_1; k1=40.0 uM_1_s_1Reaction: species_101 + species_94 => species_102, Rate Law: compartment_0*(k1*species_101*species_94-k2*species_102)
k1=3.0 uM_1_s_1; k2=0.033 sec_1Reaction: species_30 + species_33 => species_34, Rate Law: compartment_0*(k1*species_30*species_33-k2*species_34)
k1=3.0 uM_1_s_1; k2=0.5 sec_1Reaction: species_4 + species_131 => species_133, Rate Law: compartment_0*(k1*species_4*species_131-k2*species_133)
k1=1.205 sec_1Reaction: species_190 => species_143 + species_82 + species_95, Rate Law: compartment_0*k1*species_190
k2=1.0 sec_1; k1=3.0 uM_1_s_1Reaction: species_58 + species_57 => species_59, Rate Law: compartment_0*(k1*species_58*species_57-k2*species_59)
k1=10.0 sec_1Reaction: species_103 => species_101 + species_95, Rate Law: compartment_0*k1*species_103
k1=3.0 uM_1_s_1; k2=0.05 sec_1Reaction: species_4 + species_12 => species_20, Rate Law: compartment_0*(k1*species_4*species_12-k2*species_20)
k2=0.01 sec_1; k1=0.1 uM_1_s_1Reaction: species_4 + species_44 => species_87, Rate Law: compartment_0*(k1*species_4*species_44-k2*species_87)
k1=25.0 sec_1Reaction: species_56 => species_52 + species_57, Rate Law: compartment_0*k1*species_56
k1=2.845 uM_1_s_1; k2=0.96 sec_1Reaction: species_95 + species_86 => species_104, Rate Law: compartment_0*(k1*species_95*species_86-k2*species_104)
k2=0.0214 sec_1; k1=10.0 uM_1_s_1Reaction: species_57 + species_73 => species_74, Rate Law: compartment_0*(k1*species_57*species_73-k2*species_74)
k1=10.0 uM_1_s_1; k2=0.02 sec_1Reaction: species_2 => species_3, Rate Law: compartment_0*(k1*species_2*species_2-k2*species_3)
k1=3.0 sec_1Reaction: species_61 => species_62 + species_60, Rate Law: compartment_0*k1*species_61
k1=2.014 sec_1Reaction: species_3 => species_4, Rate Law: compartment_0*k1*species_3
k1=0.058 sec_1Reaction: species_39 => species_28 + species_38, Rate Law: compartment_0*k1*species_39
k2=0.5 sec_1; k1=1.667 uM_1_s_1Reaction: species_133 + species_25 => species_166, Rate Law: compartment_0*(k1*species_133*species_25-k2*species_166)
k1=5.0 uM_1_s_1; k2=0.5 sec_1Reaction: species_33 + species_41 => species_43, Rate Law: compartment_0*(k1*species_33*species_41-k2*species_43)
k2=0.6 sec_1; k1=90.0 uM_1_s_1Reaction: species_4 + species_7 => species_8, Rate Law: compartment_0*(k1*species_4*species_7-k2*species_8)
k1=1.67 sec_1Reaction: species_158 => species_151 + species_35, Rate Law: compartment_0*k1*species_158
k1=5.0 sec_1Reaction: species_72 => species_71 + species_55, Rate Law: compartment_0*k1*species_72
k1=1.1 uM_1_s_1; k2=0.033 sec_1Reaction: species_68 + species_69 => species_70, Rate Law: compartment_0*(k1*species_68*species_69-k2*species_70)
k1=2.661 sec_1Reaction: species_92 => species_3 + species_12 + species_90, Rate Law: compartment_0*k1*species_92
k1=0.1002 sec_1Reaction: species_182 => species_183, Rate Law: compartment_0*k1*species_182
k1=0.25 uM_1_s_1; k2=0.5 sec_1Reaction: species_28 + species_38 => species_40, Rate Law: compartment_0*(k1*species_28*species_38-k2*species_40)
k1=3.14 uM_1_s_1; k2=0.2 sec_1Reaction: species_4 + species_5 => species_6, Rate Law: compartment_0*(k1*species_4*species_5-k2*species_6)
k2=1.0 sec_1; k1=8.898 uM_1_s_1Reaction: species_35 + species_68 => species_129, Rate Law: compartment_0*(k1*species_35*species_68-k2*species_129)
k1=17.0 sec_1Reaction: species_57 => species_55, Rate Law: compartment_0*k1*species_57
k1=1.693 sec_1Reaction: species_163 => species_164, Rate Law: compartment_0*k1*species_163
k1=0.426 sec_1Reaction: species_46 => species_35 + species_4 + species_10 + species_12 + species_47, Rate Law: compartment_0*k1*species_46
k1=0.1298 sec_1Reaction: species_84 => species_94, Rate Law: compartment_0*k1*species_84
k1=16.0 sec_1Reaction: species_32 => species_30 + species_33, Rate Law: compartment_0*k1*species_32
k1=4.0 uM_1_s_1; k2=0.01833 sec_1Reaction: species_25 + species_26 => species_27, Rate Law: compartment_0*(k1*species_25*species_26-k2*species_27)
k1=16.67 uM_1_s_1; k2=0.05 sec_1Reaction: species_145 + species_86 => species_176, Rate Law: compartment_0*(k1*species_145*species_86-k2*species_176)
k2=5.0 uM_1_s_1; k1=1.67 sec_1Reaction: species_165 => species_162 + species_30, Rate Law: compartment_0*(k1*species_165-k2*species_162*species_30)

States:

NameDescription
species 70[Rho-associated protein kinase 1; Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN]
species 27[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
species 71[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN]
species 31[Mitogen-activated protein kinase 1]
species 62[1-phosphatidyl-1D-myo-inositol 3,4,5-trisphosphate; RAC-alpha serine/threonine-protein kinase]
species 149[GTP; Receptor protein-tyrosine kinase; Pro-epidermal growth factor; SHC-transforming protein 2; Growth factor receptor-bound protein 2; Ras-related protein R-Ras2; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase kinase kinase 1]
species 184[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; SHC-transforming protein 2; Growth factor receptor-bound protein 2; RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
species 4[Receptor protein-tyrosine kinase; Pro-epidermal growth factor]
species 28[Dual specificity mitogen-activated protein kinase kinase 1]
species 59[1-phosphatidyl-1D-myo-inositol 3,4,5-trisphosphate; RAC-alpha serine/threonine-protein kinase]
species 61[1-phosphatidyl-1D-myo-inositol 3,4,5-trisphosphate; RAC-alpha serine/threonine-protein kinase; 3-phosphoinositide-dependent protein kinase 1]
species 34[Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
species 29[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
species 30[Dual specificity mitogen-activated protein kinase kinase 1]
species 166[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; Growth factor receptor-bound protein 2; RAF proto-oncogene serine/threonine-protein kinase]
species 57[1-phosphatidyl-1D-myo-inositol 3,4,5-trisphosphate]
species 5[Tyrosine-protein phosphatase non-receptor type 11]
species 94[Rho guanine nucleotide exchange factor 12]
species 186[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; SHC-transforming protein 2; Growth factor receptor-bound protein 2; RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
species 2[Receptor protein-tyrosine kinase; Pro-epidermal growth factor]
species 163[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; SHC-transforming protein 2; Growth factor receptor-bound protein 2; RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
species 183[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; Growth factor receptor-bound protein 2; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1]
species 74[1-phosphatidyl-1D-myo-inositol 3,4,5-trisphosphate; Rho guanine nucleotide exchange factor 6]
species 33[Mitogen-activated protein kinase 1]
species 145[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; Growth factor receptor-bound protein 2; RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase kinase kinase 1]
species 146[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; Growth factor receptor-bound protein 2; RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase kinase kinase 1]
species 148[GTP; Receptor protein-tyrosine kinase; Pro-epidermal growth factor; SHC-transforming protein 2; Growth factor receptor-bound protein 2; Ras-related protein R-Ras2; Mitogen-activated protein kinase kinase kinase 1]
species 72[1-phosphatidyl-1D-myo-inositol 3,4,5-trisphosphate; Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN]
species 147[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; Growth factor receptor-bound protein 2; RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase kinase kinase 1]
species 73[Rho guanine nucleotide exchange factor 6]
species 69[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN]
species 95pRhoGAP
species 164[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; SHC-transforming protein 2; Growth factor receptor-bound protein 2; RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
species 35[Mitogen-activated protein kinase 1]
species 3[Receptor protein-tyrosine kinase; Pro-epidermal growth factor]
species 56[1-phosphatidyl-1D-myo-inositol 3,4-bisphosphate; Phosphoinositide 3-kinase regulatory subunit 5]
species 58[RAC-alpha serine/threonine-protein kinase]
species 165[Receptor protein-tyrosine kinase; Pro-epidermal growth factor; SHC-transforming protein 2; Growth factor receptor-bound protein 2; RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
species 26[Dual specificity mitogen-activated protein kinase kinase 1]

Unni2019 - Mathematical Modeling, Analysis, and Simulation of Tumor Dynamics with Drug Interventions: BIOMD0000000888v0.0.1

Mathematical Modeling, Analysis, and Simulation of Tumor Dynamics with Drug Interventions Pranav Unni 1 and Padmanabhan…

Details

Over the last few decades, there have been significant developments in theoretical, experimental, and clinical approaches to understand the dynamics of cancer cells and their interactions with the immune system. These have led to the development of important methods for cancer therapy including virotherapy, immunotherapy, chemotherapy, targeted drug therapy, and many others. Along with this, there have also been some developments on analytical and computational models to help provide insights into clinical observations. This work develops a new mathematical model that combines important interactions between tumor cells and cells in the immune systems including natural killer cells, dendritic cells, and cytotoxic CD8+ T cells combined with drug delivery to these cell sites. These interactions are described via a system of ordinary differential equations that are solved numerically. A stability analysis of this model is also performed to determine conditions for tumor-free equilibrium to be stable. We also study the influence of proliferation rates and drug interventions in the dynamics of all the cells involved. Another contribution is the development of a novel parameter estimation methodology to determine optimal parameters in the model that can reproduce a given dataset. Our results seem to suggest that the model employed is a robust candidate for studying the dynamics of tumor cells and it helps to provide the dynamic interactions between the tumor cells, immune system, and drug-response systems. link: http://identifiers.org/pubmed/31687042

Parameters:

NameDescription
f_1 = 1.0E-8; d_2 = 4.0E-6; g = 0.024; d_3 = 1.0E-4Reaction: D => ; L, N, T, Rate Law: compartment*(((f_1*L+d_2*N)-d_3*T)*D-g*D)
s_1 = 13000.0; g_1 = 0.0; h_1 = 0.0Reaction: => N; T, Rate Law: compartment*(s_1+g_1*N*T*T/(h_1+T*T))
i = 0.02; h = 3.42E-10; u = 1.8E-8Reaction: L => ; T, N, Rate Law: compartment*(h*L*T+u*N*L*L+i*L)
c_1 = 3.5E-6; j = 1.0E-7; k = 1.0E-7Reaction: T => ; N, D, L, Rate Law: compartment*(c_1*N+j*D+k*L)*T
d_1 = 1.0E-6; c_2 = 1.0E-7; e = 0.0412Reaction: N => ; T, D, Rate Law: compartment*((c_2*T+d_1*D)*N+e*N)
s_2 = 480.0Reaction: => D, Rate Law: compartment*s_2
a = 0.431; b = 2.17E-8Reaction: => T, Rate Law: compartment*a*T*(1-b*T)
f_2 = 0.01; r_1 = 0.0Reaction: => L; D, T, N, Rate Law: compartment*(f_2*D*T+r_1*N*T)

States:

NameDescription
T[Tumor Mass]
N[natural killer cell]
D[dendritic cell]
L[T-lymphocyte]

V


Valero2006_Adenine_TernaryCycle: BIOMD0000000231v0.0.1

This a model from the article: A kinetic study of a ternary cycle between adenine nucleotides. Valero E, Varón R, Ga…

Details

In the present paper, a kinetic study is made of the behavior of a moiety-conserved ternary cycle between the adenine nucleotides. The system contains the enzymes S-acetyl coenzyme A synthetase, adenylate kinase and pyruvate kinase, and converts ATP into AMP, then into ADP and finally back to ATP. L-Lactate dehydrogenase is added to the system to enable continuous monitoring of the progress of the reaction. The cycle cannot work when the only recycling substrate in the reaction medium is AMP. A mathematical model is proposed whose kinetic behavior has been analyzed both numerically by integration of the nonlinear differential equations describing the kinetics of the reactions involved, and analytically under steady-state conditions, with good agreement with the experimental results being obtained. The data obtained showed that there is a threshold value of the S-acetyl coenzyme A synthetase/adenylate kinase ratio, above which the cycle stops because all the recycling substrate has been accumulated as AMP, never reaching the steady state. In addition, the concept of adenylate energy charge has been applied to the system, obtaining the enabled values of the rate constants for a fixed adenylate energy charge value and vice versa. link: http://identifiers.org/pubmed/16884499

Parameters:

NameDescription
Vmapp1 = 2.3; Kmapp1 = 700.0Reaction: ATP => AMP, Rate Law: Vmapp1*ATP/(Kmapp1+ATP)
Vmapp3 = 65.0; Kmapp3 = 260.0Reaction: ADP => ATP + Pyr, Rate Law: Vmapp3*ADP/(Kmapp3+ADP)
K = 71000.0; Km2AMP = 110.0; Km2ATP = 25.0; Vm2 = 170.0Reaction: ATP + AMP => ADP, Rate Law: Vm2*ATP*AMP/(K+Km2ATP*AMP+Km2AMP*ATP+ATP*AMP)
k4 = 5.0Reaction: Pyr + NADH => Lac, Rate Law: k4*Pyr

States:

NameDescription
ATP[ATP; ATP]
NADH[NADH; NADH]
ADP[ADP; ADP]
Pyr[IPR001697; Pyruvate kinase]
AMP[AMP; AMP]
Lac[L-lactate dehydrogenase; IPR011304]

Valero2016 - Ascorbate-Glutathione cycle in chloroplasts under light/dark conditions: BIOMD0000000589v0.0.1

Valero2016 - Ascorbate-Glutathione cycle in chloroplasts under light/dark conditionsThis model is described in the artic…

Details

Light/dark cycles are probably the most important environmental signals that regulate plant development. Light is essential for photosynthesis, but an excess, in combination with the unavoidable presence of atmospheric oxygen inside the chloroplast, leads to excessive reactive oxygen species production. Among the defense mechanisms that activate plants to cope with environmental stress situations, it is worth noting the ascorbate-glutathione cycle, a complex metabolic pathway in which a variety of photochemical, chemical and enzymatic steps are involved.We herein studied the dynamic behavior of this pathway under light/dark conditions and for several consecutive days. For this purpose, a mathematical model was developed including a variable electron source with a rate law proportional to the intensity of solar irradiance during the photoperiod, and which is continuously turned off at night and on again the next day. The model is defined by a nonlinear system of ordinary differential equations with an on/off time-dependent input, including a parameter to simulate the fact that the photoperiod length is not constant throughout the year, and which takes into account the particular experimental kinetics of each enzyme involved in the pathway. Unlike previous models, which have only provided steady-state solutions, the present model is able to simulate diurnal fluctuations in the metabolite concentrations, fluxes and enzymatic rates involved in the network.The obtained results are broadly consistent with experimental observations and highlight the key role played by ascorbate recycling for plants to adapt to their surrounding environment. This approach provides a new strategy to in vivo studies to analyze plant defense mechanisms against oxidative stress induced by external changes, which can also be extrapolated to other complex metabolic pathways to constitute a useful tool to the scientific community in general. link: http://identifiers.org/pubmed/26797294

Parameters:

NameDescription
k6 = 720.0; k3 = 0.01; F13 = 0.0; vDHAR = 0.0; k3APX = 7560.0; k2APX = 180000.0; k1 = 1800.0; k5 = 0.0072; vMDAR = 0.0Reaction: ASC = (((((vDHAR+k1*MDA^2+k3*DHA*GSH+F13)-k2APX*ASC*CoI)-k3APX*ASC*CoII)-k6*O2neg*ASC)-2*k5*H2O2*ASC)+2*vMDAR, Rate Law: (((((vDHAR+k1*MDA^2+k3*DHA*GSH+F13)-k2APX*ASC*CoI)-k3APX*ASC*CoII)-k6*O2neg*ASC)-2*k5*H2O2*ASC)+2*vMDAR
k3 = 0.01; vDHAR = 0.0; k1 = 1800.0Reaction: DHA = ((-vDHAR)+k1*MDA^2)-k3*DHA*GSH, Rate Law: ((-vDHAR)+k1*MDA^2)-k3*DHA*GSH
vGR = 0.0; k3 = 0.01; vDHAR = 0.0; k4 = 2520.0Reaction: GSH = 2*(((vGR-vDHAR)-k4*O2neg*GSH)-k3*DHA*GSH), Rate Law: 2*(((vGR-vDHAR)-k4*O2neg*GSH)-k3*DHA*GSH)
k1APX = 43200.0; k3APX = 7560.0; Metabolite_17 = 40.0; k5APX = 1.0Reaction: APX = (-k1APX)*H2O2*APX+k3APX*ASC*CoII+k5APX*(((Metabolite_17-APX)-CoI)-CoII), Rate Law: (-k1APX)*H2O2*APX+k3APX*ASC*CoII+k5APX*(((Metabolite_17-APX)-CoI)-CoII)
vGR = 0.0; kN = 3.97846553950471E-12; F12 = 7.95693107900941E-10; vMDAR = 0.0Reaction: NADPH = (((-vGR)-kN*NADPH)+F12*0.5)-vMDAR, Rate Law: (((-vGR)-kN*NADPH)+F12*0.5)-vMDAR
k4APX = 2520.0Reaction: APXi = k4APX*H2O2*CoI, Rate Law: k4APX*H2O2*CoI
k4APX = 2520.0; k1APX = 43200.0; k2APX = 180000.0Reaction: CoI = (k1APX*H2O2*APX-k2APX*ASC*CoI)-k4APX*H2O2*CoI, Rate Law: (k1APX*H2O2*APX-k2APX*ASC*CoI)-k4APX*H2O2*CoI
k6 = 720.0; F13 = 0.0; k3APX = 7560.0; k2APX = 180000.0; k1 = 1800.0; k5 = 0.0072; vMDAR = 0.0Reaction: MDA = ((((k2APX*ASC*CoI+k3APX*ASC*CoII)-2*k1*MDA^2)+k6*O2neg*ASC+2*k5*H2O2*ASC)-F13)-2*vMDAR, Rate Law: ((((k2APX*ASC*CoI+k3APX*ASC*CoII)-2*k1*MDA^2)+k6*O2neg*ASC+2*k5*H2O2*ASC)-F13)-2*vMDAR
k3APX = 7560.0; k2APX = 180000.0Reaction: CoII = k2APX*ASC*CoI-k3APX*ASC*CoII, Rate Law: k2APX*ASC*CoI-k3APX*ASC*CoII
k2 = 720.0; k6 = 720.0; vSOD = 0.0; F11 = 1.35629466714537E-10; k4 = 2520.0Reaction: O2neg = ((((-2)*vSOD+F11)-2*k2*O2neg^2)-k6*O2neg*ASC)-k4*O2neg*GSH, Rate Law: ((((-2)*vSOD+F11)-2*k2*O2neg^2)-k6*O2neg*ASC)-k4*O2neg*GSH
k2 = 720.0; k6 = 720.0; vSOD = 0.0; k4APX = 2520.0; k1APX = 43200.0; k5 = 0.0072; k4 = 2520.0Reaction: H2O2 = (((vSOD-k1APX*H2O2*APX)-k4APX*H2O2*CoI)+k2*O2neg^2+k6*O2neg*ASC+k4*O2neg*GSH)-k5*H2O2*ASC, Rate Law: (((vSOD-k1APX*H2O2*APX)-k4APX*H2O2*CoI)+k2*O2neg^2+k6*O2neg*ASC+k4*O2neg*GSH)-k5*H2O2*ASC

States:

NameDescription
O2neg[oxide(2-)]
APXi[Probable L-ascorbate peroxidase 4; inactive]
CoI[oxidising agent]
CoII[oxidising agent]
GSSG[glutathione; oxidized]
NADPH[NADPH]
DHA[dehydroascorbic acid]
ASC[ascorbate]
H2O2[hydrogen peroxide]
NADPplus[NADP(+)]
MDA[monodehydro-L-ascorbic acid]
GSH[glutathione]
APX[Probable L-ascorbate peroxidase 4]

vanBeek2007_OxPhos_HeartMuscleCells: MODEL1006230027v0.0.1

This a model from the article: Adenine nucleotide-creatine-phosphate module in myocardial metabolic system explains fa…

Details

Computational models of a large metabolic system can be assembled from modules that represent a biological function emerging from interaction of a small subset of molecules. A "skeleton model" is tested here for a module that regulates the first phase of dynamic adaptation of oxidative phosphorylation (OxPhos) to demand in heart muscle cells. The model contains only diffusion, mitochondrial outer membrane (MOM) permeation, and two isoforms of creatine kinase (CK), in cytosol and mitochondrial intermembrane space (IMS), respectively. The communication with two neighboring modules occurs via stimulation of mitochondrial ATP production by ADP and P(i) from the IMS and via time-varying cytosolic ATP hydrolysis during contraction. Assuming normal cytosolic diffusion and high MOM permeability for ADP, the response time of OxPhos (t(mito); generalized time constant) to steps in cardiac pacing rate is predicted to be 2.4 s. In contrast, with low MOM permeability, t(mito) is predicted to be 15 s. An optimized MOM permeability of 21 mum/s gives t(mito) = 3.7 s, in agreement with experiments on rabbit heart with blocked glycolytic ATP synthesis. The model correctly predicts a lower t(mito) if CK activity is reduced by 98%. Among others, the following predictions result from the model analysis: 1) CK activity buffers large ADP oscillations; 2) ATP production is pulsatile in beating heart, although it adapts slowly to demand with "time constant" approximately 14 heartbeats; 3) if the muscle isoform of CK is overexpressed, OxPhos reacts slower to changing workload; and 4) if mitochondrial CK is overexpressed, OxPhos reacts faster. link: http://identifiers.org/pubmed/17581855

Vanee2010 - Genome-scale metabolic model of Cryptosporidium hominis (iNV213): MODEL1507180071v0.0.1

Vanee2010 - Genome-scale metabolic model of Cryptosporidium hominis (iNV213)This model is described in the article: [A…

Details

The apicomplexan Cryptosporidium is a protozoan parasite of humans and other mammals. Cryptosporidium species cause acute gastroenteritis and diarrheal disease in healthy humans and animals, and cause life-threatening infection in immunocompromised individuals such as people with AIDS. The parasite has a one-host life cycle and commonly invades intestinal epithelial cells. The current genome annotation of C. hominis, the most serious human pathogen, predicts 3884 genes of which ca. 1581 have predicted functional annotations. Using a combination of bioinformatics analysis, biochemical evidence, and high-throughput data, we have constructed a genome-scale metabolic model of C. hominis. The model is comprised of 213 gene-associated enzymes involved in 540 reactions among the major metabolic pathways and provides a link between the genotype and the phenotype of the organism, making it possible to study and predict behavior based upon genome content. This model was also used to analyze the two life stages of the parasite by integrating the stage-specific proteomic data for oocyst and sporozoite stages. Overall, this model provides a computational framework to systematically study and analyze various functional behaviors of C. hominis with respect to its life cycle and pathogenicity. link: http://identifiers.org/pubmed/20491062

vanEunen2012 - Yeast Glycolysis (glucose upshift): MODEL1403250001v0.0.1

This is corresponding to the model of yeast glycolysis "glucose upshift" condition described in the paper "Testing Bioch…

Details

A decade ago, a team of biochemists including two of us, modeled yeast glycolysis and showed that one of the most studied biochemical pathways could not be quite understood in terms of the kinetic properties of the constituent enzymes as measured in cell extract. Moreover, when the same model was later applied to different experimental steady-state conditions, it often exhibited unrestrained metabolite accumulation.Here we resolve this issue by showing that the results of such ab initio modeling are improved substantially by (i) including appropriate allosteric regulation and (ii) measuring the enzyme kinetic parameters under conditions that resemble the intracellular environment. The following modifications proved crucial: (i) implementation of allosteric regulation of hexokinase and pyruvate kinase, (ii) implementation of V(max) values measured under conditions that resembled the yeast cytosol, and (iii) redetermination of the kinetic parameters of glyceraldehyde-3-phosphate dehydrogenase under physiological conditions.Model predictions and experiments were compared under five different conditions of yeast growth and starvation. When either the original model was used (which lacked important allosteric regulation), or the enzyme parameters were measured under conditions that were, as usual, optimal for high enzyme activity, fructose 1,6-bisphosphate and some other glycolytic intermediates tended to accumulate to unrealistically high concentrations. Combining all adjustments yielded an accurate correspondence between model and experiments for all five steady-state and dynamic conditions. This enhances our understanding of in vivo metabolism in terms of in vitro biochemistry. link: http://identifiers.org/pubmed/22570597

vanEunen2013 - Network dynamics of fatty acid β-oxidation (steady-state model): BIOMD0000000505v0.0.1

vanEunen2013 - Network dynamics of fatty acid β-oxidation (steady-state model)Lipid metabolism plays an important role i…

Details

Fatty-acid metabolism plays a key role in acquired and inborn metabolic diseases. To obtain insight into the network dynamics of fatty-acid β-oxidation, we constructed a detailed computational model of the pathway and subjected it to a fat overload condition. The model contains reversible and saturable enzyme-kinetic equations and experimentally determined parameters for rat-liver enzymes. It was validated by adding palmitoyl CoA or palmitoyl carnitine to isolated rat-liver mitochondria: without refitting of measured parameters, the model correctly predicted the β-oxidation flux as well as the time profiles of most acyl-carnitine concentrations. Subsequently, we simulated the condition of obesity by increasing the palmitoyl-CoA concentration. At a high concentration of palmitoyl CoA the β-oxidation became overloaded: the flux dropped and metabolites accumulated. This behavior originated from the competition between acyl CoAs of different chain lengths for a set of acyl-CoA dehydrogenases with overlapping substrate specificity. This effectively induced competitive feedforward inhibition and thereby led to accumulation of CoA-ester intermediates and depletion of free CoA (CoASH). The mitochondrial [NAD⁺]/[NADH] ratio modulated the sensitivity to substrate overload, revealing a tight interplay between regulation of β-oxidation and mitochondrial respiration. link: http://identifiers.org/pubmed/23966849

Parameters:

NameDescription
KmvlcadC14AcylCoAMAT = 4.0 uM; KmvlcadC14EnoylCoAMAT = 1.08 uM; KmvlcadFAD = 0.12 uM; KmvlcadFADH = 24.2 uM; KmvlcadC12AcylCoAMAT = 2.7 uM; sfvlcadC14=0.42 dimensionless; KmvlcadC12EnoylCoAMAT = 1.08 uM; Vvlcad = 0.008 uM per min per mgProtein; KmvlcadC16EnoylCoAMAT = 1.08 uM; KmvlcadC16AcylCoAMAT = 6.5 uM; Keqvlcad = 6.0 dimensionlessReaction: C14AcylCoAMAT => C14EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, FADHMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, FADHMAT, Rate Law: sfvlcadC14*Vvlcad*(C14AcylCoAMAT*(FADtMAT-FADHMAT)/(KmvlcadC14AcylCoAMAT*KmvlcadFAD)-C14EnoylCoAMAT*FADHMAT/(KmvlcadC14AcylCoAMAT*KmvlcadFAD*Keqvlcad))/((1+C14AcylCoAMAT/KmvlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmvlcadC14EnoylCoAMAT+C16AcylCoAMAT/KmvlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmvlcadC16EnoylCoAMAT+C12AcylCoAMAT/KmvlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmvlcadC12EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmvlcadFAD+FADHMAT/KmvlcadFADH))
KmlcadC8AcylCoAMAT = 123.0 uM; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; sflcadC10=0.75 dimensionless; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; Keqlcad = 6.0 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C10AcylCoAMAT => C10EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C10EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C10EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, Rate Law: sflcadC10*Vlcad*(C10AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC10AcylCoAMAT*KmlcadFAD)-C10EnoylCoAMAT*FADHMAT/(KmlcadC10AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
KmmcadC12AcylCoAMAT = 5.7 uM; KmmcadC8AcylCoAMAT = 4.0 uM; KmmcadC4AcylCoAMAT = 135.0 uM; KmmcadC10AcylCoAMAT = 5.4 uM; KmmcadFAD = 0.12 uM; KmmcadC10EnoylCoAMAT = 1.08 uM; KmmcadC4EnoylCoAMAT = 1.08 uM; Vmcad = 0.081 uM per min per mgProtein; KmmcadC6AcylCoAMAT = 9.4 uM; sfmcadC10=0.8 dimensionless; KmmcadC6EnoylCoAMAT = 1.08 uM; KmmcadFADH = 24.2 uM; KmmcadC12EnoylCoAMAT = 1.08 uM; KmmcadC8EnoylCoAMAT = 1.08 uM; Keqmcad = 6.0 dimensionlessReaction: C10AcylCoAMAT => C10EnoylCoAMAT + FADHMAT; C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C10AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C10EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, C10AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C10EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: sfmcadC10*Vmcad*(C10AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC10AcylCoAMAT*KmmcadFAD)-C10EnoylCoAMAT*FADHMAT/(KmmcadC10AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))
sfmckatC10=0.65 dimensionless; KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C10KetoacylCoAMAT => C8AcylCoAMAT + AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, C10KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, C10KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC10*Vmckat*(C10KetoacylCoAMAT*CoAMAT/(KmmckatC10KetoacylCoAMAT*KmmckatCoAMAT)-C8AcylCoAMAT*AcetylCoAMAT/(KmmckatC10KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmscadFAD = 0.12 uM; KmscadC4EnoylCoAMAT = 1.08 uM; sfscadC4=1.0 dimensionless; Vscad = 0.081 uM per min per mgProtein; KmscadFADH = 24.2 uM; KmscadC4AcylCoAMAT = 10.7 uM; KmscadC6AcylCoAMAT = 285.0 uM; KmscadC6EnoylCoAMAT = 1.08 uM; Keqscad = 6.0 dimensionlessReaction: C4AcylCoAMAT => C4EnoylCoAMAT + FADHMAT; C6AcylCoAMAT, FADtMAT, C6EnoylCoAMAT, C4AcylCoAMAT, C6AcylCoAMAT, FADtMAT, C4EnoylCoAMAT, C6EnoylCoAMAT, FADHMAT, C4AcylCoAMAT, C6AcylCoAMAT, FADtMAT, C4EnoylCoAMAT, C6EnoylCoAMAT, FADHMAT, Rate Law: sfscadC4*Vscad*(C4AcylCoAMAT*(FADtMAT-FADHMAT)/(KmscadC4AcylCoAMAT*KmscadFAD)-C4EnoylCoAMAT*FADHMAT/(KmscadC4AcylCoAMAT*KmscadFAD*Keqscad))/((1+C4AcylCoAMAT/KmscadC4AcylCoAMAT+C4EnoylCoAMAT/KmscadC4EnoylCoAMAT+C6AcylCoAMAT/KmscadC6AcylCoAMAT+C6EnoylCoAMAT/KmscadC6EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmscadFAD+FADHMAT/KmscadFADH))
K1acesink=70.0 uM; Ksacesink=6000000.0 l per min per mgProteinReaction: AcetylCoAMAT => ; AcetylCoAMAT, AcetylCoAMAT, Rate Law: Ksacesink*(AcetylCoAMAT-K1acesink)
KmcactC6AcylCarMAT=15.0 uM; Vfcact = 0.42 uM per min per mgProtein; KmcactCarCYT = 130.0 uM; KmcactC6AcylCarCYT=15.0 uM; KmcactCarMAT = 130.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; KicactC6AcylCarCYT=56.0 uM; Vrcact = 0.42 uM per min per mgProteinReaction: C6AcylCarCYT => C6AcylCarMAT; CarMAT, CarCYT, C6AcylCarCYT, CarMAT, C6AcylCarMAT, CarCYT, C6AcylCarCYT, CarMAT, C6AcylCarMAT, CarCYT, Rate Law: Vfcact*(C6AcylCarCYT*CarMAT-C6AcylCarMAT*CarCYT/Keqcact)/(C6AcylCarCYT*CarMAT+KmcactCarMAT*C6AcylCarCYT+KmcactC6AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C6AcylCarMAT*(1+C6AcylCarCYT/KicactC6AcylCarCYT)+CarCYT*(KmcactC6AcylCarMAT+C6AcylCarMAT)))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; sfmckatC6=1.0 dimensionless; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C6KetoacylCoAMAT => C4AcylCoAMAT + AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C6KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C4AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, AcetylCoAMAT, C6KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C4AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC6*Vmckat*(C6KetoacylCoAMAT*CoAMAT/(KmmckatC6KetoacylCoAMAT*KmmckatCoAMAT)-C4AcylCoAMAT*AcetylCoAMAT/(KmmckatC6KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; sfcrotC14=0.2 dimensionless; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C14EnoylCoAMAT => C14HydroxyacylCoAMAT; C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C14HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C14HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC14*Vcrot*(C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT-C14HydroxyacylCoAMAT/(KmcrotC14EnoylCoAMAT*Keqcrot))/(1+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
Vfcact = 0.42 uM per min per mgProtein; KmcactCarCYT = 130.0 uM; KmcactC10AcylCarCYT=15.0 uM; KicactC10AcylCarCYT=56.0 uM; KmcactC10AcylCarMAT=15.0 uM; KmcactCarMAT = 130.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; Vrcact = 0.42 uM per min per mgProteinReaction: C10AcylCarCYT => C10AcylCarMAT; CarMAT, CarCYT, C10AcylCarCYT, CarMAT, C10AcylCarMAT, CarCYT, C10AcylCarCYT, CarMAT, C10AcylCarMAT, CarCYT, Rate Law: Vfcact*(C10AcylCarCYT*CarMAT-C10AcylCarMAT*CarCYT/Keqcact)/(C10AcylCarCYT*CarMAT+KmcactCarMAT*C10AcylCarCYT+KmcactC10AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C10AcylCarMAT*(1+C10AcylCarCYT/KicactC10AcylCarCYT)+CarCYT*(KmcactC10AcylCarMAT+C10AcylCarMAT)))
KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uM; sfmtpC14=0.9 dimensionlessReaction: C14EnoylCoAMAT => C12AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC14*Vmtp*(C14EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC14EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C12AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC14EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))
Vfcact = 0.42 uM per min per mgProtein; KmcactCarCYT = 130.0 uM; KmcactCarMAT = 130.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; KmcactC12AcylCarMAT=15.0 uM; KicactC12AcylCarCYT=56.0 uM; Vrcact = 0.42 uM per min per mgProtein; KmcactC12AcylCarCYT=15.0 uMReaction: C12AcylCarCYT => C12AcylCarMAT; CarMAT, CarCYT, C12AcylCarCYT, CarMAT, C12AcylCarMAT, CarCYT, C12AcylCarCYT, CarMAT, C12AcylCarMAT, CarCYT, Rate Law: Vfcact*(C12AcylCarCYT*CarMAT-C12AcylCarMAT*CarCYT/Keqcact)/(C12AcylCarCYT*CarMAT+KmcactCarMAT*C12AcylCarCYT+KmcactC12AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C12AcylCarMAT*(1+C12AcylCarCYT/KicactC12AcylCarCYT)+CarCYT*(KmcactC12AcylCarMAT+C12AcylCarMAT)))
KmmschadC16KetoacylCoAMAT = 1.4 uM; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; sfmschadC16=0.6 dimensionless; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C16HydroxyacylCoAMAT => C16KetoacylCoAMAT + NADHMAT; C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC16*Vmschad*(C16HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC16HydroxyacylCoAMAT*KmmschadNADMAT)-C16KetoacylCoAMAT*NADHMAT/(KmmschadC16HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; Keqcpt2 = 2.22 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; sfcpt2C10=0.95 dimensionless; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCarMAT = 51.0 uMReaction: C10AcylCarMAT => C10AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C10AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C10AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C10*Vcpt2*(C10AcylCarMAT*CoAMAT/(Kmcpt2C10AcylCarMAT*Kmcpt2CoAMAT)-C10AcylCoAMAT*CarMAT/(Kmcpt2C10AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; sfcrotC4=1.0 dimensionless; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C4EnoylCoAMAT => C4HydroxyacylCoAMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C4EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C4EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC4*Vcrot*(C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT-C4HydroxyacylCoAMAT/(KmcrotC4EnoylCoAMAT*Keqcrot))/(1+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
KmmschadC16KetoacylCoAMAT = 1.4 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; KmmschadNADMAT = 58.5 uM; sfmschadC10=0.64 dimensionless; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C10HydroxyacylCoAMAT => C10KetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C10HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C10KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, C10HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C10KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC10*Vmschad*(C10HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC10HydroxyacylCoAMAT*KmmschadNADMAT)-C10KetoacylCoAMAT*NADHMAT/(KmmschadC10HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
K1fadhsink=0.46 uM; Ksfadhsink=6000000.0 l per min per mgProteinReaction: FADHMAT => ; FADHMAT, FADHMAT, Rate Law: Ksfadhsink*(FADHMAT-K1fadhsink)
KmlcadC8AcylCoAMAT = 123.0 uM; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; sflcadC8=0.4 dimensionless; Keqlcad = 6.0 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C8AcylCoAMAT => C8EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, FADtMAT, C8EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, FADHMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, FADtMAT, C8EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, FADHMAT, Rate Law: sflcadC8*Vlcad*(C8AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC8AcylCoAMAT*KmlcadFAD)-C8EnoylCoAMAT*FADHMAT/(KmlcadC8AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
KmmschadC16KetoacylCoAMAT = 1.4 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; sfmschadC4=0.67 dimensionless; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C4HydroxyacylCoAMAT => C4AcetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, NADtMAT, C4AcetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, NADHMAT, C4HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, NADtMAT, C4AcetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC4*Vmschad*(C4HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC4HydroxyacylCoAMAT*KmmschadNADMAT)-C4AcetoacylCoAMAT*NADHMAT/(KmmschadC4HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; sfmckatC16=0.0 dimensionless; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C16KetoacylCoAMAT => C14AcylCoAMAT + AcetylCoAMAT; C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC16*Vmckat*(C16KetoacylCoAMAT*CoAMAT/(KmmckatC16KetoacylCoAMAT*KmmckatCoAMAT)-C14AcylCoAMAT*AcetylCoAMAT/(KmmckatC16KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmvlcadC14AcylCoAMAT = 4.0 uM; KmvlcadC14EnoylCoAMAT = 1.08 uM; KmvlcadFAD = 0.12 uM; KmvlcadFADH = 24.2 uM; KmvlcadC12AcylCoAMAT = 2.7 uM; KmvlcadC12EnoylCoAMAT = 1.08 uM; Vvlcad = 0.008 uM per min per mgProtein; KmvlcadC16EnoylCoAMAT = 1.08 uM; sfvlcadC12=0.11 dimensionless; KmvlcadC16AcylCoAMAT = 6.5 uM; Keqvlcad = 6.0 dimensionlessReaction: C12AcylCoAMAT => C12EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, FADHMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, FADHMAT, Rate Law: sfvlcadC12*Vvlcad*(C12AcylCoAMAT*(FADtMAT-FADHMAT)/(KmvlcadC12AcylCoAMAT*KmvlcadFAD)-C12EnoylCoAMAT*FADHMAT/(KmvlcadC12AcylCoAMAT*KmvlcadFAD*Keqvlcad))/((1+C12AcylCoAMAT/KmvlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmvlcadC12EnoylCoAMAT+C16AcylCoAMAT/KmvlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmvlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmvlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmvlcadC14EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmvlcadFAD+FADHMAT/KmvlcadFADH))
Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; sfcpt2C8=0.35 dimensionless; Keqcpt2 = 2.22 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCarMAT = 51.0 uMReaction: C8AcylCarMAT => C8AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C8AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C8AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C8*Vcpt2*(C8AcylCarMAT*CoAMAT/(Kmcpt2C8AcylCarMAT*Kmcpt2CoAMAT)-C8AcylCoAMAT*CarMAT/(Kmcpt2C8AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; sfcrotC16=0.13 dimensionless; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C16EnoylCoAMAT => C16HydroxyacylCoAMAT; C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC16*Vcrot*(C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT-C16HydroxyacylCoAMAT/(KmcrotC16EnoylCoAMAT*Keqcrot))/(1+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
KmvlcadC14AcylCoAMAT = 4.0 uM; KmvlcadC14EnoylCoAMAT = 1.08 uM; KmvlcadFAD = 0.12 uM; KmvlcadFADH = 24.2 uM; KmvlcadC12AcylCoAMAT = 2.7 uM; KmvlcadC12EnoylCoAMAT = 1.08 uM; Vvlcad = 0.008 uM per min per mgProtein; KmvlcadC16EnoylCoAMAT = 1.08 uM; KmvlcadC16AcylCoAMAT = 6.5 uM; sfvlcadC16=1.0 dimensionless; Keqvlcad = 6.0 dimensionlessReaction: C16AcylCoAMAT => C16EnoylCoAMAT + FADHMAT; C14AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, FADHMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, FADHMAT, Rate Law: sfvlcadC16*Vvlcad*(C16AcylCoAMAT*(FADtMAT-FADHMAT)/(KmvlcadC16AcylCoAMAT*KmvlcadFAD)-C16EnoylCoAMAT*FADHMAT/(KmvlcadC16AcylCoAMAT*KmvlcadFAD*Keqvlcad))/((1+C16AcylCoAMAT/KmvlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmvlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmvlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmvlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmvlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmvlcadC12EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmvlcadFAD+FADHMAT/KmvlcadFADH))
KmlcadC8AcylCoAMAT = 123.0 uM; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; Keqlcad = 6.0 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; sflcadC12=0.9 dimensionless; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C12AcylCoAMAT => C12EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, Rate Law: sflcadC12*Vlcad*(C12AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC12AcylCoAMAT*KmlcadFAD)-C14EnoylCoAMAT*FADHMAT/(KmlcadC12AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C14EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; Keqcpt2 = 2.22 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uM; sfcpt2C12=0.95 dimensionless; Kmcpt2C4AcylCarMAT = 51.0 uMReaction: C12AcylCarMAT => C12AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C12AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C12AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C12*Vcpt2*(C12AcylCarMAT*CoAMAT/(Kmcpt2C12AcylCarMAT*Kmcpt2CoAMAT)-C12AcylCoAMAT*CarMAT/(Kmcpt2C12AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uM; sfmtpC10=0.73 dimensionlessReaction: C10EnoylCoAMAT => C8AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C10EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, C10EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC10*Vmtp*(C10EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC10EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C8AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC10EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; sfcrotC8=0.58 dimensionless; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C8EnoylCoAMAT => C8HydroxyacylCoAMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C8EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C8HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C8EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C8HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC8*Vcrot*(C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT-C8HydroxyacylCoAMAT/(KmcrotC8EnoylCoAMAT*Keqcrot))/(1+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
KmmcadC8AcylCoAMAT = 4.0 uM; KmmcadC12AcylCoAMAT = 5.7 uM; KmmcadC4AcylCoAMAT = 135.0 uM; KmmcadC10AcylCoAMAT = 5.4 uM; KmmcadFAD = 0.12 uM; KmmcadC10EnoylCoAMAT = 1.08 uM; KmmcadC4EnoylCoAMAT = 1.08 uM; Vmcad = 0.081 uM per min per mgProtein; KmmcadC6AcylCoAMAT = 9.4 uM; KmmcadC6EnoylCoAMAT = 1.08 uM; KmmcadFADH = 24.2 uM; sfmcadC8=0.87 dimensionless; KmmcadC8EnoylCoAMAT = 1.08 uM; KmmcadC12EnoylCoAMAT = 1.08 uM; Keqmcad = 6.0 dimensionlessReaction: C8AcylCoAMAT => C8EnoylCoAMAT + FADHMAT; C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C8AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C8EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, C8AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C8EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: sfmcadC8*Vmcad*(C8AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC8AcylCoAMAT*KmmcadFAD)-C8EnoylCoAMAT*FADHMAT/(KmmcadC8AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; sfmckatC4=0.49 dimensionless; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C4AcetoacylCoAMAT => AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, CoAMAT, C4AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, CoAMAT, C4AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, CoAMAT, C4AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC4*Vmckat*(C4AcetoacylCoAMAT*CoAMAT/(KmmckatC4AcetoacylCoAMAT*KmmckatCoAMAT)-AcetylCoAMAT*AcetylCoAMAT/(KmmckatC4AcetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmcactC8AcylCarMAT=15.0 uM; Vfcact = 0.42 uM per min per mgProtein; KmcactCarCYT = 130.0 uM; KicactC8AcylCarCYT=56.0 uM; KmcactCarMAT = 130.0 uM; KmcactC8AcylCarCYT=15.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; Vrcact = 0.42 uM per min per mgProteinReaction: C8AcylCarCYT => C8AcylCarMAT; CarMAT, CarCYT, C8AcylCarCYT, CarMAT, C8AcylCarMAT, CarCYT, C8AcylCarCYT, CarMAT, C8AcylCarMAT, CarCYT, Rate Law: Vfcact*(C8AcylCarCYT*CarMAT-C8AcylCarMAT*CarCYT/Keqcact)/(C8AcylCarCYT*CarMAT+KmcactCarMAT*C8AcylCarCYT+KmcactC8AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C8AcylCarMAT*(1+C8AcylCarCYT/KicactC8AcylCarCYT)+CarCYT*(KmcactC8AcylCarMAT+C8AcylCarMAT)))
KmmcadC12AcylCoAMAT = 5.7 uM; KmmcadC8AcylCoAMAT = 4.0 uM; KmmcadC4AcylCoAMAT = 135.0 uM; KmmcadC10AcylCoAMAT = 5.4 uM; KmmcadFAD = 0.12 uM; KmmcadC10EnoylCoAMAT = 1.08 uM; KmmcadC4EnoylCoAMAT = 1.08 uM; Vmcad = 0.081 uM per min per mgProtein; KmmcadC6AcylCoAMAT = 9.4 uM; KmmcadC6EnoylCoAMAT = 1.08 uM; sfmcadC12=0.38 dimensionless; KmmcadFADH = 24.2 uM; KmmcadC12EnoylCoAMAT = 1.08 uM; KmmcadC8EnoylCoAMAT = 1.08 uM; Keqmcad = 6.0 dimensionlessReaction: C12AcylCoAMAT => C12EnoylCoAMAT + FADHMAT; C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: sfmcadC12*Vmcad*(C12AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC12AcylCoAMAT*KmmcadFAD)-C12EnoylCoAMAT*FADHMAT/(KmmcadC12AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; sfmckatC14=0.2 dimensionless; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C14KetoacylCoAMAT => C12AcylCoAMAT + AcetylCoAMAT; C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, C14KetoacylCoAMAT, C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, C14KetoacylCoAMAT, C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC14*Vmckat*(C14KetoacylCoAMAT*CoAMAT/(KmmckatC14KetoacylCoAMAT*KmmckatCoAMAT)-C12AcylCoAMAT*AcetylCoAMAT/(KmmckatC14KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmmschadC16KetoacylCoAMAT = 1.4 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; sfmschadC14=0.5 dimensionless; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C14HydroxyacylCoAMAT => C14KetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C14HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C14KetoacylCoAMAT, C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, C14HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C14KetoacylCoAMAT, C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC14*Vmschad*(C14HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC14HydroxyacylCoAMAT*KmmschadNADMAT)-C14KetoacylCoAMAT*NADHMAT/(KmmschadC14HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
KmcactC16AcylCarMAT=15.0 uM; Vfcact = 0.42 uM per min per mgProtein; KmcactCarCYT = 130.0 uM; KicactC16AcylCarCYT=56.0 uM; KmcactCarMAT = 130.0 uM; KmcactC16AcylCarCYT=15.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; Vrcact = 0.42 uM per min per mgProteinReaction: C16AcylCarCYT => C16AcylCarMAT; CarMAT, CarCYT, C16AcylCarCYT, CarMAT, C16AcylCarMAT, CarCYT, C16AcylCarCYT, CarMAT, C16AcylCarMAT, CarCYT, Rate Law: Vfcact*(C16AcylCarCYT*CarMAT-C16AcylCarMAT*CarCYT/Keqcact)/(C16AcylCarCYT*CarMAT+KmcactCarMAT*C16AcylCarCYT+KmcactC16AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C16AcylCarMAT*(1+C16AcylCarCYT/KicactC16AcylCarCYT)+CarCYT*(KmcactC16AcylCarMAT+C16AcylCarMAT)))
KmlcadC8AcylCoAMAT = 123.0 uM; sflcadC14=1.0 dimensionless; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; Keqlcad = 6.0 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C14AcylCoAMAT => C14EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, Rate Law: sflcadC14*Vlcad*(C14AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC14AcylCoAMAT*KmlcadFAD)-C14EnoylCoAMAT*FADHMAT/(KmlcadC14AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
KmlcadC8AcylCoAMAT = 123.0 uM; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; sflcadC16=0.9 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; Keqlcad = 6.0 dimensionless; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C16AcylCoAMAT => C16EnoylCoAMAT + FADHMAT; C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, Rate Law: sflcadC16*Vlcad*(C16AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC16AcylCoAMAT*KmlcadFAD)-C16EnoylCoAMAT*FADHMAT/(KmlcadC16AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; sfcrotC12=0.25 dimensionless; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C12EnoylCoAMAT => C12HydroxyacylCoAMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C12EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C12HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C12EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C12HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC12*Vcrot*(C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT-C12HydroxyacylCoAMAT/(KmcrotC12EnoylCoAMAT*Keqcrot))/(1+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
sfmtpC8=0.34 dimensionless; KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uMReaction: C8EnoylCoAMAT => C6AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcetoacylCoAMAT, C8EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, NADtMAT, CoAMAT, C6AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, C8EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, NADtMAT, CoAMAT, C6AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC8*Vmtp*(C8EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC8EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C6AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC8EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))
KmmschadC16KetoacylCoAMAT = 1.4 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; sfmschadC12=0.43 dimensionless; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C12HydroxyacylCoAMAT => C12KetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C12HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C12KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, C12HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C12KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC12*Vmschad*(C12HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC12HydroxyacylCoAMAT*KmmschadNADMAT)-C12KetoacylCoAMAT*NADHMAT/(KmmschadC12HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; sfmtpC12=0.81 dimensionless; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uMReaction: C12EnoylCoAMAT => C10AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C12EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, C12EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC12*Vmtp*(C12EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC12EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C10AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC12EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))
KmmcadC12AcylCoAMAT = 5.7 uM; KmmcadC8AcylCoAMAT = 4.0 uM; KmmcadC4AcylCoAMAT = 135.0 uM; KmmcadC10AcylCoAMAT = 5.4 uM; KmmcadFAD = 0.12 uM; KmmcadC10EnoylCoAMAT = 1.08 uM; KmmcadC4EnoylCoAMAT = 1.08 uM; Vmcad = 0.081 uM per min per mgProtein; KmmcadC6AcylCoAMAT = 9.4 uM; KmmcadC6EnoylCoAMAT = 1.08 uM; KmmcadFADH = 24.2 uM; KmmcadC12EnoylCoAMAT = 1.08 uM; KmmcadC8EnoylCoAMAT = 1.08 uM; sfmcadC4=0.12 dimensionless; Keqmcad = 6.0 dimensionlessReaction: C4AcylCoAMAT => C4EnoylCoAMAT + FADHMAT; C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, FADtMAT, C4EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, FADHMAT, C4AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, FADtMAT, C4EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, FADHMAT, Rate Law: sfmcadC4*Vmcad*(C4AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC4AcylCoAMAT*KmmcadFAD)-C4EnoylCoAMAT*FADHMAT/(KmmcadC4AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))
Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; sfcpt2C14=1.0 dimensionless; Keqcpt2 = 2.22 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uMReaction: C14AcylCarMAT => C14AcylCoAMAT; C16AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C14AcylCarMAT, C16AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C14AcylCarMAT, C16AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C14*Vcpt2*(C14AcylCarMAT*CoAMAT/(Kmcpt2C14AcylCarMAT*Kmcpt2CoAMAT)-C14AcylCoAMAT*CarMAT/(Kmcpt2C14AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCoAMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; KmmckatC12KetoacylCoAMAT = 1.3 uM; Keqmckat = 1051.0 dimensionless; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; sfmckatC12=0.38 dimensionless; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C12KetoacylCoAMAT => C10AcylCoAMAT + AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, C12KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, C12KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC12*Vmckat*(C12KetoacylCoAMAT*CoAMAT/(KmmckatC12KetoacylCoAMAT*KmmckatCoAMAT)-C10AcylCoAMAT*AcetylCoAMAT/(KmmckatC12KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmcactC14AcylCarMAT=15.0 uM; KmcactC14AcylCarCYT=15.0 uM; Vfcact = 0.42 uM per min per mgProtein; KicactC14AcylCarCYT=56.0 uM; KmcactCarCYT = 130.0 uM; KmcactCarMAT = 130.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; Vrcact = 0.42 uM per min per mgProteinReaction: C14AcylCarCYT => C14AcylCarMAT; CarMAT, CarCYT, C14AcylCarCYT, CarMAT, C14AcylCarMAT, CarCYT, C14AcylCarCYT, CarMAT, C14AcylCarMAT, CarCYT, Rate Law: Vfcact*(C14AcylCarCYT*CarMAT-C14AcylCarMAT*CarCYT/Keqcact)/(C14AcylCarCYT*CarMAT+KmcactCarMAT*C14AcylCarCYT+KmcactC14AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C14AcylCarMAT*(1+C14AcylCarCYT/KicactC14AcylCarCYT)+CarCYT*(KmcactC14AcylCarMAT+C14AcylCarMAT)))
KmscadFAD = 0.12 uM; KmscadC4EnoylCoAMAT = 1.08 uM; Vscad = 0.081 uM per min per mgProtein; KmscadFADH = 24.2 uM; KmscadC4AcylCoAMAT = 10.7 uM; KmscadC6AcylCoAMAT = 285.0 uM; KmscadC6EnoylCoAMAT = 1.08 uM; sfscadC6=0.3 dimensionless; Keqscad = 6.0 dimensionlessReaction: C6AcylCoAMAT => C6EnoylCoAMAT + FADHMAT; C4AcylCoAMAT, FADtMAT, C4EnoylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: sfscadC6*Vscad*(C6AcylCoAMAT*(FADtMAT-FADHMAT)/(KmscadC6AcylCoAMAT*KmscadFAD)-C6EnoylCoAMAT*FADHMAT/(KmscadC6AcylCoAMAT*KmscadFAD*Keqscad))/((1+C6AcylCoAMAT/KmscadC6AcylCoAMAT+C6EnoylCoAMAT/KmscadC6EnoylCoAMAT+C4AcylCoAMAT/KmscadC4AcylCoAMAT+C4EnoylCoAMAT/KmscadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmscadFAD+FADHMAT/KmscadFADH))
KmmschadC16KetoacylCoAMAT = 1.4 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; sfmschadC8=0.89 dimensionless; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C8HydroxyacylCoAMAT => C8KetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C8HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C8KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, C8HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C8KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC8*Vmschad*(C8HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC8HydroxyacylCoAMAT*KmmschadNADMAT)-C8KetoacylCoAMAT*NADHMAT/(KmmschadC8HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; sfcrotC10=0.33 dimensionless; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C10EnoylCoAMAT => C10HydroxyacylCoAMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C10EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C10HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C10EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C10HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC10*Vcrot*(C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT-C10HydroxyacylCoAMAT/(KmcrotC10EnoylCoAMAT*Keqcrot))/(1+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; Keqcpt2 = 2.22 dimensionless; sfcpt2C16=0.85 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCarMAT = 51.0 uMReaction: C16AcylCarMAT => C16AcylCoAMAT; C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C16*Vcpt2*(C16AcylCarMAT*CoAMAT/(Kmcpt2C16AcylCarMAT*Kmcpt2CoAMAT)-C16AcylCoAMAT*CarMAT/(Kmcpt2C16AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uM; sfmtpC16=1.0 dimensionlessReaction: C16EnoylCoAMAT => C14AcylCoAMAT + AcetylCoAMAT + NADHMAT; C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC16*Vmtp*(C16EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC16EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C14AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC16EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))

States:

NameDescription
C12AcylCarCYT[O-acylcarnitine]
C8HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C14KetoacylCoAMAT[fatty acyl-CoA]
C10AcylCarCYT[O-acylcarnitine]
C10EnoylCoAMAT[cis-2-enoyl-CoA]
C12HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C14EnoylCoAMAT[cis-2-enoyl-CoA]
C12AcylCoAMAT[fatty acyl-CoA]
C8KetoacylCoAMAT[fatty acyl-CoA]
C6AcylCarMAT[O-acylcarnitine]
C10AcylCarMAT[O-acylcarnitine]
C4HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C14HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C8AcylCoAMAT[fatty acyl-CoA]
C4AcylCoAMAT[fatty acyl-CoA]
C4EnoylCoAMAT[cis-2-enoyl-CoA]
C12KetoacylCoAMAT[fatty acyl-CoA]
C16HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
NADHMAT[NADH]
C6AcylCarCYT[O-acylcarnitine]
C8AcylCarCYT[O-acylcarnitine]
C14AcylCarCYT[O-acylcarnitine]
C10KetoacylCoAMAT[fatty acyl-CoA]
C16AcylCarMAT[O-acylcarnitine]
C10AcylCoAMAT[fatty acyl-CoA]
C16EnoylCoAMAT[cis-2-enoyl-CoA]
FADHMAT[FADH(.)]
C14AcylCoAMAT[fatty acyl-CoA]
AcetylCoAMAT[acetyl-CoA]
C16AcylCoAMAT[palmitoyl-CoA; fatty acyl-CoA]
C16KetoacylCoAMAT[fatty acyl-CoA]
C12EnoylCoAMAT[cis-2-enoyl-CoA]
C8AcylCarMAT[O-acylcarnitine]
C10HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C12AcylCarMAT[O-acylcarnitine]
C4AcetoacylCoAMAT[acetoacetyl-CoA]
C8EnoylCoAMAT[cis-2-enoyl-CoA]

vanEunen2013 - Network dynamics of fatty acid β-oxidation (time-course model): BIOMD0000000506v0.0.1

vanEunen2013 - Network dynamics of fatty acid β-oxidation (time-course model)Lipid metabolism plays an important role in…

Details

Fatty-acid metabolism plays a key role in acquired and inborn metabolic diseases. To obtain insight into the network dynamics of fatty-acid β-oxidation, we constructed a detailed computational model of the pathway and subjected it to a fat overload condition. The model contains reversible and saturable enzyme-kinetic equations and experimentally determined parameters for rat-liver enzymes. It was validated by adding palmitoyl CoA or palmitoyl carnitine to isolated rat-liver mitochondria: without refitting of measured parameters, the model correctly predicted the β-oxidation flux as well as the time profiles of most acyl-carnitine concentrations. Subsequently, we simulated the condition of obesity by increasing the palmitoyl-CoA concentration. At a high concentration of palmitoyl CoA the β-oxidation became overloaded: the flux dropped and metabolites accumulated. This behavior originated from the competition between acyl CoAs of different chain lengths for a set of acyl-CoA dehydrogenases with overlapping substrate specificity. This effectively induced competitive feedforward inhibition and thereby led to accumulation of CoA-ester intermediates and depletion of free CoA (CoASH). The mitochondrial [NAD⁺]/[NADH] ratio modulated the sensitivity to substrate overload, revealing a tight interplay between regulation of β-oxidation and mitochondrial respiration. link: http://identifiers.org/pubmed/23966849

Parameters:

NameDescription
KmvlcadC14AcylCoAMAT = 4.0 uM; KmvlcadC14EnoylCoAMAT = 1.08 uM; KmvlcadFAD = 0.12 uM; KmvlcadFADH = 24.2 uM; KmvlcadC12AcylCoAMAT = 2.7 uM; sfvlcadC14=0.42 dimensionless; KmvlcadC12EnoylCoAMAT = 1.08 uM; Vvlcad = 0.008 uM per min per mgProtein; KmvlcadC16EnoylCoAMAT = 1.08 uM; KmvlcadC16AcylCoAMAT = 6.5 uM; Keqvlcad = 6.0 dimensionlessReaction: C14AcylCoAMAT => C14EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, FADHMAT, Rate Law: sfvlcadC14*Vvlcad*(C14AcylCoAMAT*(FADtMAT-FADHMAT)/(KmvlcadC14AcylCoAMAT*KmvlcadFAD)-C14EnoylCoAMAT*FADHMAT/(KmvlcadC14AcylCoAMAT*KmvlcadFAD*Keqvlcad))/((1+C14AcylCoAMAT/KmvlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmvlcadC14EnoylCoAMAT+C16AcylCoAMAT/KmvlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmvlcadC16EnoylCoAMAT+C12AcylCoAMAT/KmvlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmvlcadC12EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmvlcadFAD+FADHMAT/KmvlcadFADH))
KmlcadC8AcylCoAMAT = 123.0 uM; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; sflcadC10=0.75 dimensionless; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; Keqlcad = 6.0 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C10AcylCoAMAT => C10EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C10EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, Rate Law: sflcadC10*Vlcad*(C10AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC10AcylCoAMAT*KmlcadFAD)-C10EnoylCoAMAT*FADHMAT/(KmlcadC10AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
KmmcadC12AcylCoAMAT = 5.7 uM; KmmcadC8AcylCoAMAT = 4.0 uM; KmmcadC4AcylCoAMAT = 135.0 uM; KmmcadC10AcylCoAMAT = 5.4 uM; KmmcadFAD = 0.12 uM; KmmcadC10EnoylCoAMAT = 1.08 uM; KmmcadC4EnoylCoAMAT = 1.08 uM; Vmcad = 0.081 uM per min per mgProtein; KmmcadC6AcylCoAMAT = 9.4 uM; sfmcadC10=0.8 dimensionless; KmmcadC6EnoylCoAMAT = 1.08 uM; KmmcadFADH = 24.2 uM; KmmcadC12EnoylCoAMAT = 1.08 uM; KmmcadC8EnoylCoAMAT = 1.08 uM; Keqmcad = 6.0 dimensionlessReaction: C10AcylCoAMAT => C10EnoylCoAMAT + FADHMAT; C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C10AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C10EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: sfmcadC10*Vmcad*(C10AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC10AcylCoAMAT*KmmcadFAD)-C10EnoylCoAMAT*FADHMAT/(KmmcadC10AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))
sfmckatC10=0.65 dimensionless; KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C10KetoacylCoAMAT => C8AcylCoAMAT + AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, C10KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC10*Vmckat*(C10KetoacylCoAMAT*CoAMAT/(KmmckatC10KetoacylCoAMAT*KmmckatCoAMAT)-C8AcylCoAMAT*AcetylCoAMAT/(KmmckatC10KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmscadFAD = 0.12 uM; KmscadC4EnoylCoAMAT = 1.08 uM; sfscadC4=1.0 dimensionless; Vscad = 0.081 uM per min per mgProtein; KmscadFADH = 24.2 uM; KmscadC4AcylCoAMAT = 10.7 uM; KmscadC6AcylCoAMAT = 285.0 uM; KmscadC6EnoylCoAMAT = 1.08 uM; Keqscad = 6.0 dimensionlessReaction: C4AcylCoAMAT => C4EnoylCoAMAT + FADHMAT; C6AcylCoAMAT, FADtMAT, C6EnoylCoAMAT, C4AcylCoAMAT, C6AcylCoAMAT, FADtMAT, C4EnoylCoAMAT, C6EnoylCoAMAT, FADHMAT, Rate Law: sfscadC4*Vscad*(C4AcylCoAMAT*(FADtMAT-FADHMAT)/(KmscadC4AcylCoAMAT*KmscadFAD)-C4EnoylCoAMAT*FADHMAT/(KmscadC4AcylCoAMAT*KmscadFAD*Keqscad))/((1+C4AcylCoAMAT/KmscadC4AcylCoAMAT+C4EnoylCoAMAT/KmscadC4EnoylCoAMAT+C6AcylCoAMAT/KmscadC6AcylCoAMAT+C6EnoylCoAMAT/KmscadC6EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmscadFAD+FADHMAT/KmscadFADH))
KmcactC6AcylCarMAT=15.0 uM; Vfcact = 0.42 uM per min per mgProtein; KmcactCarCYT = 130.0 uM; KmcactC6AcylCarCYT=15.0 uM; KmcactCarMAT = 130.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; KicactC6AcylCarCYT=56.0 uM; Vrcact = 0.42 uM per min per mgProteinReaction: C6AcylCarCYT => C6AcylCarMAT; CarMAT, CarCYT, C6AcylCarCYT, CarMAT, C6AcylCarMAT, CarCYT, Rate Law: Vfcact*(C6AcylCarCYT*CarMAT-C6AcylCarMAT*CarCYT/Keqcact)/(C6AcylCarCYT*CarMAT+KmcactCarMAT*C6AcylCarCYT+KmcactC6AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C6AcylCarMAT*(1+C6AcylCarCYT/KicactC6AcylCarCYT)+CarCYT*(KmcactC6AcylCarMAT+C6AcylCarMAT)))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; sfcrotC14=0.2 dimensionless; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C14EnoylCoAMAT => C14HydroxyacylCoAMAT; C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C14HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC14*Vcrot*(C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT-C14HydroxyacylCoAMAT/(KmcrotC14EnoylCoAMAT*Keqcrot))/(1+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uM; sfmtpC14=0.9 dimensionlessReaction: C14EnoylCoAMAT => C12AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC14*Vmtp*(C14EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC14EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C12AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC14EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))
Vfcact = 0.42 uM per min per mgProtein; KmcactCarCYT = 130.0 uM; KmcactCarMAT = 130.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; KmcactC12AcylCarMAT=15.0 uM; KicactC12AcylCarCYT=56.0 uM; Vrcact = 0.42 uM per min per mgProtein; KmcactC12AcylCarCYT=15.0 uMReaction: C12AcylCarCYT => C12AcylCarMAT; CarMAT, CarCYT, C12AcylCarCYT, CarMAT, C12AcylCarMAT, CarCYT, Rate Law: Vfcact*(C12AcylCarCYT*CarMAT-C12AcylCarMAT*CarCYT/Keqcact)/(C12AcylCarCYT*CarMAT+KmcactCarMAT*C12AcylCarCYT+KmcactC12AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C12AcylCarMAT*(1+C12AcylCarCYT/KicactC12AcylCarCYT)+CarCYT*(KmcactC12AcylCarMAT+C12AcylCarMAT)))
KmmschadC16KetoacylCoAMAT = 1.4 uM; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; sfmschadC16=0.6 dimensionless; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C16HydroxyacylCoAMAT => C16KetoacylCoAMAT + NADHMAT; C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC16*Vmschad*(C16HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC16HydroxyacylCoAMAT*KmmschadNADMAT)-C16KetoacylCoAMAT*NADHMAT/(KmmschadC16HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; Keqcpt2 = 2.22 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; sfcpt2C10=0.95 dimensionless; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCarMAT = 51.0 uMReaction: C10AcylCarMAT => C10AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C10AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C10*Vcpt2*(C10AcylCarMAT*CoAMAT/(Kmcpt2C10AcylCarMAT*Kmcpt2CoAMAT)-C10AcylCoAMAT*CarMAT/(Kmcpt2C10AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; Keqcpt2 = 2.22 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; sfcpt2C6=0.15 dimensionless; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCarMAT = 51.0 uMReaction: C6AcylCarMAT => C6AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcylCoAMAT, CarMAT, C6AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C4AcylCarMAT, CoAMAT, C6AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C6*Vcpt2*(C6AcylCarMAT*CoAMAT/(Kmcpt2C6AcylCarMAT*Kmcpt2CoAMAT)-C6AcylCoAMAT*CarMAT/(Kmcpt2C6AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; sfcrotC4=1.0 dimensionless; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C4EnoylCoAMAT => C4HydroxyacylCoAMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C4EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC4*Vcrot*(C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT-C4HydroxyacylCoAMAT/(KmcrotC4EnoylCoAMAT*Keqcrot))/(1+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
KmmschadC16KetoacylCoAMAT = 1.4 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; KmmschadNADMAT = 58.5 uM; sfmschadC10=0.64 dimensionless; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C10HydroxyacylCoAMAT => C10KetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C10HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C10KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC10*Vmschad*(C10HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC10HydroxyacylCoAMAT*KmmschadNADMAT)-C10KetoacylCoAMAT*NADHMAT/(KmmschadC10HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
Kmcpt1C16AcylCoACYT=13.8 uM; Kmcpt1CarCYT=125.0 uM; ncpt1=2.4799 dimensionless; Kicpt1MalCoACYT=9.1 uM; Kmcpt1CoACYT=40.7 uM; Vcpt1=0.012 uM per min per mgProtein; sfcpt1C16=1.0 dimensionless; Keqcpt1=0.45 dimensionless; Kmcpt1C16AcylCarCYT=136.0 uMReaction: => C16AcylCarCYT; C16AcylCoACYT, CarCYT, CoACYT, MalCoACYT, C16AcylCoACYT, CarCYT, C16AcylCarCYT, CoACYT, MalCoACYT, Rate Law: sfcpt1C16*Vcpt1*(C16AcylCoACYT*CarCYT/(Kmcpt1C16AcylCoACYT*Kmcpt1CarCYT)-C16AcylCarCYT*CoACYT/(Kmcpt1C16AcylCoACYT*Kmcpt1CarCYT*Keqcpt1))/((1+C16AcylCoACYT/Kmcpt1C16AcylCoACYT+C16AcylCarCYT/Kmcpt1C16AcylCarCYT+(MalCoACYT/Kicpt1MalCoACYT)^ncpt1)*(1+CarCYT/Kmcpt1CarCYT+CoACYT/Kmcpt1CoACYT))
KmmschadC16KetoacylCoAMAT = 1.4 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; sfmschadC4=0.67 dimensionless; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C4HydroxyacylCoAMAT => C4AcetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, NADtMAT, C4AcetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC4*Vmschad*(C4HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC4HydroxyacylCoAMAT*KmmschadNADMAT)-C4AcetoacylCoAMAT*NADHMAT/(KmmschadC4HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
K1fadhsink=0.46 uM; Ksfadhsink=6000000.0 l per min per mgProteinReaction: FADHMAT => ; FADHMAT, Rate Law: Ksfadhsink*(FADHMAT-K1fadhsink)
KmlcadC8AcylCoAMAT = 123.0 uM; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; sflcadC8=0.4 dimensionless; Keqlcad = 6.0 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C8AcylCoAMAT => C8EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, FADtMAT, C8EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, FADHMAT, Rate Law: sflcadC8*Vlcad*(C8AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC8AcylCoAMAT*KmlcadFAD)-C8EnoylCoAMAT*FADHMAT/(KmlcadC8AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; sfmckatC16=0.0 dimensionless; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C16KetoacylCoAMAT => C14AcylCoAMAT + AcetylCoAMAT; C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC16*Vmckat*(C16KetoacylCoAMAT*CoAMAT/(KmmckatC16KetoacylCoAMAT*KmmckatCoAMAT)-C14AcylCoAMAT*AcetylCoAMAT/(KmmckatC16KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmvlcadC14AcylCoAMAT = 4.0 uM; KmvlcadC14EnoylCoAMAT = 1.08 uM; KmvlcadFAD = 0.12 uM; KmvlcadFADH = 24.2 uM; KmvlcadC12AcylCoAMAT = 2.7 uM; KmvlcadC12EnoylCoAMAT = 1.08 uM; Vvlcad = 0.008 uM per min per mgProtein; KmvlcadC16EnoylCoAMAT = 1.08 uM; sfvlcadC12=0.11 dimensionless; KmvlcadC16AcylCoAMAT = 6.5 uM; Keqvlcad = 6.0 dimensionlessReaction: C12AcylCoAMAT => C12EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, FADHMAT, Rate Law: sfvlcadC12*Vvlcad*(C12AcylCoAMAT*(FADtMAT-FADHMAT)/(KmvlcadC12AcylCoAMAT*KmvlcadFAD)-C12EnoylCoAMAT*FADHMAT/(KmvlcadC12AcylCoAMAT*KmvlcadFAD*Keqvlcad))/((1+C12AcylCoAMAT/KmvlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmvlcadC12EnoylCoAMAT+C16AcylCoAMAT/KmvlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmvlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmvlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmvlcadC14EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmvlcadFAD+FADHMAT/KmvlcadFADH))
Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; sfcpt2C8=0.35 dimensionless; Keqcpt2 = 2.22 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCarMAT = 51.0 uMReaction: C8AcylCarMAT => C8AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C8AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C8*Vcpt2*(C8AcylCarMAT*CoAMAT/(Kmcpt2C8AcylCarMAT*Kmcpt2CoAMAT)-C8AcylCoAMAT*CarMAT/(Kmcpt2C8AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; sfcrotC16=0.13 dimensionless; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C16EnoylCoAMAT => C16HydroxyacylCoAMAT; C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC16*Vcrot*(C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT-C16HydroxyacylCoAMAT/(KmcrotC16EnoylCoAMAT*Keqcrot))/(1+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
KmvlcadC14AcylCoAMAT = 4.0 uM; KmvlcadC14EnoylCoAMAT = 1.08 uM; KmvlcadFAD = 0.12 uM; KmvlcadFADH = 24.2 uM; KmvlcadC12AcylCoAMAT = 2.7 uM; KmvlcadC12EnoylCoAMAT = 1.08 uM; Vvlcad = 0.008 uM per min per mgProtein; KmvlcadC16EnoylCoAMAT = 1.08 uM; KmvlcadC16AcylCoAMAT = 6.5 uM; sfvlcadC16=1.0 dimensionless; Keqvlcad = 6.0 dimensionlessReaction: C16AcylCoAMAT => C16EnoylCoAMAT + FADHMAT; C14AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, FADHMAT, Rate Law: sfvlcadC16*Vvlcad*(C16AcylCoAMAT*(FADtMAT-FADHMAT)/(KmvlcadC16AcylCoAMAT*KmvlcadFAD)-C16EnoylCoAMAT*FADHMAT/(KmvlcadC16AcylCoAMAT*KmvlcadFAD*Keqvlcad))/((1+C16AcylCoAMAT/KmvlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmvlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmvlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmvlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmvlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmvlcadC12EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmvlcadFAD+FADHMAT/KmvlcadFADH))
KmlcadC8AcylCoAMAT = 123.0 uM; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; Keqlcad = 6.0 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; sflcadC12=0.9 dimensionless; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C12AcylCoAMAT => C12EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, Rate Law: sflcadC12*Vlcad*(C12AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC12AcylCoAMAT*KmlcadFAD)-C14EnoylCoAMAT*FADHMAT/(KmlcadC12AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C14EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; Keqcpt2 = 2.22 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uM; sfcpt2C12=0.95 dimensionless; Kmcpt2C4AcylCarMAT = 51.0 uMReaction: C12AcylCarMAT => C12AcylCoAMAT; C16AcylCarMAT, C14AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C12AcylCarMAT, C16AcylCarMAT, C14AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C12*Vcpt2*(C12AcylCarMAT*CoAMAT/(Kmcpt2C12AcylCarMAT*Kmcpt2CoAMAT)-C12AcylCoAMAT*CarMAT/(Kmcpt2C12AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uM; sfmtpC10=0.73 dimensionlessReaction: C10EnoylCoAMAT => C8AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C10EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C8AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC10*Vmtp*(C10EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC10EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C8AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC10EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; sfcrotC8=0.58 dimensionless; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C8EnoylCoAMAT => C8HydroxyacylCoAMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C8EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C8HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC8*Vcrot*(C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT-C8HydroxyacylCoAMAT/(KmcrotC8EnoylCoAMAT*Keqcrot))/(1+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uM; sfcrotC6=0.83 dimensionlessReaction: C6EnoylCoAMAT => C6HydroxyacylCoAMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C6EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C4EnoylCoAMAT, C6HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC6*Vcrot*(C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT-C6HydroxyacylCoAMAT/(KmcrotC6EnoylCoAMAT*Keqcrot))/(1+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
sfmcadC6=1.0 dimensionless; KmmcadC12AcylCoAMAT = 5.7 uM; KmmcadC8AcylCoAMAT = 4.0 uM; KmmcadC4AcylCoAMAT = 135.0 uM; KmmcadC10AcylCoAMAT = 5.4 uM; KmmcadFAD = 0.12 uM; KmmcadC10EnoylCoAMAT = 1.08 uM; KmmcadC4EnoylCoAMAT = 1.08 uM; Vmcad = 0.081 uM per min per mgProtein; KmmcadC6AcylCoAMAT = 9.4 uM; KmmcadC6EnoylCoAMAT = 1.08 uM; KmmcadFADH = 24.2 uM; KmmcadC12EnoylCoAMAT = 1.08 uM; KmmcadC8EnoylCoAMAT = 1.08 uM; Keqmcad = 6.0 dimensionlessReaction: C6AcylCoAMAT => C6EnoylCoAMAT + FADHMAT; C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C4EnoylCoAMAT, C6AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C6EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: sfmcadC6*Vmcad*(C6AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC6AcylCoAMAT*KmmcadFAD)-C6EnoylCoAMAT*FADHMAT/(KmmcadC6AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))
KmmcadC8AcylCoAMAT = 4.0 uM; KmmcadC12AcylCoAMAT = 5.7 uM; KmmcadC4AcylCoAMAT = 135.0 uM; KmmcadC10AcylCoAMAT = 5.4 uM; KmmcadFAD = 0.12 uM; KmmcadC10EnoylCoAMAT = 1.08 uM; KmmcadC4EnoylCoAMAT = 1.08 uM; Vmcad = 0.081 uM per min per mgProtein; KmmcadC6AcylCoAMAT = 9.4 uM; KmmcadC6EnoylCoAMAT = 1.08 uM; KmmcadFADH = 24.2 uM; sfmcadC8=0.87 dimensionless; KmmcadC8EnoylCoAMAT = 1.08 uM; KmmcadC12EnoylCoAMAT = 1.08 uM; Keqmcad = 6.0 dimensionlessReaction: C8AcylCoAMAT => C8EnoylCoAMAT + FADHMAT; C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C8AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C8EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: sfmcadC8*Vmcad*(C8AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC8AcylCoAMAT*KmmcadFAD)-C8EnoylCoAMAT*FADHMAT/(KmmcadC8AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; sfmckatC4=0.49 dimensionless; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C4AcetoacylCoAMAT => AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, CoAMAT, C4AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, CoAMAT, C4AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC4*Vmckat*(C4AcetoacylCoAMAT*CoAMAT/(KmmckatC4AcetoacylCoAMAT*KmmckatCoAMAT)-AcetylCoAMAT*AcetylCoAMAT/(KmmckatC4AcetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmcactC8AcylCarMAT=15.0 uM; Vfcact = 0.42 uM per min per mgProtein; KmcactCarCYT = 130.0 uM; KicactC8AcylCarCYT=56.0 uM; KmcactCarMAT = 130.0 uM; KmcactC8AcylCarCYT=15.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; Vrcact = 0.42 uM per min per mgProteinReaction: C8AcylCarCYT => C8AcylCarMAT; CarMAT, CarCYT, C8AcylCarCYT, CarMAT, C8AcylCarMAT, CarCYT, Rate Law: Vfcact*(C8AcylCarCYT*CarMAT-C8AcylCarMAT*CarCYT/Keqcact)/(C8AcylCarCYT*CarMAT+KmcactCarMAT*C8AcylCarCYT+KmcactC8AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C8AcylCarMAT*(1+C8AcylCarCYT/KicactC8AcylCarCYT)+CarCYT*(KmcactC8AcylCarMAT+C8AcylCarMAT)))
KmmcadC12AcylCoAMAT = 5.7 uM; KmmcadC8AcylCoAMAT = 4.0 uM; KmmcadC4AcylCoAMAT = 135.0 uM; KmmcadC10AcylCoAMAT = 5.4 uM; KmmcadFAD = 0.12 uM; KmmcadC10EnoylCoAMAT = 1.08 uM; KmmcadC4EnoylCoAMAT = 1.08 uM; Vmcad = 0.081 uM per min per mgProtein; KmmcadC6AcylCoAMAT = 9.4 uM; KmmcadC6EnoylCoAMAT = 1.08 uM; sfmcadC12=0.38 dimensionless; KmmcadFADH = 24.2 uM; KmmcadC12EnoylCoAMAT = 1.08 uM; KmmcadC8EnoylCoAMAT = 1.08 uM; Keqmcad = 6.0 dimensionlessReaction: C12AcylCoAMAT => C12EnoylCoAMAT + FADHMAT; C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: sfmcadC12*Vmcad*(C12AcylCoAMAT*(FADtMAT-FADHMAT)/(KmmcadC12AcylCoAMAT*KmmcadFAD)-C12EnoylCoAMAT*FADHMAT/(KmmcadC12AcylCoAMAT*KmmcadFAD*Keqmcad))/((1+C12AcylCoAMAT/KmmcadC12AcylCoAMAT+C12EnoylCoAMAT/KmmcadC12EnoylCoAMAT+C10AcylCoAMAT/KmmcadC10AcylCoAMAT+C10EnoylCoAMAT/KmmcadC10EnoylCoAMAT+C8AcylCoAMAT/KmmcadC8AcylCoAMAT+C8EnoylCoAMAT/KmmcadC8EnoylCoAMAT+C6AcylCoAMAT/KmmcadC6AcylCoAMAT+C6EnoylCoAMAT/KmmcadC6EnoylCoAMAT+C4AcylCoAMAT/KmmcadC4AcylCoAMAT+C4EnoylCoAMAT/KmmcadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmmcadFAD+FADHMAT/KmmcadFADH))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; sfmckatC14=0.2 dimensionless; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C14KetoacylCoAMAT => C12AcylCoAMAT + AcetylCoAMAT; C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, C14KetoacylCoAMAT, C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C12AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC14*Vmckat*(C14KetoacylCoAMAT*CoAMAT/(KmmckatC14KetoacylCoAMAT*KmmckatCoAMAT)-C12AcylCoAMAT*AcetylCoAMAT/(KmmckatC14KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmmschadC16KetoacylCoAMAT = 1.4 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; sfmschadC14=0.5 dimensionless; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C14HydroxyacylCoAMAT => C14KetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C14HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C14KetoacylCoAMAT, C16KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC14*Vmschad*(C14HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC14HydroxyacylCoAMAT*KmmschadNADMAT)-C14KetoacylCoAMAT*NADHMAT/(KmmschadC14HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
KmcactC16AcylCarMAT=15.0 uM; Vfcact = 0.42 uM per min per mgProtein; KmcactCarCYT = 130.0 uM; KicactC16AcylCarCYT=56.0 uM; KmcactCarMAT = 130.0 uM; KmcactC16AcylCarCYT=15.0 uM; Keqcact = 1.0 dimensionless; KicactCarCYT = 200.0 uM; Vrcact = 0.42 uM per min per mgProteinReaction: C16AcylCarCYT => C16AcylCarMAT; CarMAT, CarCYT, C16AcylCarCYT, CarMAT, C16AcylCarMAT, CarCYT, Rate Law: Vfcact*(C16AcylCarCYT*CarMAT-C16AcylCarMAT*CarCYT/Keqcact)/(C16AcylCarCYT*CarMAT+KmcactCarMAT*C16AcylCarCYT+KmcactC16AcylCarCYT*CarMAT*(1+CarCYT/KicactCarCYT)+Vfcact/(Vrcact*Keqcact)*(KmcactCarCYT*C16AcylCarMAT*(1+C16AcylCarCYT/KicactC16AcylCarCYT)+CarCYT*(KmcactC16AcylCarMAT+C16AcylCarMAT)))
KmlcadC8AcylCoAMAT = 123.0 uM; sflcadC14=1.0 dimensionless; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; Keqlcad = 6.0 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C14AcylCoAMAT => C14EnoylCoAMAT + FADHMAT; C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C16EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, Rate Law: sflcadC14*Vlcad*(C14AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC14AcylCoAMAT*KmlcadFAD)-C14EnoylCoAMAT*FADHMAT/(KmlcadC14AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; sfcrotC12=0.25 dimensionless; KmcrotC10EnoylCoAMAT = 25.0 uM; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C12EnoylCoAMAT => C12HydroxyacylCoAMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C12EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C12HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC12*Vcrot*(C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT-C12HydroxyacylCoAMAT/(KmcrotC12EnoylCoAMAT*Keqcrot))/(1+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
KmlcadC8AcylCoAMAT = 123.0 uM; Vlcad = 0.01 uM per min per mgProtein; KmlcadC14EnoylCoAMAT = 1.08 uM; KmlcadFAD = 0.12 uM; KmlcadFADH = 24.2 uM; KmlcadC12EnoylCoAMAT = 1.08 uM; KmlcadC8EnoylCoAMAT = 1.08 uM; sflcadC16=0.9 dimensionless; KmlcadC16AcylCoAMAT = 2.5 uM; Keqlcad = 6.0 dimensionless; KmlcadC12AcylCoAMAT = 9.0 uM; KmlcadC10AcylCoAMAT = 24.3 uM; KmlcadC10EnoylCoAMAT = 1.08 uM; KmlcadC14AcylCoAMAT = 7.4 uM; KmlcadC16EnoylCoAMAT = 1.08 uMReaction: C16AcylCoAMAT => C16EnoylCoAMAT + FADHMAT; C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, FADtMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, FADHMAT, Rate Law: sflcadC16*Vlcad*(C16AcylCoAMAT*(FADtMAT-FADHMAT)/(KmlcadC16AcylCoAMAT*KmlcadFAD)-C16EnoylCoAMAT*FADHMAT/(KmlcadC16AcylCoAMAT*KmlcadFAD*Keqlcad))/((1+C16AcylCoAMAT/KmlcadC16AcylCoAMAT+C16EnoylCoAMAT/KmlcadC16EnoylCoAMAT+C14AcylCoAMAT/KmlcadC14AcylCoAMAT+C14EnoylCoAMAT/KmlcadC14EnoylCoAMAT+C12AcylCoAMAT/KmlcadC12AcylCoAMAT+C12EnoylCoAMAT/KmlcadC12EnoylCoAMAT+C10AcylCoAMAT/KmlcadC10AcylCoAMAT+C10EnoylCoAMAT/KmlcadC10EnoylCoAMAT+C8AcylCoAMAT/KmlcadC8AcylCoAMAT+C8EnoylCoAMAT/KmlcadC8EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmlcadFAD+FADHMAT/KmlcadFADH))
sfmtpC8=0.34 dimensionless; KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uMReaction: C8EnoylCoAMAT => C6AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcetoacylCoAMAT, C8EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, NADtMAT, CoAMAT, C6AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC8*Vmtp*(C8EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC8EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C6AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC8EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))
KmmschadC16KetoacylCoAMAT = 1.4 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; sfmschadC12=0.43 dimensionless; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C12HydroxyacylCoAMAT => C12KetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C12HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C12KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC12*Vmschad*(C12HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC12HydroxyacylCoAMAT*KmmschadNADMAT)-C12KetoacylCoAMAT*NADHMAT/(KmmschadC12HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; KmmckatC8KetoacylCoAMAT = 3.2 uM; Keqmckat = 1051.0 dimensionless; KmmckatC12KetoacylCoAMAT = 1.3 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; sfmckatC8=0.81 dimensionless; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C8KetoacylCoAMAT => C6AcylCoAMAT + AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcylCoAMAT, C8KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C6AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC8*Vmckat*(C8KetoacylCoAMAT*CoAMAT/(KmmckatC8KetoacylCoAMAT*KmmckatCoAMAT)-C6AcylCoAMAT*AcetylCoAMAT/(KmmckatC8KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; sfmtpC12=0.81 dimensionless; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uMReaction: C12EnoylCoAMAT => C10AcylCoAMAT + AcetylCoAMAT + NADHMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C12EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC12*Vmtp*(C12EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC12EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C10AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC12EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))
Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; sfcpt2C14=1.0 dimensionless; Keqcpt2 = 2.22 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uMReaction: C14AcylCarMAT => C14AcylCoAMAT; C16AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C14AcylCarMAT, C16AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C14*Vcpt2*(C14AcylCarMAT*CoAMAT/(Kmcpt2C14AcylCarMAT*Kmcpt2CoAMAT)-C14AcylCoAMAT*CarMAT/(Kmcpt2C14AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCoAMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmmckatC14AcylCoAMAT = 13.83 uM; KmmckatC10KetoacylCoAMAT = 2.1 uM; KmmckatCoAMAT = 26.6 uM; KmmckatC6KetoacylCoAMAT = 6.7 uM; KmmckatC12KetoacylCoAMAT = 1.3 uM; Keqmckat = 1051.0 dimensionless; KmmckatC8KetoacylCoAMAT = 3.2 uM; KmmckatC4AcetoacylCoAMAT = 12.4 uM; KmmckatC16KetoacylCoAMAT = 1.1 uM; KmmckatC12AcylCoAMAT = 13.83 uM; sfmckatC12=0.38 dimensionless; KmmckatC8AcylCoAMAT = 13.83 uM; KmmckatC10AcylCoAMAT = 13.83 uM; KmmckatC6AcylCoAMAT = 13.83 uM; Vmckat = 0.377 uM per min per mgProtein; KmmckatC16AcylCoAMAT = 13.83 uM; KmmckatC14KetoacylCoAMAT = 1.2 uM; KmmckatC4AcylCoAMAT = 13.83 uM; KmmckatAcetylCoAMAT = 30.0 uMReaction: C12KetoacylCoAMAT => C10AcylCoAMAT + AcetylCoAMAT; C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, C12KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, CoAMAT, C10AcylCoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, AcetylCoAMAT, Rate Law: sfmckatC12*Vmckat*(C12KetoacylCoAMAT*CoAMAT/(KmmckatC12KetoacylCoAMAT*KmmckatCoAMAT)-C10AcylCoAMAT*AcetylCoAMAT/(KmmckatC12KetoacylCoAMAT*KmmckatCoAMAT*Keqmckat))/((1+C12KetoacylCoAMAT/KmmckatC12KetoacylCoAMAT+C10AcylCoAMAT/KmmckatC10AcylCoAMAT+C16KetoacylCoAMAT/KmmckatC16KetoacylCoAMAT+C16AcylCoAMAT/KmmckatC16AcylCoAMAT+C14KetoacylCoAMAT/KmmckatC14KetoacylCoAMAT+C14AcylCoAMAT/KmmckatC14AcylCoAMAT+C10KetoacylCoAMAT/KmmckatC10KetoacylCoAMAT+C12AcylCoAMAT/KmmckatC12AcylCoAMAT+C8KetoacylCoAMAT/KmmckatC8KetoacylCoAMAT+C8AcylCoAMAT/KmmckatC8AcylCoAMAT+C6KetoacylCoAMAT/KmmckatC6KetoacylCoAMAT+C6AcylCoAMAT/KmmckatC6AcylCoAMAT+C4AcetoacylCoAMAT/KmmckatC4AcetoacylCoAMAT+C4AcylCoAMAT/KmmckatC4AcylCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT)*(1+CoAMAT/KmmckatCoAMAT+AcetylCoAMAT/KmmckatAcetylCoAMAT))
KmmschadC16KetoacylCoAMAT = 1.4 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; sfmschadC6=1.0 dimensionless; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C6HydroxyacylCoAMAT => C6KetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C4AcetoacylCoAMAT, C6HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C6KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C8KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC6*Vmschad*(C6HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC6HydroxyacylCoAMAT*KmmschadNADMAT)-C6KetoacylCoAMAT*NADHMAT/(KmmschadC6HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
KmscadFAD = 0.12 uM; KmscadC4EnoylCoAMAT = 1.08 uM; Vscad = 0.081 uM per min per mgProtein; KmscadFADH = 24.2 uM; KmscadC4AcylCoAMAT = 10.7 uM; KmscadC6AcylCoAMAT = 285.0 uM; KmscadC6EnoylCoAMAT = 1.08 uM; sfscadC6=0.3 dimensionless; Keqscad = 6.0 dimensionlessReaction: C6AcylCoAMAT => C6EnoylCoAMAT + FADHMAT; C4AcylCoAMAT, FADtMAT, C4EnoylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, FADtMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, FADHMAT, Rate Law: sfscadC6*Vscad*(C6AcylCoAMAT*(FADtMAT-FADHMAT)/(KmscadC6AcylCoAMAT*KmscadFAD)-C6EnoylCoAMAT*FADHMAT/(KmscadC6AcylCoAMAT*KmscadFAD*Keqscad))/((1+C6AcylCoAMAT/KmscadC6AcylCoAMAT+C6EnoylCoAMAT/KmscadC6EnoylCoAMAT+C4AcylCoAMAT/KmscadC4AcylCoAMAT+C4EnoylCoAMAT/KmscadC4EnoylCoAMAT)*(1+(FADtMAT-FADHMAT)/KmscadFAD+FADHMAT/KmscadFADH))
KmmschadC16KetoacylCoAMAT = 1.4 uM; Keqmschad = 2.17E-4 dimensionless; KmmschadC16HydroxyacylCoAMAT = 1.5 uM; KmmschadC14HydroxyacylCoAMAT = 1.8 uM; Vmschad = 1.0 uM per min per mgProtein; KmmschadC6HydroxyacylCoAMAT = 28.6 uM; KmmschadC12KetoacylCoAMAT = 1.6 uM; KmmschadC4HydroxyacylCoAMAT = 69.9 uM; KmmschadC14KetoacylCoAMAT = 1.4 uM; KmmschadC12HydroxyacylCoAMAT = 3.7 uM; KmmschadC6KetoacylCoAMAT = 5.8 uM; KmmschadNADMAT = 58.5 uM; KmmschadC10HydroxyacylCoAMAT = 8.8 uM; KmmschadC8HydroxyacylCoAMAT = 16.3 uM; KmmschadC4AcetoacylCoAMAT = 16.9 uM; KmmschadC10KetoacylCoAMAT = 2.3 uM; KmmschadNADHMAT = 5.4 uM; sfmschadC8=0.89 dimensionless; KmmschadC8KetoacylCoAMAT = 4.1 uMReaction: C8HydroxyacylCoAMAT => C8KetoacylCoAMAT + NADHMAT; C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, C8HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C10HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, NADtMAT, C8KetoacylCoAMAT, C16KetoacylCoAMAT, C14KetoacylCoAMAT, C12KetoacylCoAMAT, C10KetoacylCoAMAT, C6KetoacylCoAMAT, C4AcetoacylCoAMAT, NADHMAT, Rate Law: sfmschadC8*Vmschad*(C8HydroxyacylCoAMAT*(NADtMAT-NADHMAT)/(KmmschadC8HydroxyacylCoAMAT*KmmschadNADMAT)-C8KetoacylCoAMAT*NADHMAT/(KmmschadC8HydroxyacylCoAMAT*KmmschadNADMAT*Keqmschad))/((1+C8HydroxyacylCoAMAT/KmmschadC8HydroxyacylCoAMAT+C8KetoacylCoAMAT/KmmschadC8KetoacylCoAMAT+C16HydroxyacylCoAMAT/KmmschadC16HydroxyacylCoAMAT+C16KetoacylCoAMAT/KmmschadC16KetoacylCoAMAT+C14HydroxyacylCoAMAT/KmmschadC14HydroxyacylCoAMAT+C14KetoacylCoAMAT/KmmschadC14KetoacylCoAMAT+C12HydroxyacylCoAMAT/KmmschadC12HydroxyacylCoAMAT+C12KetoacylCoAMAT/KmmschadC12KetoacylCoAMAT+C10HydroxyacylCoAMAT/KmmschadC10HydroxyacylCoAMAT+C10KetoacylCoAMAT/KmmschadC10KetoacylCoAMAT+C6HydroxyacylCoAMAT/KmmschadC6HydroxyacylCoAMAT+C6KetoacylCoAMAT/KmmschadC6KetoacylCoAMAT+C4HydroxyacylCoAMAT/KmmschadC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KmmschadC4AcetoacylCoAMAT)*(1+(NADtMAT-NADHMAT)/KmmschadNADMAT+NADHMAT/KmmschadNADHMAT))
KmcrotC16EnoylCoAMAT = 150.0 uM; KmcrotC12EnoylCoAMAT = 25.0 uM; KicrotC4AcetoacylCoA = 1.6 uM; KmcrotC4EnoylCoAMAT = 40.0 uM; Vcrot = 3.6 uM per min per mgProtein; Keqcrot = 3.13 dimensionless; KmcrotC14EnoylCoAMAT = 100.0 uM; KmcrotC8EnoylCoAMAT = 25.0 uM; KmcrotC10HydroxyacylCoAMAT = 45.0 uM; KmcrotC8HydroxyacylCoAMAT = 45.0 uM; KmcrotC6EnoylCoAMAT = 25.0 uM; KmcrotC10EnoylCoAMAT = 25.0 uM; sfcrotC10=0.33 dimensionless; KmcrotC14HydroxyacylCoAMAT = 45.0 uM; KmcrotC4HydroxyacylCoAMAT = 45.0 uM; KmcrotC16HydroxyacylCoAMAT = 45.0 uM; KmcrotC12HydroxyacylCoAMAT = 45.0 uM; KmcrotC6HydroxyacylCoAMAT = 45.0 uMReaction: C10EnoylCoAMAT => C10HydroxyacylCoAMAT; C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, C10EnoylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C8EnoylCoAMAT, C6EnoylCoAMAT, C4EnoylCoAMAT, C10HydroxyacylCoAMAT, C16HydroxyacylCoAMAT, C14HydroxyacylCoAMAT, C12HydroxyacylCoAMAT, C8HydroxyacylCoAMAT, C6HydroxyacylCoAMAT, C4HydroxyacylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfcrotC10*Vcrot*(C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT-C10HydroxyacylCoAMAT/(KmcrotC10EnoylCoAMAT*Keqcrot))/(1+C10EnoylCoAMAT/KmcrotC10EnoylCoAMAT+C10HydroxyacylCoAMAT/KmcrotC10HydroxyacylCoAMAT+C16EnoylCoAMAT/KmcrotC16EnoylCoAMAT+C16HydroxyacylCoAMAT/KmcrotC16HydroxyacylCoAMAT+C14EnoylCoAMAT/KmcrotC14EnoylCoAMAT+C14HydroxyacylCoAMAT/KmcrotC14HydroxyacylCoAMAT+C12EnoylCoAMAT/KmcrotC12EnoylCoAMAT+C12HydroxyacylCoAMAT/KmcrotC12HydroxyacylCoAMAT+C8EnoylCoAMAT/KmcrotC8EnoylCoAMAT+C8HydroxyacylCoAMAT/KmcrotC8HydroxyacylCoAMAT+C6EnoylCoAMAT/KmcrotC6EnoylCoAMAT+C6HydroxyacylCoAMAT/KmcrotC6HydroxyacylCoAMAT+C4EnoylCoAMAT/KmcrotC4EnoylCoAMAT+C4HydroxyacylCoAMAT/KmcrotC4HydroxyacylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)
Kmcpt2C14AcylCarMAT = 51.0 uM; Kmcpt2C10AcylCarMAT = 51.0 uM; Kmcpt2CoAMAT = 30.0 uM; Kmcpt2C6AcylCarMAT = 51.0 uM; Kmcpt2C14AcylCoAMAT = 38.0 uM; Keqcpt2 = 2.22 dimensionless; sfcpt2C16=0.85 dimensionless; Kmcpt2C8AcylCoAMAT = 38.0 uM; Kmcpt2C6AcylCoAMAT = 1000.0 uM; Kmcpt2C16AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCarMAT = 51.0 uM; Kmcpt2C12AcylCoAMAT = 38.0 uM; Vcpt2 = 0.391 uM per min per mgProtein; Kmcpt2C8AcylCarMAT = 51.0 uM; Kmcpt2CarMAT = 350.0 uM; Kmcpt2C16AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCoAMAT = 1000000.0 uM; Kmcpt2C10AcylCoAMAT = 38.0 uM; Kmcpt2C4AcylCarMAT = 51.0 uMReaction: C16AcylCarMAT => C16AcylCoAMAT; C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, C16AcylCarMAT, C14AcylCarMAT, C12AcylCarMAT, C10AcylCarMAT, C8AcylCarMAT, C6AcylCarMAT, C4AcylCarMAT, CoAMAT, C16AcylCoAMAT, C14AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcylCoAMAT, CarMAT, Rate Law: sfcpt2C16*Vcpt2*(C16AcylCarMAT*CoAMAT/(Kmcpt2C16AcylCarMAT*Kmcpt2CoAMAT)-C16AcylCoAMAT*CarMAT/(Kmcpt2C16AcylCarMAT*Kmcpt2CoAMAT*Keqcpt2))/((1+C16AcylCarMAT/Kmcpt2C16AcylCarMAT+C16AcylCoAMAT/Kmcpt2C16AcylCoAMAT+C14AcylCarMAT/Kmcpt2C14AcylCarMAT+C14AcylCoAMAT/Kmcpt2C14AcylCoAMAT+C12AcylCarMAT/Kmcpt2C12AcylCarMAT+C12AcylCoAMAT/Kmcpt2C12AcylCoAMAT+C10AcylCarMAT/Kmcpt2C10AcylCarMAT+C10AcylCoAMAT/Kmcpt2C10AcylCoAMAT+C8AcylCarMAT/Kmcpt2C8AcylCarMAT+C8AcylCoAMAT/Kmcpt2C8AcylCoAMAT+C6AcylCarMAT/Kmcpt2C6AcylCarMAT+C6AcylCoAMAT/Kmcpt2C6AcylCoAMAT+C4AcylCarMAT/Kmcpt2C4AcylCarMAT+C4AcylCoAMAT/Kmcpt2C4AcylCoAMAT)*(1+CoAMAT/Kmcpt2CoAMAT+CarMAT/Kmcpt2CarMAT))
KmmtpC14EnoylCoAMAT = 25.0 uM; KmmtpC8EnoylCoAMAT = 25.0 uM; KmmtpC16AcylCoAMAT = 13.83 uM; KmmtpCoAMAT = 30.0 uM; KmmtpC16EnoylCoAMAT = 25.0 uM; KmmtpC12EnoylCoAMAT = 25.0 uM; KmmtpAcetylCoAMAT = 30.0 uM; Vmtp = 2.84 uM per min per mgProtein; KmmtpC12AcylCoAMAT = 13.83 uM; KmmtpC8AcylCoAMAT = 13.83 uM; KmmtpC14AcylCoAMAT = 13.83 uM; KmmtpC6AcylCoAMAT = 13.83 uM; KmmtpC10EnoylCoAMAT = 25.0 uM; Keqmtp = 0.71 dimensionless; KicrotC4AcetoacylCoA = 1.6 uM; KmmtpNADMAT = 60.0 uM; KmmtpNADHMAT = 50.0 uM; KmmtpC10AcylCoAMAT = 13.83 uM; sfmtpC16=1.0 dimensionlessReaction: C16EnoylCoAMAT => C14AcylCoAMAT + AcetylCoAMAT + NADHMAT; C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, C4AcetoacylCoAMAT, C16EnoylCoAMAT, C14EnoylCoAMAT, C12EnoylCoAMAT, C10EnoylCoAMAT, C8EnoylCoAMAT, NADtMAT, CoAMAT, C14AcylCoAMAT, C16AcylCoAMAT, C12AcylCoAMAT, C10AcylCoAMAT, C8AcylCoAMAT, C6AcylCoAMAT, NADHMAT, AcetylCoAMAT, C4AcetoacylCoAMAT, Rate Law: sfmtpC16*Vmtp*(C16EnoylCoAMAT*(NADtMAT-NADHMAT)*CoAMAT/(KmmtpC16EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT)-C14AcylCoAMAT*NADHMAT*AcetylCoAMAT/(KmmtpC16EnoylCoAMAT*KmmtpNADMAT*KmmtpCoAMAT*Keqmtp))/((1+C16EnoylCoAMAT/KmmtpC16EnoylCoAMAT+C14AcylCoAMAT/KmmtpC14AcylCoAMAT+C14EnoylCoAMAT/KmmtpC14EnoylCoAMAT+C16AcylCoAMAT/KmmtpC16AcylCoAMAT+C12EnoylCoAMAT/KmmtpC12EnoylCoAMAT+C12AcylCoAMAT/KmmtpC12AcylCoAMAT+C10EnoylCoAMAT/KmmtpC10EnoylCoAMAT+C10AcylCoAMAT/KmmtpC10AcylCoAMAT+C8EnoylCoAMAT/KmmtpC8EnoylCoAMAT+C8AcylCoAMAT/KmmtpC8AcylCoAMAT+C6AcylCoAMAT/KmmtpC6AcylCoAMAT+C4AcetoacylCoAMAT/KicrotC4AcetoacylCoA)*(1+(NADtMAT-NADHMAT)/KmmtpNADMAT+NADHMAT/KmmtpNADHMAT)*(1+CoAMAT/KmmtpCoAMAT+AcetylCoAMAT/KmmtpAcetylCoAMAT))

States:

NameDescription
C12AcylCarCYT[O-acylcarnitine]
C8HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C14KetoacylCoAMAT[fatty acyl-CoA]
C14EnoylCoAMAT[cis-2-enoyl-CoA]
C10EnoylCoAMAT[cis-2-enoyl-CoA]
C16AcylCarCYT[O-acylcarnitine]
C12AcylCoAMAT[fatty acyl-CoA]
C8KetoacylCoAMAT[fatty acyl-CoA]
C4HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C14HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C6AcylCoAMAT[fatty acyl-CoA]
C6AcylCarMAT[O-acylcarnitine]
C8AcylCoAMAT[fatty acyl-CoA]
C16AcylCarMAT[O-acylcarnitine]
C16AcylCoACYT[palmitoyl-CoA; fatty acyl-CoA]
NADHMAT[NADH]
C8AcylCarCYT[O-acylcarnitine]
C10KetoacylCoAMAT[fatty acyl-CoA]
C6AcylCarCYT[O-acylcarnitine]
C16EnoylCoAMAT[cis-2-enoyl-CoA]
C6HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C10AcylCoAMAT[fatty acyl-CoA]
C16HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
FADHMAT[FADH(.)]
C14AcylCoAMAT[myristoyl-CoA; fatty acyl-CoA]
AcetylCoAMAT[acetyl-CoA]
C6KetoacylCoAMAT[fatty acyl-CoA]
C16AcylCoAMAT[fatty acyl-CoA; palmitoyl-CoA]
CoAMAT[coenzyme A]
C6EnoylCoAMAT[cis-2-enoyl-CoA]
C12EnoylCoAMAT[cis-2-enoyl-CoA]
C10HydroxyacylCoAMAT[hydroxy fatty acyl-CoA]
C12AcylCarMAT[O-acylcarnitine]
C8AcylCarMAT[O-acylcarnitine]
C4AcetoacylCoAMAT[fatty acyl-CoA]
C8EnoylCoAMAT[cis-2-enoyl-CoA]

VanTol2020 - Thalassiosira pseudonana CCMP 1335 GEM, acclimated to high light intensity: MODEL2010230002v0.0.1

Thalassiosira pseudonana CCMP 1335, acclimated to 200 umol photons m-2 s-1 (iTps1432_HL). doi: https://doi.org/10.1101/…

Details

Diatoms are unicellular photosynthetic algae known to secrete organic matter that fuels secondary production in the ocean, though our knowledge of how their physiology impacts the character of dissolved organic matter remains limited. Like all photosynthetic organisms, their use of light for energy and reducing power creates the challenge of avoiding cellular damage. To better understand the interplay between redox balance and organic matter secretion, we reconstructed a genome-scale metabolic model of Thalassiosira pseudonana strain CCMP 1335, a model for diatom molecular biology and physiology, with a 60-year history of studies. The model simulates the metabolic activities of 1,432 genes via a network of 2,792 metabolites produced through 6,079 reactions distributed across six subcellular compartments. Growth was simulated under different steady-state light conditions (5-200 µmol photons m-2 s-1) and in a batch culture progressing from exponential growth to nitrate-limitation and nitrogen-starvation. We used the model to examine the dissipation of reductants generated through light-dependent processes and found that when available, nitrate assimilation is an important means of dissipating reductants in the plastid; under nitrate-limiting conditions, sulfate assimilation plays a similar role. The use of either nitrate or sulfate uptake to balance redox reactions leads to the secretion of distinct organic nitrogen and sulfur compounds. Such compounds can be accessed by bacteria in the surface ocean. The model of the diatom Thalassiosira pseudonana provides a mechanistic explanation for the production of ecologically and climatologically relevant compounds that may serve as the basis for intricate, cross-kingdom microbial networks. Diatom metabolism has an important influence on global biogeochemistry; metabolic models of marine microorganisms link genes to ecosystems and may be key to integrating molecular data with models of ocean biogeochemistry. link: http://identifiers.org/doi/10.1101/2020.10.26.355578

VanTol2020 - Thalassiosira pseudonana CCMP 1335 GEM, acclimated to low light intensity: MODEL2010230003v0.0.1

Thalassiosira pseudonana CCMP 1335, acclimated to 5 umol photons m-2 s-1 (iTps1432_LL). doi: https://doi.org/10.1101/20…

Details

Diatoms are unicellular photosynthetic algae known to secrete organic matter that fuels secondary production in the ocean, though our knowledge of how their physiology impacts the character of dissolved organic matter remains limited. Like all photosynthetic organisms, their use of light for energy and reducing power creates the challenge of avoiding cellular damage. To better understand the interplay between redox balance and organic matter secretion, we reconstructed a genome-scale metabolic model of Thalassiosira pseudonana strain CCMP 1335, a model for diatom molecular biology and physiology, with a 60-year history of studies. The model simulates the metabolic activities of 1,432 genes via a network of 2,792 metabolites produced through 6,079 reactions distributed across six subcellular compartments. Growth was simulated under different steady-state light conditions (5-200 µmol photons m-2 s-1) and in a batch culture progressing from exponential growth to nitrate-limitation and nitrogen-starvation. We used the model to examine the dissipation of reductants generated through light-dependent processes and found that when available, nitrate assimilation is an important means of dissipating reductants in the plastid; under nitrate-limiting conditions, sulfate assimilation plays a similar role. The use of either nitrate or sulfate uptake to balance redox reactions leads to the secretion of distinct organic nitrogen and sulfur compounds. Such compounds can be accessed by bacteria in the surface ocean. The model of the diatom Thalassiosira pseudonana provides a mechanistic explanation for the production of ecologically and climatologically relevant compounds that may serve as the basis for intricate, cross-kingdom microbial networks. Diatom metabolism has an important influence on global biogeochemistry; metabolic models of marine microorganisms link genes to ecosystems and may be key to integrating molecular data with models of ocean biogeochemistry. link: http://identifiers.org/doi/10.1101/2020.10.26.355578

VanTol2020 - Thalassiosira pseudonana CCMP 1335 GEM, acclimated to medium light intensity: MODEL2010230001v0.0.1

Thalassiosira pseudonana CCMP 1335, acclimated to 60 umol photons m-2 s-1 (iTps1432_ML). doi: https://doi.org/10.1101/2…

Details

Diatoms are unicellular photosynthetic algae known to secrete organic matter that fuels secondary production in the ocean, though our knowledge of how their physiology impacts the character of dissolved organic matter remains limited. Like all photosynthetic organisms, their use of light for energy and reducing power creates the challenge of avoiding cellular damage. To better understand the interplay between redox balance and organic matter secretion, we reconstructed a genome-scale metabolic model of Thalassiosira pseudonana strain CCMP 1335, a model for diatom molecular biology and physiology, with a 60-year history of studies. The model simulates the metabolic activities of 1,432 genes via a network of 2,792 metabolites produced through 6,079 reactions distributed across six subcellular compartments. Growth was simulated under different steady-state light conditions (5-200 µmol photons m-2 s-1) and in a batch culture progressing from exponential growth to nitrate-limitation and nitrogen-starvation. We used the model to examine the dissipation of reductants generated through light-dependent processes and found that when available, nitrate assimilation is an important means of dissipating reductants in the plastid; under nitrate-limiting conditions, sulfate assimilation plays a similar role. The use of either nitrate or sulfate uptake to balance redox reactions leads to the secretion of distinct organic nitrogen and sulfur compounds. Such compounds can be accessed by bacteria in the surface ocean. The model of the diatom Thalassiosira pseudonana provides a mechanistic explanation for the production of ecologically and climatologically relevant compounds that may serve as the basis for intricate, cross-kingdom microbial networks. Diatom metabolism has an important influence on global biogeochemistry; metabolic models of marine microorganisms link genes to ecosystems and may be key to integrating molecular data with models of ocean biogeochemistry. link: http://identifiers.org/doi/10.1101/2020.10.26.355578

Varusai2018 - Dynamic modelling of the mTOR signalling network reveals complex emergent behaviours conferred by DEPTOR: BIOMD0000000823v0.0.1

This is a mathematical describing the effect that DEP domain-containing mTOR-interacting protein (DEPTOR) has on the mam…

Details

The mechanistic Target of Rapamycin (mTOR) signalling network is an evolutionarily conserved network that controls key cellular processes, including cell growth and metabolism. Consisting of the major kinase complexes mTOR Complex 1 and 2 (mTORC1/2), the mTOR network harbours complex interactions and feedback loops. The DEP domain-containing mTOR-interacting protein (DEPTOR) was recently identified as an endogenous inhibitor of both mTORC1 and 2 through direct interactions, and is in turn degraded by mTORC1/2, adding an extra layer of complexity to the mTOR network. Yet, the dynamic properties of the DEPTOR-mTOR network and the roles of DEPTOR in coordinating mTORC1/2 activation dynamics have not been characterised. Using computational modelling, systems analysis and dynamic simulations we show that DEPTOR confers remarkably rich and complex dynamic behaviours to mTOR signalling, including abrupt, bistable switches, oscillations and co-existing bistable/oscillatory responses. Transitions between these distinct modes of behaviour are enabled by modulating DEPTOR expression alone. We characterise the governing conditions for the observed dynamics by elucidating the network in its vast multi-dimensional parameter space, and develop strategies to identify core network design motifs underlying these dynamics. Our findings provide new systems-level insights into the complexity of mTOR signalling contributed by DEPTOR. link: http://identifiers.org/pubmed/29330362

Parameters:

NameDescription
Km16 = 50.0; V16 = 1.0Reaction: iIRS => IRS, Rate Law: compartment*V16*iIRS/(Km16+iIRS)
kd18 = 0.0Reaction: pDEPTOR =>, Rate Law: compartment*kd18*pDEPTOR
Km8 = 1.0; V8 = 6.0Reaction: pmTORC1 => mTORC1, Rate Law: compartment*V8*pmTORC1/(Km8+pmTORC1)
ks17 = 0.0Reaction: => DEPTOR, Rate Law: compartment*ks17
Km6 = 34.0; V6 = 2.0Reaction: pAkt => Akt, Rate Law: compartment*V6*pAkt/(Km6+pAkt)
V4 = 1.0; Km4 = 50.0Reaction: pIRS => IRS, Rate Law: compartment*V4*pIRS/(Km4+pIRS)
Km2 = 35.0; V2 = 1.0Reaction: pIR => IR, Rate Law: compartment*V2*pIR/(Km2+pIR)
k9c = 0.3; Km9 = 160.0Reaction: mTORC2 => pmTORC2; pIR, Rate Law: compartment*k9c*mTORC2*pIR/(Km9+mTORC2)
Km12 = 7.0; V12 = 4.0Reaction: pDEPTOR => DEPTOR, Rate Law: compartment*V12*pDEPTOR/(Km12+pDEPTOR)
Km3 = 50.0; k3c = 0.1Reaction: IRS => pIRS; pIR, Rate Law: compartment*k3c*IRS*pIR/(Km3+IRS)
V10 = 3.0; Km10 = 7.0Reaction: pmTORC2 => mTORC2, Rate Law: compartment*V10*pmTORC2/(Km10+pmTORC2)
k7c = 0.1; Km7 = 2.0Reaction: mTORC1 => pmTORC1; pAkt, Rate Law: compartment*k7c*mTORC1*pAkt/(Km7+mTORC1)
k13r = 0.006; k13f = 0.001Reaction: mTORC1 + DEPTOR => mTORC1_DEPTOR, Rate Law: compartment*(k13f*mTORC1*DEPTOR-k13r*mTORC1_DEPTOR)
k15c = 0.1; Km15 = 50.0Reaction: IRS => iIRS; pmTORC1, Rate Law: compartment*k15c*IRS*pmTORC1/(Km15+IRS)
k5ca = 0.05; k5cb = 1.5; Km5a = 7.0; Km5b = 4.0Reaction: Akt => pAkt; pIRS, pmTORC2, Rate Law: compartment*(k5ca*pIRS*Akt/(Km5a+Akt)+k5cb*pmTORC2*Akt/(Km5b+Akt))
k14f = 0.007; k14r = 0.006Reaction: mTORC2 + DEPTOR => mTORC2_DEPTOR, Rate Law: compartment*(k14f*mTORC2*DEPTOR-k14r*mTORC2_DEPTOR)
V1 = 1.0; Km1 = 95.0Reaction: IR => pIR, Rate Law: compartment*V1*IR/(Km1+IR)
Km11b = 11.0; k11ca = 0.1; Km11a = 120.0; k11cb = 0.13Reaction: DEPTOR => pDEPTOR; pmTORC1, pmTORC2, Rate Law: compartment*(k11ca*pmTORC1*DEPTOR/(Km11a+pDEPTOR)+k11cb*pmTORC2*DEPTOR/(Km11b+DEPTOR))

States:

NameDescription
mTORC2 DEPTOR[DEP Domain-Containing mTOR-Interacting Protein; mTORC2]
mTORC2[mTORC2]
IRS[Insulin Receptor Substrate 1]
Akt[AKT kinase]
pIR[insulin receptor]
pmTORC1[mTORC1]
iIRS[Insulin Receptor Substrate 1]
mTORC1[mTORC1]
pmTORC2[mTORC2]
pIRS[Insulin Receptor Substrate 1]
pDEPTOR[DEP Domain-Containing mTOR-Interacting Protein]
pAkt[AKT kinase]
IR[insulin receptor]
DEPTOR[DEP Domain-Containing mTOR-Interacting Protein]
mTORC1 DEPTOR[mTORC1; DEP Domain-Containing mTOR-Interacting Protein]

Vasalou2010_Pacemaker_Neuron_SCN: BIOMD0000000246v0.0.1

This the single cell model from the article: A multiscale model to investigate circadian rhythmicity of pacemaker neur…

Details

The suprachiasmatic nucleus (SCN) of the hypothalamus is a multicellular system that drives daily rhythms in mammalian behavior and physiology. Although the gene regulatory network that produces daily oscillations within individual neurons is well characterized, less is known about the electrophysiology of the SCN cells and how firing rate correlates with circadian gene expression. We developed a firing rate code model to incorporate known electrophysiological properties of SCN pacemaker cells, including circadian dependent changes in membrane voltage and ion conductances. Calcium dynamics were included in the model as the putative link between electrical firing and gene expression. Individual ion currents exhibited oscillatory patterns matching experimental data both in current levels and phase relationships. VIP and GABA neurotransmitters, which encode synaptic signals across the SCN, were found to play critical roles in daily oscillations of membrane excitability and gene expression. Blocking various mechanisms of intracellular calcium accumulation by simulated pharmacological agents (nimodipine, IP3- and ryanodine-blockers) reproduced experimentally observed trends in firing rate dynamics and core-clock gene transcription. The intracellular calcium concentration was shown to regulate diverse circadian processes such as firing frequency, gene expression and system periodicity. The model predicted a direct relationship between firing frequency and gene expression amplitudes, demonstrated the importance of intracellular pathways for single cell behavior and provided a novel multiscale framework which captured characteristics of the SCN at both the electrophysiological and gene regulatory levels. link: http://identifiers.org/pubmed/20300645

Parameters:

NameDescription
k1 = 0.45 per_h; k2 = 0.2 per_hReaction: PC_C => PC_N, Rate Law: cytoplasm*k1*PC_C-nucleus*k2*PC_N
vP = 1.0 nM_per_h; v_K = NaN nM_per_h; K_2_CB = 0.01 nM; K_1_CB = 0.01 nM; WT = 1.0 dimensionlessReaction: => CB, Rate Law: cytoplasm*(v_K*(1-CB)/((K_1_CB+1)-CB)-vP*CB/(K_2_CB+CB))/WT
V1_C = 0.6 nM_per_h; K_dp = 0.1 nM; V2_C = 0.1 nM_per_h; K_p = 0.1 nMReaction: C_C => C_CP, Rate Law: cytoplasm*(V1_C*C_C/(K_p+C_C)-V2_C*C_CP/(K_dp+C_CP))
k3 = 0.4 per_nM_per_h; k4 = 0.2 per_hReaction: P_C + C_C => PC_C, Rate Law: cytoplasm*(k3*P_C*C_C-k4*PC_C)
v_mC = 1.0 nM_per_h; kd_mC = 0.01 per_h; K_mC = 0.4 nMReaction: M_C =>, Rate Law: cytoplasm*(v_mC*M_C/(K_mC+M_C)+kd_mC*M_C)
Kd = 0.3 nM; v_dPC = 0.7 per_nM_per_h; kd_n = 0.01 per_hReaction: P_CP =>, Rate Law: cytoplasm*(v_dPC*P_CP/(Kd+P_CP)+kd_n*P_CP)
Kd = 0.3 nM; v_dCC = 0.7 nM_per_h; kd_n = 0.01 per_hReaction: C_CP =>, Rate Law: cytoplasm*(v_dCC*C_CP/(Kd+C_CP)+kd_n*C_CP)
m_BN = 2.0 dimensionless; K_IB = 2.2 nM; v_sB = 1.0 nM_per_hReaction: => M_B; B_N, Rate Law: cytoplasm*v_sB*K_IB^m_BN/(K_IB^m_BN+B_N^m_BN)
n_BN = 4.0 dimensionless; v_sC = 1.1 nM_per_h; K_sC = 0.6 nMReaction: => M_C; B_N, Rate Law: cytoplasm*v_sC*B_N^n_BN/(K_sC^n_BN+B_N^n_BN)
n_BN = 4.0 dimensionless; K_C = 0.15 nM; v_sP0 = 1.0 nM_per_h; K_AP = 0.6 nM; C_T = 1.6 nM_per_hReaction: => M_P; CB, B_N, Rate Law: cytoplasm*(v_sP0+C_T*CB/(K_C+CB))*B_N^n_BN/(K_AP^n_BN+B_N^n_BN)
K_VIP = 15.0; v_VIP = 0.5 nM_per_h; n_VIP = 1.9 dimensionless; f_r = NaN HzReaction: => VIP, Rate Law: cytoplasm*v_VIP*f_r^n_VIP/(K_VIP+f_r^n_VIP)
K_dp = 0.1 nM; V2_P = 0.3 nM_per_h; K_p = 0.1 nM; V1_P = NaN nM_per_hReaction: P_C => P_CP, Rate Law: cytoplasm*(V1_P*P_C/(K_p+P_C)-V2_P*P_CP/(K_dp+P_CP))
n_M3 = 6.0 dimensionless; p_A = 4.2 dimensionless; K_R_Ca = 3.0 uM; K_A = 0.67 uM; V_M3 = 400.0 uM_per_hReaction: Ca_store => Ca_in, Rate Law: 1000*store*V_M3*Ca_store^n_M3/(K_R_Ca^n_M3+Ca_store^n_M3)*Ca_in^p_A/(K_A^p_A+Ca_in^p_A)
theta_Na = NaN milliVoltReaction: Na_in = Na_ex/theta_Na, Rate Law: missing
kd_nc = 0.12 per_hReaction: C_C =>, Rate Law: cytoplasm*kd_nc*C_C
k5 = 0.4; k6 = 0.2Reaction: B_C => B_N, Rate Law: cytoplasm*k5*B_C-nucleus*k6*B_N
Kd = 0.3 nM; vd_IN = 0.8 nM_per_h; kd_n = 0.01 per_hReaction: I_N =>, Rate Law: nucleus*(vd_IN*I_N/(Kd+I_N)+kd_n*I_N)
K_dp = 0.1 nM; V4_B = 0.2 nM_per_h; V3_B = 0.5 nM_per_h; K_p = 0.1 nMReaction: B_N => B_NP, Rate Law: nucleus*(V3_B*B_N/(K_p+B_N)-V4_B*B_NP/(K_dp+B_NP))
v_kk = 3.3 per_uM_per_h; K_kk = 0.02 nM; n_kCa = 2.0 dimensionless; n_kk = 0.1 dimensionlessReaction: Ca_in => ; C_C, Rate Law: 1000*cytoplasm*v_kk*C_C^n_kk/(K_kk+C_C^n_kk)*Ca_in^n_kCa
K_vo = 4.5 nM; n_vo = 4.5 dimensionless; v_vo = 0.09 uM_per_hReaction: => Ca_in; B_C, Rate Law: 1000*cytoplasm*v_vo*B_C^n_vo/(K_vo+B_C^n_vo)
k7 = 0.5 per_nM_per_h; k8 = 0.1 per_hReaction: B_N + PC_N => I_N, Rate Law: cytoplasm*(k7*B_N*PC_N-k8*I_N)
Kd = 0.3 nM; vd_BC = 0.5 nM_per_h; kd_n = 0.01 per_hReaction: B_CP =>, Rate Law: cytoplasm*(vd_BC*B_CP/(Kd+B_CP)+kd_n*B_CP)
theta_K = NaN milliVoltReaction: K_in = K_ex/theta_K, Rate Law: missing
V_M2 = 149.5 uM_per_h; n_M2 = 2.2 dimensionless; K_2 = 5.0 uMReaction: Ca_in => Ca_store, Rate Law: 1000*cytoplasm*V_M2*Ca_in^n_M2/(K_2^n_M2+Ca_in^n_M2)
v_mP = 1.1 nM_per_h; K_mP = 0.31 nM; kd_mP = 0.01 per_hReaction: M_P =>, Rate Law: cytoplasm*(v_mP*M_P/(K_mP+M_P)+kd_mP*M_P)
Kd = 0.3 nM; vd_PCC = 0.7 nM_per_h; kd_n = 0.01 per_hReaction: PC_CP =>, Rate Law: cytoplasm*(vd_PCC*PC_CP/(Kd+PC_CP)+kd_n*PC_CP)
V1_B = 0.5 nM_per_h; K_dp = 0.1 nM; V2_B = 0.1 nM_per_h; K_p = 0.1 nMReaction: B_C => B_CP, Rate Law: cytoplasm*(V1_B*B_C/(K_p+B_C)-V2_B*B_CP/(K_dp+B_CP))
V3_PC = NaN nM_per_h; V4_PC = 0.1 nM_per_h; K_dp = 0.1 nM; K_p = 0.1 nMReaction: PC_N => PC_NP, Rate Law: nucleus*(V3_PC*PC_N/(K_p+PC_N)-V4_PC*PC_NP/(K_dp+PC_NP))
kd_n = 0.01 per_hReaction: B_N =>, Rate Law: nucleus*kd_n*B_N
kd_mB = 0.01 per_h; K_mB = 0.4 nM; v_mB = 0.8 nM_per_hReaction: M_B =>, Rate Law: cytoplasm*(v_mB*M_B/(K_mB+M_B)+kd_mB*M_B)
n_dVIP = 0.2 dimensionless; k_dVIP = 0.5Reaction: VIP =>, Rate Law: cytoplasm*k_dVIP*VIP^n_dVIP
ks_P = 0.6 per_hReaction: => P_C; M_P, Rate Law: cytoplasm*ks_P*M_P
V1_PC = NaN nM_per_h; K_dp = 0.1 nM; K_p = 0.1 nM; V2_PC = 0.1 nM_per_hReaction: PC_C => PC_CP, Rate Law: cytoplasm*(V1_PC*PC_C/(K_p+PC_C)-V2_PC*PC_CP/(K_dp+PC_CP))
k_f = 0.001 per_hReaction: Ca_store => Ca_in, Rate Law: 1000*store*k_f*Ca_store
ks_C = 1.6 per_hReaction: => C_C; M_C, Rate Law: cytoplasm*ks_C*M_C
beta_IP3 = 0.5 dimensionless; V_M1 = 3.0E-4 uM_per_hReaction: => Ca_in, Rate Law: 1000*cytoplasm*V_M1*beta_IP3
v_GABA = 19.0 nM; K_GABA = 3.0 nMReaction: GABA = GABA_o+v_GABA*VIP/(K_GABA+VIP), Rate Law: missing
ksB = 0.12Reaction: => B_C; M_B, Rate Law: cytoplasm*ksB*M_B
Kd = 0.3 nM; vd_BN = 0.6 nM_per_h; kd_n = 0.01 per_hReaction: B_NP =>, Rate Law: nucleus*(vd_BN*B_NP/(Kd+B_NP)+kd_n*B_NP)
Kd = 0.3 nM; vd_PCN = 0.7 nM_per_h; kd_n = 0.01 per_hReaction: PC_NP =>, Rate Law: nucleus*(vd_PCN*PC_NP/(Kd+PC_NP)+kd_n*PC_NP)

States:

NameDescription
Na in[CHEBI_9175; Sodium cation]
M B[Aryl hydrocarbon receptor nuclear translocator-like protein 1; messenger RNA]
B N[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
C C[Cryptochrome-2; Cryptochrome-1]
GABA[4-Aminobutanoate; gamma-aminobutyric acid]
PC NP[Cryptochrome-1; Period circadian protein homolog 3; Cryptochrome-1; Period circadian protein homolog 2; Cryptochrome-1; Period circadian protein homolog 1]
B C[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
C CP[Cryptochrome-2; Cryptochrome-1; Phosphoprotein]
Ca store[calcium(2+); Calcium cation]
K in[potassium(1+); Potassium cation]
P CP[Period circadian protein homolog 3; Period circadian protein homolog 2; Period circadian protein homolog 1]
B CP[Phosphoprotein; Aryl hydrocarbon receptor nuclear translocator-like protein 1]
CB[Cyclic AMP-responsive element-binding protein 1]
M C[Cryptochrome-2; Cryptochrome-1; messenger RNA]
VIP[Vasoactive intestinal polypeptide receptor 1]
M P[Period circadian protein homolog 3; Period circadian protein homolog 2; Period circadian protein homolog 1; messenger RNA]
PC C[Cryptochrome-1; Period circadian protein homolog 3; Cryptochrome-1; Period circadian protein homolog 1]
PC CP[Phosphoprotein; Cryptochrome-1; Period circadian protein homolog 1; Cryptochrome-1; Period circadian protein homolog 3]
Ca in[calcium(2+); Calcium cation]
I NI_N
P C[Period circadian protein homolog 3; Period circadian protein homolog 2; Period circadian protein homolog 1]
B NP[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
PC N[Cryptochrome-2; Period circadian protein homolog 3; Cryptochrome-1; Period circadian protein homolog 1]

Vaseghi1999_Pentose_PP_yeast: MODEL1004070001v0.0.1

Model as described in: In vivo dynamics of the pentose phosphate pathway in Saccharomyces cerevisiae Vaseghi S, Baum…

Details

The in vivo dynamics of the pentose phosphate pathway has been studied with transient experiments in continuous culture of Saccharomyces cerevisiae. Rapid sampling was performed with a special sampling device after disturbing the steady state with a pulse of glucose. The time span of observation was 120 s after the pulse. During this short time period the dynamic effect of protein biosynthesis can be neglected. The metabolites of interest (glucose 6-phosphate, NADP, NADPH, 6-phosphogluconate, and MgATP2-) we determined with enzymatic assays and HPLC. The experimental observations were then used for the identification of kinetic rate equations and parameters under in vivo conditions. In accordance with results from in vitro studies the in vivo diagnosis supports an ordered Bi-Bi mechanism with noncompetitive inhibition by MgATP2- for the enzyme glucose-6-phosphate dehydrogenase. In the case of 6-phosphogluconate dehydrogenase an ordered Bi-Ter mechanism with a competitive inhibition by MgATP2- has been found. Because the MgATP2- concentration decreases abruptly after the pulse of glucose the inhibitory effect vanishes and the flux through the pentose phosphate pathway increases. This regulation phenomenon guarantees the balance of fluxes through glycolysis and pentose phosphate pathway during the dynamic time period. link: http://identifiers.org/pubmed/10935926

Vazquez2014 - Chemical inhibition from amyloid protein aggregation kinetics: BIOMD0000000532v0.0.1

Vazquez2014 - Chemical inhibition from amyloid protein aggregation kineticsThis model is described in the article: [Mod…

Details

BACKGROUNDS: The process of amyloid proteins aggregation causes several human neuropathologies. In some cases, e.g. fibrillar deposits of insulin, the problems are generated in the processes of production and purification of protein and in the pump devices or injectable preparations for diabetics. Experimental kinetics and adequate modelling of chemical inhibition from amyloid aggregation are of practical importance in order to study the viable processing, formulation and storage as well as to predict and optimize the best conditions to reduce the effect of protein nucleation. RESULTS: In this manuscript, experimental data of insulin, Aβ42 amyloid protein and apomyoglobin fibrillation from recent bibliography were selected to evaluate the capability of a bivariate sigmoid equation to model them. The mathematical functions (logistic combined with Weibull equation) were used in reparameterized form and the effect of inhibitor concentrations on kinetic parameters from logistic equation were perfectly defined and explained. The surfaces of data were accurately described by proposed model and the presented analysis characterized the inhibitory influence on the protein aggregation by several chemicals. Discrimination between true and apparent inhibitors was also confirmed by the bivariate equation. EGCG for insulin (working at pH = 7.4/T = 37°C) and taiwaniaflavone for Aβ42 were the compounds studied that shown the greatest inhibition capacity. CONCLUSIONS: An accurate, simple and effective model to investigate the inhibition of chemicals on amyloid protein aggregation has been developed. The equation could be useful for the clear quantification of inhibitor potential of chemicals and rigorous comparison among them. link: http://identifiers.org/pubmed/24572069

Parameters:

NameDescription
lambda = 3.0; C = 1.0; mlambda = 2.0; alambda = 2.0; klambda = 1.0Reaction: Lambda = lambda*(1+klambda*(1-exp((-ln(2))*(C/mlambda)^alambda))), Rate Law: missing
kx = 1.0; C = 1.0; ax = 2.0; mx = 5.0; xm = 1.0Reaction: Xm = xm*(1-kx*(1-exp((-ln(2))*(C/mx)^ax))), Rate Law: missing
av = 2.0; C = 1.0; kv = 1.0; mv = 4.0; vm = 0.25Reaction: Vm = vm*(1-kv*(1-exp((-ln(2))*(C/mv)^av))), Rate Law: missing

States:

NameDescription
XmXm
X[amyloid plaque]
LambdaLambda
VmVm

Veening2008_DegU_Regulation: BIOMD0000000240v0.0.1

This a model from the article: Transient heterogeneity in extracellular protease production by Bacillus subtilis. V…

Details

The most sophisticated survival strategy Bacillus subtilis employs is the differentiation of a subpopulation of cells into highly resistant endospores. To examine the expression patterns of non-sporulating cells within heterogeneous populations, we used buoyant density centrifugation to separate vegetative cells from endospore-containing cells and compared the transcriptome profiles of both subpopulations. This demonstrated the differential expression of various regulons. Subsequent single-cell analyses using promoter-gfp fusions confirmed our microarray results. Surprisingly, only part of the vegetative subpopulation highly and transiently expresses genes encoding the extracellular proteases Bpr (bacillopeptidase) and AprE (subtilisin), both of which are under the control of the DegU transcriptional regulator. As these proteases and their degradation products freely diffuse within the liquid growth medium, all cells within the clonal population are expected to benefit from their activities, suggesting that B. subtilis employs cooperative or even altruistic behavior. To unravel the mechanisms by which protease production heterogeneity within the non-sporulating subpopulation is established, we performed a series of genetic experiments combined with mathematical modeling. Simulations with our model yield valuable insights into how population heterogeneity may arise by the relatively long and variable response times within the DegU autoactivating pathway. link: http://identifiers.org/pubmed/18414485

Parameters:

NameDescription
ksyn1 = 0.04Reaction: => DegU; mDegU, Rate Law: ksyn1*mDegU*univ
ka = 0.025Reaction: DegUP => Dim, Rate Law: ka*DegUP^2
kdeg = 4.0E-4Reaction: DegU =>, Rate Law: kdeg*DegU*univ
kdegm = 0.01Reaction: mAprE =>, Rate Law: kdegm*mAprE
kdephos = NaNReaction: DegUP => DegU, Rate Law: kdephos*DegUP
kd = 0.1Reaction: Dim => DegUP, Rate Law: kd*Dim
kphos = NaNReaction: DegU => DegUP, Rate Law: kphos*DegU
ksyn = 0.04Reaction: => AprE; mAprE, Rate Law: ksyn*mAprE*univ
Kr1 = 7.0; R = 7.0; Kdim = 12.0; Iro = 0.02; Kr = 7.0; Irmax = 0.4Reaction: => mAprE; Dim, Rate Law: Kr1/(R+Kr1)*(Iro*(Dim*univ/Kdim+1)/(1+Dim*univ/Kdim+(Dim*univ)^2/Kdim^2+R/Kr)+Irmax*(Dim*univ)^2/(Kdim^2*(1+Dim*univ/Kdim+(Dim*univ)^2/Kdim^2+R/Kr)))
K = 7.0; Imax = 0.048; Io = 0.004Reaction: => mDegU; Dim, Rate Law: Io*K/(Dim*univ+K)+Imax*Dim*univ/(Dim*univ+K)

States:

NameDescription
Dim[protein homodimerization activity]
DegU[Transcriptional regulatory protein DegU]
mAprE[Subtilisin E]
AprE[Subtilisin E]
DegUP[Transcriptional regulatory protein DegU]
mDegU[Transcriptional regulatory protein DegU]

Veith2015 - Genome-scale metabolic model of Enterococcus faecalis V583: MODEL1510010000v0.0.1

efa201208

Details

Increasing antibiotic resistance in pathogenic bacteria necessitates the development of new medication strategies. Interfering with the metabolic network of the pathogen can provide novel drug targets but simultaneously requires a deeper and more detailed organism-specific understanding of the metabolism, which is often surprisingly sparse. In light of this, we reconstructed a genome-scale metabolic model of the pathogen Enterococcus faecalis V583. The manually curated metabolic network comprises 642 metabolites and 706 reactions. We experimentally determined metabolic profiles of E. faecalis grown in chemically defined medium in an anaerobic chemostat setup at different dilution rates and calculated the net uptake and product fluxes to constrain the model. We computed growth-associated energy and maintenance parameters and studied flux distributions through the metabolic network. Amino acid auxotrophies were identified experimentally for model validation and revealed seven essential amino acids. In addition, the important metabolic hub of glutamine/glutamate was altered by constructing a glutamine synthetase knockout mutant. The metabolic profile showed a slight shift in the fermentation pattern toward ethanol production and increased uptake rates of multiple amino acids, especially l-glutamine and l-glutamate. The model was used to understand the altered flux distributions in the mutant and provided an explanation for the experimentally observed redirection of the metabolic flux. We further highlighted the importance of gene-regulatory effects on the redirection of the metabolic fluxes upon perturbation. The genome-scale metabolic model presented here includes gene-protein-reaction associations, allowing a further use for biotechnological applications, for studying essential genes, proteins, or reactions, and the search for novel drug targets. link: http://identifiers.org/pubmed/25527553

Vempati2007_MMP9_Regulation: MODEL7888000034v0.0.1

This is the model described in the article: A biochemical model of matrix metalloproteinase 9 activation and inhibitio…

Details

Matrix metalloproteinases (MMPs) are a class of extracellular and membrane-bound proteases involved in an array of physiological processes, including angiogenesis. We present a detailed computational model of MMP9 activation and inhibition. Our model is validated to existing biochemical experimental data. We determine kinetic rate constants for the processes of MMP9 activation by MMP3, MMP10, MMP13, and trypsin; inhibition by the tissue inhibitors of metalloproteinases (TIMPs) 1 and 2; and MMP9 deactivation. This computational approach allows us to investigate discrepancies in our understanding of the interaction of MMP9 with TIMP1. Specifically, we find that inhibition due to a single binding event cannot describe MMP9 inhibition by TIMP1. Temporally accurate biphasic inhibition requires either an additional isomerization step or a second lower affinity isoform of MMP9. We also theoretically characterize the MMP3/TIMP2/pro-MMP9 and MMP3/TIMP1/pro-MMP9 systems. We speculate that these systems differ significantly in their time scales of activation and inhibition such that MMP9 is able to temporarily overshoot its final equilibrium value in the latter. Our numerical simulations suggest that the ability of pro-MMP9 to complex TIMP1 increases this overshoot. In all, our analysis serves as a summary of existing kinetic data for MMP9 and a foundation for future models utilizing MMP9 or other MMPs under physiologically well defined microenvironments. link: http://identifiers.org/pubmed/17848556

Venkatraman2011 - PLS-UPA behaviour in the presence of substrate competition_1_1_1_1: BIOMD0000000630v0.0.1

Venkatraman2011 - PLS-UPA behaviour in the presence of substrate competitionThe posibility of ultrasensitivity and bista…

Details

Plasmin (PLS) and urokinase-type plasminogen activator (UPA) are ubiquitous proteases that regulate the extracellular environment. Although they are secreted in inactive forms, they can activate each other through proteolytic cleavage. This mutual interplay creates the potential for complex dynamics, which we investigated using mathematical modeling and in vitro experiments. We constructed ordinary differential equations to model the conversion of precursor plasminogen into active PLS, and precursor urokinase (scUPA) into active urokinase (tcUPA). Although neither PLS nor UPA exhibits allosteric cooperativity, modeling showed that cooperativity occurred at the system level because of substrate competition. Computational simulations and bifurcation analysis predicted that the system would be bistable over a range of parameters for cooperativity and positive feedback. Cell-free experiments with recombinant proteins tested key predictions of the model. PLS activation in response to scUPA stimulus was found to be cooperative in vitro. Finally, bistability was demonstrated in vitro by the presence of two significantly different steady-state levels of PLS activation for the same levels of stimulus. We conclude that ultrasensitive, bistable activation of UPA-PLS is possible in the presence of substrate competition. An ultrasensitive threshold for activation of PLS and UPA would have ramifications for normal and disease processes, including angiogenesis, metastasis, wound healing, and fibrosis. link: http://identifiers.org/pubmed/22004735

Parameters:

NameDescription
parameter_1 = 0.084Reaction: species_2 => ; species_2, Rate Law: compartment_1*parameter_1*species_2
k1=0.9Reaction: species_4 + species_1 => species_2 + species_4; species_4, species_1, Rate Law: compartment_1*k1*species_4*species_1
k1=0.035Reaction: species_3 + species_1 => species_2 + species_3; species_3, species_1, Rate Law: compartment_1*k1*species_3*species_1
parameter_13 = 2.0; parameter_8=40.0Reaction: species_2 + species_3 => species_4 + species_2; species_2, species_3, Rate Law: compartment_1*parameter_8*species_2^parameter_13*species_3
v=0.01Reaction: => species_5, Rate Law: compartment_1*v
v=0.0032Reaction: => species_3, Rate Law: compartment_1*v
k1=0.02Reaction: species_6 => species_2; species_6, Rate Law: compartment_1*k1*species_6
parameter_2 = 0.032Reaction: species_5 => ; species_5, Rate Law: compartment_1*parameter_2*species_5
k2=0.016; k1=0.0Reaction: species_2 + species_5 => species_6; species_2, species_5, species_6, Rate Law: compartment_1*(k1*species_2*species_5-k2*species_6)

States:

NameDescription
species 2[Plasminogen]
species 6[Plasminogen; 10370340]
species 3[Urokinase-type plasminogen activator]
species 1[Plasminogen]
species 4[Urokinase-type plasminogen activator]
species 5[10370340]

Venkatraman2012 - Interplay between PLS and TSP1 in TGF-β1 activation: BIOMD0000000447v0.0.1

Venkatraman2012 - Interplay between PLS and TSP1 in TGF-β1 activationThe interplay between PLS (Plasmin) and TSP1 (Throm…

Details

Transforming growth factor-β1 (TGF-β1) is a potent regulator of extracellular matrix production, wound healing, differentiation, and immune response, and is implicated in the progression of fibrotic diseases and cancer. Extracellular activation of TGF-β1 from its latent form provides spatiotemporal control over TGF-β1 signaling, but the current understanding of TGF-β1 activation does not emphasize cross talk between activators. Plasmin (PLS) and thrombospondin-1 (TSP1) have been studied individually as activators of TGF-β1, and in this work we used a systems-level approach with mathematical modeling and in vitro experiments to study the interplay between PLS and TSP1 in TGF-β1 activation. Simulations and steady-state analysis predicted a switch-like bistable transition between two levels of active TGF-β1, with an inverse correlation between PLS and TSP1. In particular, the model predicted that increasing PLS breaks a TSP1-TGF-β1 positive feedback loop and causes an unexpected net decrease in TGF-β1 activation. To test these predictions in vitro, we treated rat hepatocytes and hepatic stellate cells with PLS, which caused proteolytic cleavage of TSP1 and decreased activation of TGF-β1. The TGF-β1 activation levels showed a cooperative dose response, and a test of hysteresis in the cocultured cells validated that TGF-β1 activation is bistable. We conclude that switch-like behavior arises from natural competition between two distinct modes of TGF-β1 activation: a TSP1-mediated mode of high activation and a PLS-mediated mode of low activation. This switch suggests an explanation for the unexpected effects of the plasminogen activation system on TGF-β1 in fibrotic diseases in vivo, as well as novel prognostic and therapeutic approaches for diseases with TGF-β dysregulation. link: http://identifiers.org/pubmed/23009856

Parameters:

NameDescription
parameter_2 = 0.35Reaction: species_2 + species_3 => species_4 + species_2; species_2, species_3, Rate Law: compartment_1*parameter_2*species_2*species_3
parameter_7 = 0.35Reaction: species_6 => species_6 + species_7; species_6, Rate Law: compartment_1*parameter_7*species_6
parameter_9 = 17.5; parameter_10 = 0.0245Reaction: species_7 + species_2 => species_9; species_7, species_2, species_9, Rate Law: compartment_1*(parameter_9*species_7*species_2-parameter_10*species_9)
parameter_8 = 1.05Reaction: species_6 => species_6 + species_8; species_6, Rate Law: compartment_1*parameter_8*species_6
parameter_17 = 0.0035; parameter_16 = 0.07Reaction: species_8 + species_3 => species_13; species_8, species_3, species_13, Rate Law: compartment_1*(parameter_16*species_8*species_3-parameter_17*species_13)
parameter_1 = 0.035Reaction: species_3 + species_1 => species_2 + species_3; species_3, species_1, Rate Law: compartment_1*parameter_1*species_3*species_1
parameter_20 = 0.0525Reaction: species_2 => ; species_2, Rate Law: compartment_1*parameter_20*species_2
parameter_22 = 0.0035Reaction: => species_3, Rate Law: compartment_1*parameter_22
parameter_14 = 0.035; parameter_15 = 0.0035Reaction: species_8 + species_4 => species_12; species_8, species_4, species_12, Rate Law: compartment_1*(parameter_14*species_8*species_4-parameter_15*species_12)
parameter_18 = 24.5Reaction: species_9 => ; species_9, Rate Law: compartment_1*parameter_18*species_9
parameter_23 = 0.035Reaction: => species_1, Rate Law: compartment_1*parameter_23
parameter_21 = 0.0175Reaction: species_7 => ; species_7, Rate Law: compartment_1*parameter_21*species_7
parameter_19 = 0.21Reaction: species_6 => ; species_6, Rate Law: compartment_1*parameter_19*species_6
parameter_6 = 0.005Reaction: species_5 => species_6; species_5, Rate Law: compartment_1*parameter_6*species_5
parameter_4 = 0.035Reaction: species_2 + species_5 => species_6 + species_2; species_2, species_5, Rate Law: compartment_1*parameter_4*species_2*species_5
parameter_5 = 24.5Reaction: species_7 + species_5 => species_6; species_7, species_5, Rate Law: compartment_1*parameter_5*species_7*species_5
parameter_3 = 1.4Reaction: species_4 + species_1 => species_2 + species_4; species_4, species_1, Rate Law: compartment_1*parameter_3*species_4*species_1
parameter_11 = 0.35Reaction: species_9 => species_2; species_9, Rate Law: compartment_1*parameter_11*species_9
parameter_12 = 24.5; parameter_13 = 0.0105Reaction: species_10 + species_2 => species_11; species_10, species_2, species_11, Rate Law: compartment_1*(parameter_12*species_10*species_2-parameter_13*species_11)

States:

NameDescription
species 9[Plasminogen; Thrombospondin 1]
species 2[Plasminogen]
species 6[Transforming growth factor beta-1]
species 10[Alpha-2-macroglobulin]
species 11[Plasminogen; Alpha-2-macroglobulin]
species 1[Plasminogen]
species 4[Urokinase-type plasminogen activator; active]
species 3[Urokinase-type plasminogen activator; inactive]
species 8[Plasminogen activator inhibitor 1]
species 12[Plasminogen activator inhibitor 1; Urokinase-type plasminogen activator; active]
species 7[Thrombospondin 1]
species 5[Transforming growth factor beta-1; inactive]
species 13[Urokinase-type plasminogen activator; Plasminogen activator inhibitor 1; inactive]

Verlingue2016 - Signalling pathway that control S-phase entry and geroconversion - Boolean Model: MODEL1611180000v0.0.1

Verlingue2016 - Signalling pathway that control S-phase entry and geroconversion - Boolean ModelThis model is described…

Details

Altered molecular responses to insulin and growth factors (GF) are responsible for late-life shortening diseases such as type-2 diabetes mellitus (T2DM) and cancers. We have built a network of the signaling pathways that control S-phase entry and a specific type of senescence called geroconversion. We have translated this network into a Boolean model to study possible cell phenotype outcomes under diverse molecular signaling conditions. In the context of insulin resistance, the model was able to reproduce the variations of the senescence level observed in tissues related to T2DM's main morbidity and mortality. Furthermore, by calibrating the pharmacodynamics of mTOR inhibitors, we have been able to reproduce the dose-dependent effect of rapamycin on liver degeneration and lifespan expansion in wild-type and HER2-neu mice. Using the model, we have finally performed an in silico prospective screen of the risk-benefit ratio of rapamycin dosage for healthy lifespan expansion strategies. We present here a comprehensive prognostic and predictive systems biology tool for human aging. link: http://identifiers.org/pubmed/27613445

Verma2016 - Ca(2+) Signal Propagation Along Hepatocyte Cords: BIOMD0000000834v0.0.1

Verma2016 - Ca(2+) Signal Propagation Along Hepatocyte CordsThis model is described in the article: [Computational Mode…

Details

The purpose of this study is to model the dynamics of lobular Ca(2+) wave propagation induced by an extracellular stimulus, and to analyze the effect of spatially systematic variations in cell-intrinsic signaling parameters on sinusoidal Ca(2+) response.We developed a computational model of lobular scale Ca(2+) signaling that accounts for receptor- mediated initiation of cell-intrinsic Ca(2+) signal in hepatocytes and its propagation to neighboring hepatocytes through gap junction-mediated molecular exchange.Analysis of the simulations showed that a pericentral-to-periportal spatial gradient in hormone sensitivity and/or rates of IP3 synthesis underlies the Ca(2+) wave propagation. We simulated specific cases corresponding to localized disruptions in the graded pattern of these parameters along a hepatic sinusoid. Simulations incorporating locally altered parameters exhibited Ca(2+) waves that do not propagate throughout the hepatic plate. Increased gap junction coupling restored normal Ca(2+) wave propagation when hepatocytes with low Ca(2+) signaling ability were localized in the midlobular or the pericentral region.Multiple spatial patterns in intracellular signaling parameters can lead to Ca(2+) wave propagation that is consistent with the experimentally observed spatial patterns of Ca(2+) dynamics. Based on simulations and analysis, we predict that increased gap junction-mediated intercellular coupling can induce robust Ca(2+) signals in otherwise poorly responsive hepatocytes, at least partly restoring the sinusoidally oriented Ca (2+) waves.Our bottom-up model of agonist-evoked spatial Ca(2+) patterns can be integrated with detailed descriptions of liver histology to study Ca(2+) regulation at the tissue level. link: http://identifiers.org/pubmed/27076052

Parameters:

NameDescription
k_ip315=0.8; H = 1.8E-4; k3 = 1.0; kcat = 0.45Reaction: => IP3_15; CaI_15, r_15, Rate Law: cytosol15*k_ip315*H*r_15*(1+(-k3*1/(CaI_15+k3)))*1/(kcat+r_15)
k_r3 = 1.928571Reaction: => r_3, Rate Law: cytosol3*k_r3
k_r6 = 1.785714; k_d = 0.34; H = 1.8E-4; k_HR = 1.0Reaction: r_6 =>, Rate Law: cytosol6*(k_d*r_6+k_HR*H*r_6+k_r6*r_6)
k_r7 = 1.571429; k_d = 0.34; H = 1.8E-4; k_HR = 1.0Reaction: r_7 =>, Rate Law: cytosol7*(k_d*r_7+k_HR*H*r_7+k_r7*r_7)
D = 1.6Reaction: IP3_4 =>, Rate Law: cytosol4*0.5*D*IP3_4
v=1.28571Reaction: => r_15, Rate Law: cytosol15*v
G = 0.9Reaction: IP3_3 => IP3_2, Rate Law: G*(IP3_3+(-IP3_2))*cytosol3
k_ip34=0.857143; H = 1.8E-4; k3 = 1.0; kcat = 0.45Reaction: => IP3_4; CaI_4, r_4, Rate Law: cytosol4*k_ip34*H*r_4*(1+(-k3*1/(CaI_4+k3)))*1/(kcat+r_4)
E = 1.0Reaction: => g_3; CaI_3, Rate Law: cytosol3*E*CaI_3^4*(1+(-g_3))
H = 1.8E-4; k3 = 1.0; k_ip311=0.714286; kcat = 0.45Reaction: => IP3_11; CaI_11, r_11, Rate Law: cytosol11*k_ip311*H*r_11*(1+(-k3*1/(CaI_11+k3)))*1/(kcat+r_11)
k_d = 0.34; H = 1.8E-4; k_r12 = 2.0; k_HR = 1.0Reaction: r_12 =>, Rate Law: cytosol12*(k_d*r_12+k_HR*H*r_12+k_r12*r_12)
k_r13 = 1.214286Reaction: => r_13, Rate Law: cytosol13*k_r13
k_d = 0.34; H = 1.8E-4; k_r9 = 1.142857; k_HR = 1.0Reaction: r_9 =>, Rate Law: cytosol9*(k_d*r_9+k_HR*H*r_9+k_r9*r_9)
B = 0.082; k2 = 0.15Reaction: CaI_3 => CaT_3, Rate Law: B*CaI_3^2*1/(k2^2+CaI_3^2)*cytosol3
k_r14 = 1.071429Reaction: => r_14, Rate Law: cytosol14*k_r14
k_d = 0.34; H = 1.8E-4; k_r4 = 1.357143; k_HR = 1.0Reaction: r_4 =>, Rate Law: cytosol4*(k_d*r_4+k_HR*H*r_4+k_r4*r_4)
k_ip314=0.7; H = 1.8E-4; k3 = 1.0; kcat = 0.45Reaction: => IP3_14; CaI_14, r_14, Rate Law: cytosol14*k_ip314*H*r_14*(1+(-k3*1/(CaI_14+k3)))*1/(kcat+r_14)
k_r13 = 1.214286; k_d = 0.34; H = 1.8E-4; k_HR = 1.0Reaction: r_13 =>, Rate Law: cytosol13*(k_d*r_13+k_HR*H*r_13+k_r13*r_13)
k_r10 = 1.642857Reaction: => r_10, Rate Law: cytosol10*k_r10
k_r15=1.28571; k_d = 0.34; H = 1.8E-4; k_HR = 1.0Reaction: r_15 =>, Rate Law: cytosol15*(k_d*r_15+k_HR*H*r_15+k_r15*r_15)
k_r8 = 1.714286; k_d = 0.34; H = 1.8E-4; k_HR = 1.0Reaction: r_8 =>, Rate Law: cytosol8*(k_d*r_8+k_HR*H*r_8+k_r8*r_8)
k_r14 = 1.071429; k_d = 0.34; H = 1.8E-4; k_HR = 1.0Reaction: r_14 =>, Rate Law: cytosol14*(k_d*r_14+k_HR*H*r_14+k_r14*r_14)
k_r7 = 1.571429Reaction: => r_7, Rate Law: cytosol7*k_r7
k_ip37=0.742857; H = 1.8E-4; k3 = 1.0; kcat = 0.45Reaction: => IP3_7; CaI_7, r_7, Rate Law: cytosol7*k_ip37*H*r_7*(1+(-k3*1/(CaI_7+k3)))*1/(kcat+r_7)
L = 1.5E-4; A = 0.2; k1 = 0.5Reaction: CaT_10 => CaI_10; g_10, IP3_10, Rate Law: (1+(-g_10))*(A*(0.5*IP3_10)^4*1/(k1+0.5*IP3_10)^4+L)*(CaT_10+(-CaI_10))*store10
k_ip38=0.828571; H = 1.8E-4; k3 = 1.0; kcat = 0.45Reaction: => IP3_8; CaI_8, r_8, Rate Law: cytosol8*k_ip38*H*r_8*(1+(-k3*1/(CaI_8+k3)))*1/(kcat+r_8)
k_d = 0.34; H = 1.8E-4; k_r10 = 1.642857; k_HR = 1.0Reaction: r_10 =>, Rate Law: cytosol10*(k_d*r_10+k_HR*H*r_10+k_r10*r_10)
k_r8 = 1.714286Reaction: => r_8, Rate Law: cytosol8*k_r8
k_ip35=0.842857; H = 1.8E-4; k3 = 1.0; kcat = 0.45Reaction: => IP3_5; CaI_5, r_5, Rate Law: cytosol5*k_ip35*H*r_5*(1+(-k3*1/(CaI_5+k3)))*1/(kcat+r_5)
k_d = 0.34; k_r11 = 1.428571; H = 1.8E-4; k_HR = 1.0Reaction: r_11 =>, Rate Law: cytosol11*(k_d*r_11+k_HR*H*r_11+k_r11*r_11)
F = 0.01Reaction: g_3 => ; CaI_3, Rate Law: cytosol3*F

States:

NameDescription
CaI 12cytosolic Ca2+_CaI_12
CaI 7cytosolic Ca2+_CaI_7
r 10total receptor levels _r_10
CaI 1[Calcium cation]
r 3total receptor levels _r_3
r 9total receptor levels _r_9
g 5total IP3R_g_5
IP3 1cytosolic IP3_IP3_1
IP3 5cytosolic IP3_IP3_5
g 3total IP3R_g_3
CaT 4total intracellular store Ca2+ content_CaT_4
CaI 15cytosolic Ca2+_CaI_15
CaT 3total intracellular store Ca2+ content_CaT_3
CaT 12total intracellular store Ca2+ content_CaT_12
CaI 5cytosolic Ca2+_CaI_5
IP3 6cytosolic IP3_IP3_6
r 12total receptor levels _r_12
g 6total IP3R_g_6
r 11total receptor levels _r_11
g 12total IP3R_g_12
CaI 13cytosolic Ca2+_CaI_13
CaT 5total intracellular store Ca2+ content_CaT_5
CaI 6cytosolic Ca2+_CaI_6
r 15total receptor levels _r_15
IP3 4cytosolic IP3_IP3_4
CaT 7total intracellular store Ca2+ content_CaT_7
CaT 11total intracellular store Ca2+ content_CaT_11
g 4total IP3R_g_4
CaT 15total intracellular store Ca2+ content_CaT_15
CaI 10cytosolic Ca2+_CaI_10
g 11total IP3R_g_11
r 7total receptor levels_ r_7
CaI 4cytosolic Ca2+_CaI_4
r 13total receptor levels _r_13
IP3 15cytosolic IP3_IP3_15
g 9total IP3R_g_9
g 14total IP3R_g_14
CaT 14total intracellular store Ca2+ content_CaT_14
IP3 9cytosolic IP3_IP3_9
CaT 13total intracellular store Ca2+ content_CaT_13
CaI 9cytosolic Ca2+_CaI_9
IP3 7cytosolic IP3_IP3_7
IP3 11cytosolic IP3_IP3_11
r 14total receptor levels _r_14
g 7total IP3R_g_7
r 4total receptor levels _r_4
g 15total IP3R_g_15
CaT 6total intracellular store Ca2+ content_CaT_6
CaT 9total intracellular store Ca2+ content_CaT_9
IP3 14cytosolic IP3_IP3_14
g 8total IP3R_g_8
IP3 13cytosolic IP3_IP3_13
CaI 8cytosolic Ca2+_CaI_8
IP3 8cytosolic IP3_IP3_8
IP3 3cytosolic IP3_IP3_3
r 6total receptor levels _r_6
r 8total receptor levels _r_8
CaI 14cytosolic Ca2+_CaI_14
g 13total IP3R_g_13
CaT 8total intracellular store Ca2+ content_CaT_8

Verma2016 - HIV and HPV co-infection, T-cell response: BIOMD0000000872v0.0.1

This is a COPASI version of the HIV/HPV coinfection model submitted to PLoS One. Title: Modeling the mechanisms by whic…

Details

Human immunodeficiency virus (HIV)-infected patients are at an increased risk of co-infection with human papilloma virus (HPV), and subsequent malignancies such as oral cancer. To determine the role of HIV-associated immune suppression on HPV persistence and pathogenesis, and to investigate the mechanisms underlying the modulation of HPV infection and oral cancer by HIV, we developed a mathematical model of HIV/HPV co-infection. Our model captures known immunological and molecular features such as impaired HPV-specific effector T helper 1 (Th1) cell responses, and enhanced HPV infection due to HIV. We used the model to determine HPV prognosis in the presence of HIV infection, and identified conditions under which HIV infection alters HPV persistence in the oral mucosa system. The model predicts that conditions leading to HPV persistence during HIV/HPV co-infection are the permissive immune environment created by HIV and molecular interactions between the two viruses. The model also determines when HPV infection continues to persist in the short run in a co-infected patient undergoing antiretroviral therapy. Lastly, the model predicts that, under efficacious antiretroviral treatment, HPV infections will decrease in the long run due to the restoration of CD4+ T cell numbers and protective immune responses. link: http://identifiers.org/doi/10.1371/journal.pone.0168133

Parameters:

NameDescription
mu = 0.048; k2=1000.0Reaction: => s14; s13, Rate Law: default*Production_of_HPV_due_to_HPV_self_proliferating_1(k2, mu, s13)
k1=1.0Reaction: s3 =>, Rate Law: default*k1*s3
beta = 1.97002141327623E-7; e_rt = 0.0Reaction: s4 => s3; s2, Rate Law: default*Rate_Law_for_production_of_HIV_infected_cells_1(beta, e_rt, s2, s4)
k1=0.05Reaction: s14 =>, Rate Law: default*k1*s14
epi = 0.5; r=0.1Reaction: s13 => s13; s13, Rate Law: default*Proliferation_of_HPV_self_proliferating(epi, r, s13)
c1 = 23.0Reaction: s2 =>, Rate Law: default*c1*s2
k1=1000.0; mu = 0.048Reaction: => s14; s12, Rate Law: default*Rate_Law_for_Production_of_HPV_due_to_HPVinfected_1(k1, mu, s12)
s = 4864.02569593148Reaction: => s4, Rate Law: default*Constant_flux__irreversible(s)
e_pi = 0.0; delta=1.0; N1 = 467.0Reaction: => s2; s3, Rate Law: default*Rate_Law_for_Production_of_HIV_virion_1(N1, delta, e_pi, s3)
b=3.5E-5; omega = 0.001Reaction: s16 => ; s13, s4, Rate Law: default*Logistic_term_for_Effector_cells_1(b, omega, s13, s16, s4)
N2=10000.0; psi=0.0067; phi=1000000.0; p=2.0833E-5Reaction: s12 => s12; s14, s2, Rate Law: default*Productionof_HPV_infected_cells(N2, p, phi, psi, s12, s14, s2)
d = 0.01Reaction: s4 =>, Rate Law: default*d*s4
k1=0.048Reaction: s13 =>, Rate Law: default*k1*s13
epi = 0.5Reaction: s12 => s13, Rate Law: default*epi*s12
mu = 0.048Reaction: s12 =>, Rate Law: default*mu*s12
a=0.01Reaction: s13 => ; s16, Rate Law: default*Death_of_HPV_self_proliferating_cells_due_to_effector_cells(a, s13, s16)
omega = 0.001Reaction: s16 => s16; s13, Rate Law: default*Rate_Law_for_Production_of_Effector_cell_1_1(omega, s13, s16)

States:

NameDescription
s14[0004510]
s13[Epithelial Cell]
s12[Epithelial Cell]
s2[Human Immunodeficiency Virus]
s4[C97350]
s3[C97350]
s16[C12543]

Vernoux2011_AuxinSignaling_AuxinFluctuating: BIOMD0000000352v0.0.1

This model is from the article: The auxin signalling network translates dynamic input into robust patterning at the sh…

Details

The plant hormone auxin is thought to provide positional information for patterning during development. It is still unclear, however, precisely how auxin is distributed across tissues and how the hormone is sensed in space and time. The control of gene expression in response to auxin involves a complex network of over 50 potentially interacting transcriptional activators and repressors, the auxin response factors (ARFs) and Aux/IAAs. Here, we perform a large-scale analysis of the Aux/IAA-ARF pathway in the shoot apex of Arabidopsis, where dynamic auxin-based patterning controls organogenesis. A comprehensive expression map and full interactome uncovered an unexpectedly simple distribution and structure of this pathway in the shoot apex. A mathematical model of the Aux/IAA-ARF network predicted a strong buffering capacity along with spatial differences in auxin sensitivity. We then tested and confirmed these predictions using a novel auxin signalling sensor that reports input into the signalling pathway, in conjunction with the published DR5 transcriptional output reporter. Our results provide evidence that the auxin signalling network is essential to create robust patterns at the shoot apex. link: http://identifiers.org/pubmed/21734647

Parameters:

NameDescription
d_A = 0.003Reaction: A =>, Rate Law: d_A*A
d_r = 0.007Reaction: R =>, Rate Law: d_r*R
pi_A = 1.0Reaction: => A, Rate Law: pi_A
d_IA = 0.003Reaction: D_IA =>, Rate Law: d_IA*D_IA
d_II = 0.003Reaction: D_II =>, Rate Law: d_II*D_II
pi_I = 1.0Reaction: => I; R, Rate Law: pi_I*R
w_A = 10.0; f_c = 10.0; B_d = 100.0; f_A = 10.0; K_IA = 10.0; w_I = 10.0; k_Am = 10.0; w_D = 10.0Reaction: => R; A, D_IA, I, Rate Law: (1+f_c/B_d*A*(1+w_A*f_A*A/B_d))/(1+A/B_d*(1+w_A*A/B_d)+w_I*A*I/(K_IA*B_d)+w_D*D_IA/B_d+k_Am)
K_aux = 1.0; d_I = 0.05; gamma_I = 10.0Reaction: I => ; aux, Rate Law: gamma_I*d_I*K_aux*aux/(K_aux*aux+1)*I
kprime_II = 10.0; k_II = 1.0Reaction: I + I => D_II, Rate Law: k_II*I*I-kprime_II*D_II
k_IA = 1.0; kprime_IA = 10.0Reaction: A + I => D_IA, Rate Law: k_IA*I*A-kprime_IA*D_IA

States:

NameDescription
I[Auxin-responsive protein IAA1]
A[Auxin response factor 2]
aux[Auxin transporter protein 1]
D II[Auxin-responsive protein IAA1]
R[messenger RNA]
D IA[Auxin-responsive protein IAA1; Auxin response factor 2]

Vernoux2011_AuxinSignaling_AuxinSingleStepInput: BIOMD0000000351v0.0.1

This model is from the article: The auxin signalling network translates dynamic input into robust patterning at the sh…

Details

The plant hormone auxin is thought to provide positional information for patterning during development. It is still unclear, however, precisely how auxin is distributed across tissues and how the hormone is sensed in space and time. The control of gene expression in response to auxin involves a complex network of over 50 potentially interacting transcriptional activators and repressors, the auxin response factors (ARFs) and Aux/IAAs. Here, we perform a large-scale analysis of the Aux/IAA-ARF pathway in the shoot apex of Arabidopsis, where dynamic auxin-based patterning controls organogenesis. A comprehensive expression map and full interactome uncovered an unexpectedly simple distribution and structure of this pathway in the shoot apex. A mathematical model of the Aux/IAA-ARF network predicted a strong buffering capacity along with spatial differences in auxin sensitivity. We then tested and confirmed these predictions using a novel auxin signalling sensor that reports input into the signalling pathway, in conjunction with the published DR5 transcriptional output reporter. Our results provide evidence that the auxin signalling network is essential to create robust patterns at the shoot apex. link: http://identifiers.org/pubmed/21734647

Parameters:

NameDescription
d_A = 0.003Reaction: A =>, Rate Law: d_A*A
d_r = 0.007Reaction: R =>, Rate Law: d_r*R
pi_A = 1.0Reaction: => A, Rate Law: pi_A
d_IA = 0.003Reaction: D_IA =>, Rate Law: d_IA*D_IA
d_II = 0.003Reaction: D_II =>, Rate Law: d_II*D_II
pi_I = 1.0Reaction: => I; R, Rate Law: pi_I*R
w_A = 10.0; f_c = 10.0; B_d = 100.0; f_A = 10.0; K_IA = 10.0; w_I = 10.0; k_Am = 10.0; w_D = 10.0Reaction: => R; A, D_IA, I, Rate Law: (1+f_c/B_d*A*(1+w_A*f_A*A/B_d))/(1+A/B_d*(1+w_A*A/B_d)+w_I*A*I/(K_IA*B_d)+w_D*D_IA/B_d+k_Am)
K_aux = 1.0; d_I = 0.05; gamma_I = 10.0Reaction: I => ; aux, Rate Law: gamma_I*d_I*K_aux*aux/(K_aux*aux+1)*I
kprime_II = 10.0; k_II = 1.0Reaction: I + I => D_II, Rate Law: k_II*I*I-kprime_II*D_II
k_IA = 1.0; kprime_IA = 10.0Reaction: A + I => D_IA, Rate Law: k_IA*I*A-kprime_IA*D_IA

States:

NameDescription
I[Auxin-responsive protein IAA1]
A[Auxin response factor 2]
aux[Auxin transporter protein 1]
D II[Auxin-responsive protein IAA1]
R[messenger RNA]
D IA[Auxin response factor 2; Auxin-responsive protein IAA1]

Vibert2017 - Tcell proliferation model: MODEL2003170002v0.0.1

model allows us to infer proliferation rates and cell cycle phase durations from complex experimental 5-ethynyl-2'-deoxy…

Details

Cell proliferation is the common characteristic of all biological systems. The immune system insures the maintenance of body integrity on the basis of a continuous production of diversified T lymphocytes in the thymus. This involves processes of proliferation, differentiation, selection, death and migration of lymphocytes to peripheral tissues, where proliferation also occurs upon antigen recognition. Quantification of cell proliferation dynamics requires specific experimental methods and mathematical modelling. Here, we assess the impact of genetics and aging on the immune system by investigating the dynamics of proliferation of T lymphocytes across their differentiation through thymus and spleen in mice. Our investigation is based on single-cell multicolour flow cytometry analysis revealing the active incorporation of a thymidine analogue during S phase after pulse-chase-pulse experiments in vivo, versus cell DNA content. A generic mathematical model of state transition simulates through Ordinary Differential Equations (ODEs) the evolution of single cell behaviour during various durations of labelling. It allows us to fit our data, to deduce proliferation rates and estimate cell cycle durations in sub-populations. Our model is simple and flexible and is validated with other durations of pulse/chase experiments. Our results reveal that T cell proliferation is highly heterogeneous but with a specific "signature" that depends upon genetic origins, is specific to cell differentiation stages in thymus and spleen and is altered with age. In conclusion, our model allows us to infer proliferation rates and cell cycle phase durations from complex experimental 5-ethynyl-2'-deoxyuridine (EdU) data, revealing T cell proliferation heterogeneity and specific signatures. link: http://identifiers.org/pubmed/28288157

Viertel2019 - A Computational model of the mammalian external tufted cell: BIOMD0000000844v0.0.1

This is a mathematical conductance-based model of the bursting activity in external tufted (ET) cells of the olfactory b…

Details

We introduce a novel detailed conductance-based model of the bursting activity in external tufted (ET) cells of the olfactory bulb. We investigate the mechanisms underlying their bursting, and make experimentally-testable predictions. The ionic currents included in the model are specific to ET cells, and their kinetic and other parameters are based on experimental recordings. We validate the model by showing that its bursting characteristics under various conditions (e.g. blocking various currents) are consistent with experimental observations. Further, we identify the bifurcation structure and dynamics that explain bursting behavior. This analysis allows us to make predictions of the response of the cell to current pulses at different burst phases. We find that depolarizing (but not hyperpolarizing) inputs received during the interburst interval can advance burst timing, creating the substrate for synchronization by excitatory connections. It has been hypothesized that such synchronization among the ET cells within one glomerulus might help coordinate the glomerular output. Next we investigate model parameter sensitivity and identify parameters that play the most prominent role in controlling each burst characteristic, such as the burst frequency and duration. Finally, the response of the cell to periodic inputs is examined, reflecting the sniffing-modulated input that these cell receive in vivo. We find that individual cells can be better entrained by inputs with higher, rather than lower, frequencies than the intrinsic bursting frequency of the cell. Nevertheless, a heterogeneous population of ET cells (as may be found in a glomerulus) is able to produce reliable periodic population responses even at lower input frequencies. link: http://identifiers.org/pubmed/30290156

Parameters:

NameDescription
hNaP_tau = 483.668077978459; hNaP_inf = 0.373251468077049Reaction: => hNaP, Rate Law: compartment*(hNaP_inf-hNaP)/hNaP_tau
nHVK_tau = 1000.00007479349; nHVK_inf = 2.244842984971E-5Reaction: => nHVK, Rate Law: compartment*(nHVK_inf-nHVK)/nHVK_tau
tau_Ca = 8.0; Ca0 = 2.0E-5Reaction: => Ca, Rate Law: compartment*(Ca0-Ca)/tau_Ca
hH_tau = 7.13025057731447; hH_inf = 0.155405252349385Reaction: => hH, Rate Law: compartment*(hH_inf-hH)/hH_tau
hLVA_tau = 329.955639297499; hLVA_inf = 0.333222156222541Reaction: => hLVA, Rate Law: compartment*(hLVA_inf-hLVA)/hLVA_tau
mLVA_tau = 17.5876479384678; mLVA_inf = 0.0509254933768459Reaction: => mLVA, Rate Law: compartment*(mLVA_inf-mLVA)/mLVA_tau
IBK = 31.678826380681; INa = -0.012838612439222; IHVA = -0.585636135043006; INaP = -6.09894732028694; C = 21.0; ILVA = -1.31979373465397; IK = 0.00668833084914886; IHVK = 0.0878809960822716; IL = 27.7286628129031; IH = -51.7633892852534Reaction: V =>, Rate Law: compartment*(INa+IK+ILVA+IH+INaP+IL+IHVA+IBK+IHVK)/C
d = 1.0; Ca_buffer = 0.5; F = 96485.0; IHVA = -0.585636135043006; ILVA = -1.31979373465397; Ca_z = 2.0Reaction: Ca =>, Rate Law: compartment*Ca_buffer*10*(ILVA+IHVA)/(Ca_z*F*d)
nK_Inf = 0.0560848507623637; nK_tau = 4.60171012895541Reaction: => nK, Rate Law: compartment*(nK_Inf-nK)/nK_tau
mBK_inf = 2.00990799551082E-5; mBK_tau = 219.103190338819Reaction: => mBK, Rate Law: compartment*(mBK_inf-mBK)/mBK_tau

States:

NameDescription
hNaP[Electrical Current; C830]
nHVK[C765; Electrical Current]
mLVA[Electrical Current; C331]
V[SBO:0000259]
hLVA[C331; Electrical Current]
Ca[C331]
hH[Electrical Current]
nK[Electrical Current; C765]
mBK[C765; Electrical Current]

Viladomiu2012 - PPARgamma role in C.diff associated disease: MODEL1210260004v0.0.1

"Modeling the Role of Peroxisome Proliferator-Activated Receptor γ and MicroRNA-146 in Mucosal Immune Responses to Clost…

Details

Clostridium difficile is an anaerobic bacterium that has re-emerged as a facultative pathogen and can cause nosocomial diarrhea, colitis or even death. Peroxisome proliferator-activated receptor (PPAR) γ has been implicated in the prevention of inflammation in autoimmune and infectious diseases; however, its role in the immunoregulatory mechanisms modulating host responses to C. difficile and its toxins remains largely unknown. To characterize the role of PPARγ in C. difficile-associated disease (CDAD), immunity and gut pathology, we used a mouse model of C. difficile infection in wild-type and T cell-specific PPARγ null mice. The loss of PPARγ in T cells increased disease activity and colonic inflammatory lesions following C. difficile infection. Colonic expression of IL-17 was upregulated and IL-10 downregulated in colons of T cell-specific PPARγ null mice. Also, both the loss of PPARγ in T cells and C. difficile infection favored Th17 responses in spleen and colonic lamina propria of mice with CDAD. MicroRNA (miRNA)-sequencing analysis and RT-PCR validation indicated that miR-146b was significantly overexpressed and nuclear receptor co-activator 4 (NCOA4) suppressed in colons of C. difficile-infected mice. We next developed a computational model that predicts the upregulation of miR-146b, downregulation of the PPARγ co-activator NCOA4, and PPARγ, leading to upregulation of IL-17. Oral treatment of C. difficile-infected mice with the PPARγ agonist pioglitazone ameliorated colitis and suppressed pro-inflammatory gene expression. In conclusion, our data indicates that miRNA-146b and PPARγ activation may be implicated in the regulation of Th17 responses and colitis in C. difficile-infected mice. link: http://identifiers.org/pubmed/23071818

Vilar2002_Oscillator: BIOMD0000000035v0.0.1

# # Minimal Model for Circadian Oscillations CitationVilar JMG, Kueh HY, Barkai N, Leibler S, (2002) . M…

Details

A wide range of organisms use circadian clocks to keep internal sense of daily time and regulate their behavior accordingly. Most of these clocks use intracellular genetic networks based on positive and negative regulatory elements. The integration of these "circuits" at the cellular level imposes strong constraints on their functioning and design. Here, we study a recently proposed model [Barkai, N. & Leibler, S. (2000) Nature (London), 403, 267-268] that incorporates just the essential elements found experimentally. We show that this type of oscillator is driven mainly by two elements: the concentration of a repressor protein and the dynamics of an activator protein forming an inactive complex with the repressor. Thus, the clock does not need to rely on mRNA dynamics to oscillate, which makes it especially resistant to fluctuations. Oscillations can be present even when the time average of the number of mRNA molecules goes below one. Under some conditions, this oscillator is not only resistant to but, paradoxically, also enhanced by the intrinsic biochemical noise. link: http://identifiers.org/pubmed/11972055

Parameters:

NameDescription
alphaAp=500.0Reaction: DAp => DAp + MA, Rate Law: DAp*alphaAp
alphaA=50.0Reaction: DA => DA + MA, Rate Law: DA*alphaA
alphaR=0.01Reaction: DR => DR + MR, Rate Law: DR*alphaR
deltaR=0.2Reaction: R => EmptySet, Rate Law: R*deltaR
betaR=5.0Reaction: MR => MR + R, Rate Law: MR*betaR
deltaMR=0.5Reaction: MR => EmptySet, Rate Law: MR*deltaMR
betaA=50.0Reaction: MA => A + MA, Rate Law: MA*betaA
gammaA=1.0Reaction: A + DA => DAp, Rate Law: A*DA*gammaA
alphaRp=50.0Reaction: DRp => DRp + MR, Rate Law: DRp*alphaRp
thetaR=100.0Reaction: DRp => A + DR, Rate Law: DRp*thetaR
deltaMA=10.0Reaction: MA => EmptySet, Rate Law: MA*deltaMA
gammaC=2.0Reaction: A + R => C, Rate Law: A*R*gammaC
thetaA=50.0Reaction: DAp => A + DA, Rate Law: DAp*thetaA
gammaR=1.0Reaction: A + DR => DRp, Rate Law: A*DR*gammaR
deltaA=1.0Reaction: A => EmptySet, Rate Law: A*deltaA

States:

NameDescription
DRDR
A[protein]
MR[messenger RNA]
C[protein]
DRpDRp
MA[messenger RNA]
DADA
R[protein]
DApDAp

Vilar2006_TGFbeta: BIOMD0000000101v0.0.1

The model reproduces Fig 5A of the paper. The ligand concentration is increased from 3E-5 to 0.01 at time t=2500 to ensu…

Details

The TGF-beta pathway plays a central role in tissue homeostasis and morphogenesis. It transduces a variety of extracellular signals into intracellular transcriptional responses that control a plethora of cellular processes, including cell growth, apoptosis, and differentiation. We use computational modeling to show that coupling of signaling with receptor trafficking results in a highly versatile signal-processing unit, able to sense by itself absolute levels of ligand, temporal changes in ligand concentration, and ratios of multiple ligands. This coupling controls whether the response of the receptor module is transient or permanent and whether or not different signaling channels behave independently of each other. Our computational approach unifies seemingly disparate experimental observations and suggests specific changes in receptor trafficking patterns that can lead to phenotypes that favor tumor progression. link: http://identifiers.org/pubmed/16446785

Parameters:

NameDescription
klid = 0.25Reaction: lRIRII =>, Rate Law: klid*lRIRII
ki = 0.3333333333333Reaction: lRIRII => lRIRII_endo, Rate Law: ki*lRIRII
pRI = 8.0Reaction: => RI, Rate Law: pRI
ka = 1.0; ligand = 3.0E-5Reaction: RII + RI => lRIRII, Rate Law: ka*ligand*RI*RII
pRII = 4.0Reaction: => RII, Rate Law: pRII
kcd = 0.0277777778Reaction: RII =>, Rate Law: kcd*RII
kr = 0.0333333333333333Reaction: RII_endo => RII, Rate Law: kr*RII_endo

States:

NameDescription
RI endo[TGF-beta receptor type-1]
lRIRII[Transforming growth factor beta-1; TGF-beta receptor type-1; TGF-beta receptor type-2]
RII[TGF-beta receptor type-2]
lRIRII endo[Transforming growth factor beta-1; TGF-beta receptor type-1; TGF-beta receptor type-2]
RI[TGF-beta receptor type-1]
RII endo[TGF-beta receptor type-2]

Villanova2017 - Mixotrophic metabolism in the model diatom Phaeodactylum tricornutum: MODEL2102080001v0.0.1

Genome-scale metabolic model of Phaeodactylum tricornutum

Details

Diatoms are prominent marine microalgae, interesting not only from an ecological point of view, but also for their possible use in biotechnology applications. They can be cultivated in phototrophic conditions, using sunlight as the sole energy source. Some diatoms, however, can also grow in a mixotrophic mode, wherein both light and external reduced carbon contribute to biomass accumulation. In this study, we investigated the consequences of mixotrophy on the growth and metabolism of the pennate diatom Phaeodactylum tricornutum, using glycerol as the source of reduced carbon. Transcriptomics, metabolomics, metabolic modelling and physiological data combine to indicate that glycerol affects the central-carbon, carbon-storage and lipid metabolism of the diatom. In particular, provision of glycerol mimics typical responses of nitrogen limitation on lipid metabolism at the level of triacylglycerol accumulation and fatty acid composition. The presence of glycerol, despite provoking features reminiscent of nutrient limitation, neither diminishes photosynthetic activity nor cell growth, revealing essential aspects of the metabolic flexibility of these microalgae and suggesting possible biotechnological applications of mixotrophy. link: http://identifiers.org/doi/10.1098/rstb.2016.0404

Vinnakota2006_MuscleGlycogenolysis_ModelA: MODEL1006230077v0.0.1

This a model from the article: Dynamics of muscle glycogenolysis modeled with pH time course computation and pH-depend…

Details

Cellular metabolites are moieties defined by their specific binding constants to H+, Mg2+, and K+ or anions without ligands. As a consequence, every biochemical reaction in the cytoplasm has an associated proton stoichiometry that is generally noninteger- and pH-dependent. Therefore, with metabolic flux, pH is altered in a medium with finite buffer capacity. Apparent equilibrium constants and maximum enzyme velocities, which are functions of pH, are also altered. We augmented an earlier mathematical model of skeletal muscle glycogenolysis with pH-dependent enzyme kinetics and reaction equilibria to compute the time course of pH changes. Analysis shows that kinetics and final equilibrium states of the closed system are highly constrained by the pH-dependent parameters. This kinetic model of glycogenolysis, coupled to creatine kinase and adenylate kinase, simulated published experiments made with a cell-free enzyme mixture to reconstitute the network and to synthesize PCr and lactate in vitro. Using the enzyme kinetic and thermodynamic data in the literature, the simulations required minimal adjustments of parameters to describe the data. These results show that incorporation of appropriate physical chemistry of the reactions with accurate kinetic modeling gives a reasonable simulation of experimental data and is necessary for a physically correct representation of the metabolic network. The approach is general for modeling metabolic networks beyond the specific pathway and conditions presented here. link: http://identifiers.org/pubmed/16617075

Vinnakota2006_MuscleGlycogenolysis_ModelB: MODEL1006230053v0.0.1

This a model from the article: Dynamics of muscle glycogenolysis modeled with pH time course computation and pH-depend…

Details

Cellular metabolites are moieties defined by their specific binding constants to H+, Mg2+, and K+ or anions without ligands. As a consequence, every biochemical reaction in the cytoplasm has an associated proton stoichiometry that is generally noninteger- and pH-dependent. Therefore, with metabolic flux, pH is altered in a medium with finite buffer capacity. Apparent equilibrium constants and maximum enzyme velocities, which are functions of pH, are also altered. We augmented an earlier mathematical model of skeletal muscle glycogenolysis with pH-dependent enzyme kinetics and reaction equilibria to compute the time course of pH changes. Analysis shows that kinetics and final equilibrium states of the closed system are highly constrained by the pH-dependent parameters. This kinetic model of glycogenolysis, coupled to creatine kinase and adenylate kinase, simulated published experiments made with a cell-free enzyme mixture to reconstitute the network and to synthesize PCr and lactate in vitro. Using the enzyme kinetic and thermodynamic data in the literature, the simulations required minimal adjustments of parameters to describe the data. These results show that incorporation of appropriate physical chemistry of the reactions with accurate kinetic modeling gives a reasonable simulation of experimental data and is necessary for a physically correct representation of the metabolic network. The approach is general for modeling metabolic networks beyond the specific pathway and conditions presented here. link: http://identifiers.org/pubmed/16617075

Vinnakota2006_MuscleGlycogenolysis_ModelC: MODEL1006230049v0.0.1

This a model from the article: Dynamics of muscle glycogenolysis modeled with pH time course computation and pH-depend…

Details

Cellular metabolites are moieties defined by their specific binding constants to H+, Mg2+, and K+ or anions without ligands. As a consequence, every biochemical reaction in the cytoplasm has an associated proton stoichiometry that is generally noninteger- and pH-dependent. Therefore, with metabolic flux, pH is altered in a medium with finite buffer capacity. Apparent equilibrium constants and maximum enzyme velocities, which are functions of pH, are also altered. We augmented an earlier mathematical model of skeletal muscle glycogenolysis with pH-dependent enzyme kinetics and reaction equilibria to compute the time course of pH changes. Analysis shows that kinetics and final equilibrium states of the closed system are highly constrained by the pH-dependent parameters. This kinetic model of glycogenolysis, coupled to creatine kinase and adenylate kinase, simulated published experiments made with a cell-free enzyme mixture to reconstitute the network and to synthesize PCr and lactate in vitro. Using the enzyme kinetic and thermodynamic data in the literature, the simulations required minimal adjustments of parameters to describe the data. These results show that incorporation of appropriate physical chemistry of the reactions with accurate kinetic modeling gives a reasonable simulation of experimental data and is necessary for a physically correct representation of the metabolic network. The approach is general for modeling metabolic networks beyond the specific pathway and conditions presented here. link: http://identifiers.org/pubmed/16617075

Vinnakota2010_TranscientAnoia_EDLmuscle: MODEL1006230100v0.0.1

This a model from the article: Common phenotype of resting mouse extensor digitorum longus and soleus muscles: equal A…

Details

Rates of ATPase and glycolysis are several times faster in actively contracting mouse extensor digitorum longus muscle (EDL) than soleus (SOL), but we find these rates are not distinguishable at rest. We used a transient anoxic perturbation of steady state energy balance to decrease phosphocreatine (PCr) reversibly and to measure the rates of ATPase and of lactate production without muscle activation or contraction. The rate of glycolytic ATP synthesis is less than the ATPase rate, accounting for the continual PCr decrease during anoxia in both muscles. We fitted a mathematical model validated with properties of enzymes and solutes measured in vitro and appropriate for the transient perturbation of these muscles to experimental data to test whether the model accounts for the results. Simulations showed equal rates of ATPase and lactate production in both muscles. ATPase controls glycolytic flux by feedback from its products. Adenylate kinase function is critical because a rise in [AMP] is necessary to activate glycogen phosphorylase. ATPase is the primary source of H+ production. The sum of contributions of the 13 reactions of the glycogenolytic and glycolytic network to total proton load is negligible. The stoichiometry of lactate and H+ production is near unity. These results identify a default state of energy metabolism for resting muscle in which there is no difference in the metabolic phenotype of EDL and SOL. Therefore, additional control mechanisms, involving higher ATPase flux and [Ca2+], must exist to explain the well-known difference in glycolytic rates in fast-twitch and slow-twitch muscles in actively contracting muscle. link: http://identifiers.org/pubmed/20308252

Vinnakotta2010_TranscientAnoxia_SOLmuscle: MODEL1006230112v0.0.1

This a model from the article: Common phenotype of resting mouse extensor digitorum longus and soleus muscles: equal A…

Details

Rates of ATPase and glycolysis are several times faster in actively contracting mouse extensor digitorum longus muscle (EDL) than soleus (SOL), but we find these rates are not distinguishable at rest. We used a transient anoxic perturbation of steady state energy balance to decrease phosphocreatine (PCr) reversibly and to measure the rates of ATPase and of lactate production without muscle activation or contraction. The rate of glycolytic ATP synthesis is less than the ATPase rate, accounting for the continual PCr decrease during anoxia in both muscles. We fitted a mathematical model validated with properties of enzymes and solutes measured in vitro and appropriate for the transient perturbation of these muscles to experimental data to test whether the model accounts for the results. Simulations showed equal rates of ATPase and lactate production in both muscles. ATPase controls glycolytic flux by feedback from its products. Adenylate kinase function is critical because a rise in [AMP] is necessary to activate glycogen phosphorylase. ATPase is the primary source of H+ production. The sum of contributions of the 13 reactions of the glycogenolytic and glycolytic network to total proton load is negligible. The stoichiometry of lactate and H+ production is near unity. These results identify a default state of energy metabolism for resting muscle in which there is no difference in the metabolic phenotype of EDL and SOL. Therefore, additional control mechanisms, involving higher ATPase flux and [Ca2+], must exist to explain the well-known difference in glycolytic rates in fast-twitch and slow-twitch muscles in actively contracting muscle. link: http://identifiers.org/pubmed/20308252

Vinod2011_MitoticExit: BIOMD0000000370v0.0.1

This model is from the article: Computational modelling of mitotic exit in budding yeast: the role of separase and Cdc…

Details

The operating principles of complex regulatory networks are best understood with the help of mathematical modelling rather than by intuitive reasoning. Hereby, we study the dynamics of the mitotic exit (ME) control system in budding yeast by further developing the Queralt's model. A comprehensive systems view of the network regulating ME is provided based on classical experiments in the literature. In this picture, Cdc20-APC is a critical node controlling both cyclin (Clb2 and Clb5) and phosphatase (Cdc14) branches of the regulatory network. On the basis of experimental situations ranging from single to quintuple mutants, the kinetic parameters of the network are estimated. Numerical analysis of the model quantifies the dependence of ME control on the proteolytic and non-proteolytic functions of separase. We show that the requirement of the non-proteolytic function of separase for ME depends on cyclin-dependent kinase activity. The model is also used for the systematic analysis of the recently discovered Cdc14 endocycles. The significance of Cdc14 endocycles in eukaryotic cell cycle control is discussed as well. link: http://identifiers.org/pubmed/21288956

Parameters:

NameDescription
kssic_1 = 0.2; Vdsic_1 = NaN; kssic_2 = 0.004Reaction: Sic1T_1 = (kssic_2+kssic_1*Swi5_1)-Vdsic_1*Sic1T_1, Rate Law: (kssic_2+kssic_1*Swi5_1)-Vdsic_1*Sic1T_1
Clb2nd_1 = 0.0Reaction: Clb2_2 = (Clb2T_1+Clb2nd_1)-Trim2_1, Rate Law: missing
Vd_1 = NaN; Vp_1 = NaN; Net1T_1 = 1.0Reaction: Net1dep_1 = Vd_1*(Net1T_1-Net1dep_1)-Vp_1*Net1dep_1, Rate Law: Vd_1*(Net1T_1-Net1dep_1)-Vp_1*Net1dep_1
Viswi_1 = NaN; Vaswi_1 = NaN; Swi5T_1 = 1.0; Jswi_1 = 0.1Reaction: Swi5_1 = Vaswi_1*(Swi5T_1-Swi5_1)/((Jswi_1+Swi5T_1)-Swi5_1)-Viswi_1*Swi5_1/(Jswi_1+Swi5_1), Rate Law: Vaswi_1*(Swi5T_1-Swi5_1)/((Jswi_1+Swi5T_1)-Swi5_1)-Viswi_1*Swi5_1/(Jswi_1+Swi5_1)
kp_1 = 2.0; ldnet_1 = 1.0; Vd_1 = NaN; Vp_1 = NaN; lanet_1 = 500.0; Net1T_1 = 1.0Reaction: RENTp_1 = (((Vp_1*(RENT_1-RENTp_1)-Vd_1*RENTp_1)+lanet_1*(((Net1T_1-Net1dep_1)-Net1pp_1)-RENTp_1)*Cdc14n_1)-ldnet_1*RENTp_1)-kp_1*Polo_1*RENTp_1, Rate Law: (((Vp_1*(RENT_1-RENTp_1)-Vd_1*RENTp_1)+lanet_1*(((Net1T_1-Net1dep_1)-Net1pp_1)-RENTp_1)*Cdc14n_1)-ldnet_1*RENTp_1)-kp_1*Polo_1*RENTp_1
kimbf_1 = 0.5; Jmbf_1 = 0.01; kambf_1 = 0.1; kimbf_3 = 0.5Reaction: MBF_1 = kambf_1*(1-MBF_1)/((Jmbf_1+1)-MBF_1)-(kimbf_1*Clb2_2+kimbf_3*Clb5_1)*MBF_1/(Jmbf_1+MBF_1), Rate Law: kambf_1*(1-MBF_1)/((Jmbf_1+1)-MBF_1)-(kimbf_1*Clb2_2+kimbf_3*Clb5_1)*MBF_1/(Jmbf_1+MBF_1)
kdsic2_1 = 0.1; V2_1 = NaN; Vdsic_1 = NaN; kasic2_1 = 40.0Reaction: Trim2_1 = kasic2_1*Clb2_2*Sic1_1-(kdsic2_1+V2_1+Vdsic_1)*Trim2_1, Rate Law: kasic2_1*Clb2_2*Sic1_1-(kdsic2_1+V2_1+Vdsic_1)*Trim2_1
ksclb5_1 = 0.01; V6_1 = NaN; ksclb5_2 = 0.002Reaction: Clb5T_1 = (ksclb5_2+ksclb5_1*MBF_1)-V6_1*Clb5T_1, Rate Law: (ksclb5_2+ksclb5_1*MBF_1)-V6_1*Clb5T_1
katem_2 = 0.6; kitem_3 = 0.1; kitem_1 = 20.0; kitem_2 = 1.0; Jtem1_1 = 0.005; katem_1 = 0.0Reaction: Tem1_1 = (katem_1+katem_2*Polo_1)*(1-Tem1_1)/((Jtem1_1+1)-Tem1_1)-(kitem_3+kitem_2/(1+kitem_1*Esp1_1))/(Jtem1_1+Tem1_1)*Tem1_1, Rate Law: (katem_1+katem_2*Polo_1)*(1-Tem1_1)/((Jtem1_1+1)-Tem1_1)-(kitem_3+kitem_2/(1+kitem_1*Esp1_1))/(Jtem1_1+Tem1_1)*Tem1_1
ksclb2_1 = 0.015; V2_1 = NaN; ksclb2_2 = 0.005Reaction: Clb2T_1 = (ksclb2_1+ksclb2_2*Mcm_1)-V2_1*Clb2T_1, Rate Law: (ksclb2_1+ksclb2_2*Mcm_1)-V2_1*Clb2T_1
kd20_1 = 0.1; ks20_2 = 0.001; ks20_1 = 0.05; kd20_2 = 1.0Reaction: Cdc20_1 = (ks20_2+ks20_1*Mcm_1)-(kd20_1+kd20_2*Cdh1_1)*Cdc20_1, Rate Law: (ks20_2+ks20_1*Mcm_1)-(kd20_1+kd20_2*Cdh1_1)*Cdc20_1
kapolo_1 = 0.0; kipolo_1 = 0.1; kapolo_2 = 1.0; kdpolo_1 = 0.05; kdpolo_2 = 0.5; Jpolo_1 = 0.1Reaction: Polo_1 = ((kapolo_1+kapolo_2*Clb2_2)*(PoloT_1-Polo_1)/((Jpolo_1+PoloT_1)-Polo_1)-kipolo_1*Polo_1/(Jpolo_1+Polo_1))-(kdpolo_1+kdpolo_2*Cdh1_1)*Polo_1, Rate Law: ((kapolo_1+kapolo_2*Clb2_2)*(PoloT_1-Polo_1)/((Jpolo_1+PoloT_1)-Polo_1)-kipolo_1*Polo_1/(Jpolo_1+Polo_1))-(kdpolo_1+kdpolo_2*Cdh1_1)*Polo_1
Jcdh_1 = 0.01; Vicdh_1 = NaN; Vacdh_1 = NaNReaction: Cdh1_1 = Vacdh_1*(1-Cdh1_1)/((Jcdh_1+1)-Cdh1_1)-Vicdh_1*Cdh1_1/(Jcdh_1+Cdh1_1), Rate Law: Vacdh_1*(1-Cdh1_1)/((Jcdh_1+1)-Cdh1_1)-Vicdh_1*Cdh1_1/(Jcdh_1+Cdh1_1)
kp_1 = 2.0; ldnet_1 = 1.0; lanet_1 = 500.0; kimp_1 = 1.0; Vexp_1 = NaN; Net1T_1 = 1.0Reaction: Cdc14n_1 = (((kp_1*Polo_1*RENTp_1-lanet_1*((Net1T_1-Net1pp_1)-RENT_1)*Cdc14n_1)+ldnet_1*RENT_1)-Vexp_1*Cdc14n_1)+kimp_1*Cdc14c_1, Rate Law: (((kp_1*Polo_1*RENTp_1-lanet_1*((Net1T_1-Net1pp_1)-RENT_1)*Cdc14n_1)+ldnet_1*RENT_1)-Vexp_1*Cdc14n_1)+kimp_1*Cdc14c_1
kdcln_1 = 0.25; kscln_1 = 0.1; kscln_2 = 0.01Reaction: Cln_1 = (kscln_2+kscln_1*MBF_1)-kdcln_1*Cln_1, Rate Law: (kscln_2+kscln_1*MBF_1)-kdcln_1*Cln_1
Net1T_1 = 1.0Reaction: Net1_2 = ((Net1T_1-Net1p_1)-RENT_1)-Net1pp_1, Rate Law: missing
ksmcm_1 = 1.0; Jmcm_1 = 0.01; kdmcm_1 = 0.25; ksmcm_3 = 0.01Reaction: Mcm_1 = (ksmcm_3+ksmcm_1*Clb2_2)*(1-Mcm_1)/((Jmcm_1+1)-Mcm_1)-kdmcm_1*Mcm_1/(Jmcm_1+Mcm_1), Rate Law: (ksmcm_3+ksmcm_1*Clb2_2)*(1-Mcm_1)/((Jmcm_1+1)-Mcm_1)-kdmcm_1*Mcm_1/(Jmcm_1+Mcm_1)
Jcdc15_1 = 1.0; ldmen_1 = 0.1; kitem_3 = 0.1; kic15_2 = 0.2; kitem_2 = 1.0; lamen_1 = 100.0; kic15_1 = 0.03; Jtem1_1 = 0.005Reaction: MEN_1 = ((lamen_1*(Tem1_1-MEN_1)*(Cdc15_1-MEN_1)-ldmen_1*MEN_1)-(kitem_3+kitem_2/(1+kitem_3*Esp1_1))/(Jtem1_1+Tem1_1)*MEN_1)-(kic15_1+kic15_2*Clb2_2)/(Jcdc15_1+Cdc15_1)*MEN_1, Rate Law: ((lamen_1*(Tem1_1-MEN_1)*(Cdc15_1-MEN_1)-ldmen_1*MEN_1)-(kitem_3+kitem_2/(1+kitem_3*Esp1_1))/(Jtem1_1+Tem1_1)*MEN_1)-(kic15_1+kic15_2*Clb2_2)/(Jcdc15_1+Cdc15_1)*MEN_1
lapds_1 = 500.0; ldpds_1 = 1.0; kdesp_1 = 0.004; kdpds_2 = 2.0; kdpds_1 = 0.01Reaction: Esp1b_1 = lapds_1*Pds1_1*Esp1_1-(ldpds_1+kdesp_1+kdpds_1+kdpds_2*Cdc20_1)*Esp1b_1, Rate Law: lapds_1*Pds1_1*Esp1_1-(ldpds_1+kdesp_1+kdpds_1+kdpds_2*Cdc20_1)*Esp1b_1
kp_1 = 2.0; Vd_1 = NaN; Net1T_1 = 1.0Reaction: Net1pp_1 = kp_1*Polo_1*((Net1T_1-Net1dep_1)-Net1pp_1)-Vd_1*Net1pp_1, Rate Law: kp_1*Polo_1*((Net1T_1-Net1dep_1)-Net1pp_1)-Vd_1*Net1pp_1
kp_1 = 2.0; ldnet_1 = 1.0; lanet_1 = 500.0; Net1T_1 = 1.0Reaction: RENT_1 = (lanet_1*((Net1T_1-Net1pp_1)-RENT_1)*Cdc14n_1-ldnet_1*RENT_1)-kp_1*Polo_1*RENTp_1, Rate Law: (lanet_1*((Net1T_1-Net1pp_1)-RENT_1)*Cdc14n_1-ldnet_1*RENT_1)-kp_1*Polo_1*RENTp_1
kasic5_1 = 10.0; kdsic5_1 = 0.1; V6_1 = NaN; Vdsic_1 = NaNReaction: Trim5_1 = kasic5_1*Clb5_1*Sic1_1-(kdsic5_1+V6_1+Vdsic_1)*Trim5_1, Rate Law: kasic5_1*Clb5_1*Sic1_1-(kdsic5_1+V6_1+Vdsic_1)*Trim5_1
kdesp_1 = 0.004; ksesp_1 = 0.001Reaction: Esp1T_1 = ksesp_1-kdesp_1*Esp1T_1, Rate Law: ksesp_1-kdesp_1*Esp1T_1
kspds_1 = 0.01; kdpds_2 = 2.0; kspds_2 = 0.006; kdpds_1 = 0.01Reaction: Pds1T_1 = (kspds_2+kspds_1*MBF_1)-(kdpds_1+kdpds_2*Cdc20_1)*Pds1T_1, Rate Law: (kspds_2+kspds_1*MBF_1)-(kdpds_1+kdpds_2*Cdc20_1)*Pds1T_1
Cdc14T_1 = 0.5Reaction: Cdc14c_1 = (Cdc14T_1-Cdc14n_1)-RENT_1, Rate Law: missing
Jcdc15_1 = 1.0; kac15_2 = 0.5; kic15_2 = 0.2; kac15_1 = 0.03; kic15_1 = 0.03Reaction: Cdc15_1 = (kac15_1+kac15_2*Cdc14c_1)*(1-Cdc15_1)/((Jcdc15_1+1)-Cdc15_1)-(kic15_1+kic15_2*Clb2_2)*Cdc15_1/(Jcdc15_1+Cdc15_1), Rate Law: (kac15_1+kac15_2*Cdc14c_1)*(1-Cdc15_1)/((Jcdc15_1+1)-Cdc15_1)-(kic15_1+kic15_2*Clb2_2)*Cdc15_1/(Jcdc15_1+Cdc15_1)
kspolo_1 = 0.05; kdpolo_1 = 0.05; kdpolo_2 = 0.5; kspolo_2 = 0.001Reaction: PoloT_1 = (kspolo_2+kspolo_1*Mcm_1)-(kdpolo_1+kdpolo_2*Cdh1_1)*PoloT_1, Rate Law: (kspolo_2+kspolo_1*Mcm_1)-(kdpolo_1+kdpolo_2*Cdh1_1)*PoloT_1

States:

NameDescription
Pds1 1[Securin]
Sic1 1[Protein SIC1]
Esp1T 1[Separin]
Cdc15 1[Cell division control protein 15]
Esp1 1[Separin]
Cdh1 1[APC/C activator protein CDH1]
MEN 1[Cell division control protein 15; Protein TEM1]
Trim5 1[Cyclin-dependent kinase 1; S-phase entry cyclin-5; Protein SIC1]
Esp1b 1[Separin]
Net1dep 1[Nucleolar protein NET1]
Cdc14n 1[Tyrosine-protein phosphatase CDC14]
Tem1 1[Protein TEM1]
Pds1T 1[Securin]
PoloT 1[Cell cycle serine/threonine-protein kinase CDC5/MSD2]
Net1pp 1[Nucleolar protein NET1; Phosphoprotein]
MBF 1[Multiprotein-bridging factor 1]
Swi5 1[Transcriptional factor SWI5]
RENTp 1[Phosphoprotein; Tyrosine-protein phosphatase CDC14; Nucleolar protein NET1]
Cdc14c 1[Tyrosine-protein phosphatase CDC14]
Clb5T 1[S-phase entry cyclin-5]
Sic1T 1[Protein SIC1]
Trim2 1[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin-2; Protein SIC1]
Clb2 2[G2/mitotic-specific cyclin-2]
Polo 1[Cell cycle serine/threonine-protein kinase CDC5/MSD2]
Cln 1[G1/S-specific cyclin CLN2]
Net1 2[Nucleolar protein NET1]
Cdc20 1[APC/C activator protein CDC20]
Mcm 1[Nuclear division defective protein 1; Fork head protein homolog 2; Pheromone receptor transcription factor]
Clb2T 1[G2/mitotic-specific cyclin-2]
Clb5 1[S-phase entry cyclin-5]
RENT 1[Tyrosine-protein phosphatase CDC14; Nucleolar protein NET1]

Voit2003 - Trehalose Cycle: BIOMD0000000266v0.0.1

Voit2003 - Trehalose CycleThis model is described in the article: [Biochemical and genomic regulation of the trehalose…

Details

The physiological hallmark of heat-shock response in yeast is a rapid, enormous increase in the concentration of trehalose. Normally found in growing yeast cells and other organisms only as traces, trehalose becomes a crucial protector of proteins and membranes against a variety of stresses, including heat, cold, starvation, desiccation, osmotic or oxidative stress, and exposure to toxicants. Trehalose is produced from glucose 6-phosphate and uridine diphosphate glucose in a two-step process, and recycled to glucose by trehalases. Even though the trehalose cycle consists of only a few metabolites and enzymatic steps, its regulatory structure and operation are surprisingly complex. The article begins with a review of experimental observations on the regulation of the trehalose cycle in yeast and proposes a canonical model for its analysis. The first part of this analysis demonstrates the benefits of the various regulatory features by means of controlled comparisons with models of otherwise equivalent pathways lacking these features. The second part elucidates the significance of the expression pattern of the trehalose cycle genes in response to heat shock. Interestingly, the genes contributing to trehalose formation are up-regulated to very different degrees, and even the trehalose degrading trehalases show drastically increased activity during heat-shock response. Again using the method of controlled comparisons, the model provides rationale for the observed pattern of gene expression and reveals benefits of the counterintuitive trehalase up-regulation. link: http://identifiers.org/pubmed/12782117

Parameters:

NameDescription
flux_X1_in = NaN mM per minute; flux_X1_out = NaN mM per minuteReaction: X1 = flux_X1_in-flux_X1_out, Rate Law: flux_X1_in-flux_X1_out
flux_X5_out = NaN mM per minute; flux_X5_in = NaN mM per minuteReaction: X5 = flux_X5_in-flux_X5_out, Rate Law: flux_X5_in-flux_X5_out
flux_X3_in = NaN mM per minute; flux_X3_out = NaN mM per minuteReaction: X3 = flux_X3_in-flux_X3_out, Rate Law: flux_X3_in-flux_X3_out
flux_X6_in = NaN mM per minute; flux_X6_out = NaN mM per minuteReaction: X6 = flux_X6_in-flux_X6_out, Rate Law: flux_X6_in-flux_X6_out
flux_X4_out = NaN mM per minute; flux_X4_in = NaN mM per minuteReaction: X4 = flux_X4_in-flux_X4_out, Rate Law: flux_X4_in-flux_X4_out
flux_X2_in = NaN mM per minute; flux_X2_out = NaN mM per minuteReaction: X2 = flux_X2_in-flux_X2_out, Rate Law: flux_X2_in-flux_X2_out
flux_X7_in = NaN mM per minute; flux_X7_out = NaN mM per minuteReaction: X7 = flux_X7_in-flux_X7_out, Rate Law: flux_X7_in-flux_X7_out

States:

NameDescription
X3[D-glucopyranose 1-phosphate]
X7[alpha,alpha-trehalose]
X4[UDP-D-glucose]
X5[glycogen]
X2[alpha-D-glucose 6-phosphate]
X1[alpha-D-glucose]
X6[alpha,alpha-trehalose 6-phosphate]

Voliotis2019-GnRH Pulse Generation: BIOMD0000000931v0.0.1

Fertility critically depends on the gonadotropin-releasing hormone (GnRH) pulse generator, a neural construct comprised…

Details

Fertility critically depends on the gonadotropin-releasing hormone (GnRH) pulse generator, a neural construct comprised of hypothalamic neurons coexpressing kisspeptin, neurokoinin-B and dynorphin. Here, using mathematical modeling and in vivo optogenetics we reveal for the first time how this neural construct initiates and sustains the appropriate ultradian frequency essential for reproduction. Prompted by mathematical modeling, we show experimentally using female estrous mice that robust pulsatile release of luteinizing hormone, a proxy for GnRH, emerges abruptly as we increase the basal activity of the neuronal network using continuous low-frequency optogenetic stimulation. Further increase in basal activity markedly increases pulse frequency and eventually leads to pulse termination. Additional model predictions that pulsatile dynamics emerge from nonlinear positive and negative feedback interactions mediated through neurokinin-B and dynorphin signaling respectively are confirmed neuropharmacologically. Our results shed light on the long-elusive GnRH pulse generator offering new horizons for reproductive health and wellbeing.SIGNIFICANCE STATEMENT The gonadotropin-releasing hormone (GnRH) pulse generator controls the pulsatile secretion of the gonadotropic hormones LH and FSH and is critical for fertility. The hypothalamic arcuate kisspeptin neurons are thought to represent the GnRH pulse generator, since their oscillatory activity is coincident with LH pulses in the blood; a proxy for GnRH pulses. However, the mechanisms underlying GnRH pulse generation remain elusive. We developed a mathematical model of the kisspeptin neuronal network and confirmed its predictions experimentally, showing how LH secretion is frequency-modulated as we increase the basal activity of the arcuate kisspeptin neurons in vivo using continuous optogenetic stimulation. Our model provides a quantitative framework for understanding the reproductive neuroendocrine system and opens new horizons for fertility regulation. link: http://identifiers.org/pubmed/31645462

Parameters:

NameDescription
d_N = 1.0Reaction: N =>, Rate Law: compartment*d_N*N
f_N = 0.302353418222975Reaction: => N, Rate Law: compartment*f_N
f_v = 3016.26432543932Reaction: => v, Rate Law: compartment*f_v
d_D = 0.25Reaction: D =>, Rate Law: compartment*d_D*D
f_D = 0.439705882352941Reaction: => D, Rate Law: compartment*f_D
d_v = 10.0Reaction: v =>, Rate Law: compartment*d_v*v

States:

NameDescription
vv
NN
DD

Vongsangnak2008 - Genome-scale metabolic network of Aspergillus oryzae (iWV1314): MODEL1507180056v0.0.1

Vongsangnak2008 - Genome-scale metabolic network of Aspergillus oryzae (iWV1314)This model is described in the article:…

Details

BACKGROUND: Since ancient times the filamentous fungus Aspergillus oryzae has been used in the fermentation industry for the production of fermented sauces and the production of industrial enzymes. Recently, the genome sequence of A. oryzae with 12,074 annotated genes was released but the number of hypothetical proteins accounted for more than 50% of the annotated genes. Considering the industrial importance of this fungus, it is therefore valuable to improve the annotation and further integrate genomic information with biochemical and physiological information available for this microorganism and other related fungi. Here we proposed the gene prediction by construction of an A. oryzae Expressed Sequence Tag (EST) library, sequencing and assembly. We enhanced the function assignment by our developed annotation strategy. The resulting better annotation was used to reconstruct the metabolic network leading to a genome scale metabolic model of A. oryzae. RESULTS: Our assembled EST sequences we identified 1,046 newly predicted genes in the A. oryzae genome. Furthermore, it was possible to assign putative protein functions to 398 of the newly predicted genes. Noteworthy, our annotation strategy resulted in assignment of new putative functions to 1,469 hypothetical proteins already present in the A. oryzae genome database. Using the substantially improved annotated genome we reconstructed the metabolic network of A. oryzae. This network contains 729 enzymes, 1,314 enzyme-encoding genes, 1,073 metabolites and 1,846 (1,053 unique) biochemical reactions. The metabolic reactions are compartmentalized into the cytosol, the mitochondria, the peroxisome and the extracellular space. Transport steps between the compartments and the extracellular space represent 281 reactions, of which 161 are unique. The metabolic model was validated and shown to correctly describe the phenotypic behavior of A. oryzae grown on different carbon sources. CONCLUSION: A much enhanced annotation of the A. oryzae genome was performed and a genome-scale metabolic model of A. oryzae was reconstructed. The model accurately predicted the growth and biomass yield on different carbon sources. The model serves as an important resource for gaining further insight into our understanding of A. oryzae physiology. link: http://identifiers.org/pubmed/18500999

Väremo2015 - Human myocyte metabolic model: MODEL1503240000v0.0.1

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

Details

Skeletal myocytes are metabolically active and susceptible to insulin resistance and are thus implicated in type 2 diabetes (T2D). This complex disease involves systemic metabolic changes, and their elucidation at the systems level requires genome-wide data and biological networks. Genome-scale metabolic models (GEMs) provide a network context for the integration of high-throughput data. We generated myocyte-specific RNA-sequencing data and investigated their correlation with proteome data. These data were then used to reconstruct a comprehensive myocyte GEM. Next, we performed a meta-analysis of six studies comparing muscle transcription in T2D versus healthy subjects. Transcriptional changes were mapped on the myocyte GEM, revealing extensive transcriptional regulation in T2D, particularly around pyruvate oxidation, branched-chain amino acid catabolism, and tetrahydrofolate metabolism, connected through the downregulated dihydrolipoamide dehydrogenase. Strikingly, the gene signature underlying this metabolic regulation successfully classifies the disease state of individual samples, suggesting that regulation of these pathways is a ubiquitous feature of myocytes in response to T2D. link: http://identifiers.org/pubmed/25937284

W


Wajima2009_BloodCoagulation_aPTTtest: BIOMD0000000338v0.0.1

This model is from the article: A comprehensive model for the humoral coagulation network in humans. Wajima T, Is…

Details

Coagulation is an important process in hemostasis and comprises a complicated interaction of multiple enzymes and proteins. We have developed a mechanistic quantitative model of the coagulation network. The model accurately describes the time courses of coagulation factors following in vivo activation as well as in vitro blood coagulation tests of prothrombin time (PT, often reported as international normalized ratio (INR)) and activated partial thromboplastin time (aPTT). The model predicts the concentration-time and time-effect profiles of warfarin, heparins, and vitamin K in humans. The model can be applied to predict the time courses of coagulation kinetics in clinical situations (e.g., hemophilia) and for biomarker identification during drug development. The model developed in this study is the first quantitative description of the comprehensive coagulation network. link: http://identifiers.org/pubmed/19516255

Parameters:

NameDescription
v=20000.0; k=0.5Reaction: Fg => F; IIa, Rate Law: compartment_1*v*Fg*IIa/(k+IIa)
VKH20 = 0.1; II0 = 1394.4; d_II = 0.01Reaction: => II; VKH2, Rate Law: compartment_1*d_II*II0*VKH2/VKH20
c=0.5Reaction: VIIa_TF + Xa_TFPI => VIIa_TF_Xa_TFPI, Rate Law: compartment_1*VIIa_TF*Xa_TFPI/c
k1=0.05Reaction: F => FDP, Rate Law: compartment_1*k1*F
v=50000.0; k=1.0E-6Reaction: VIII => VIIIa; IIa, Rate Law: compartment_1*v*VIII*IIa/(k+IIa)
k=1.0; v=1.0Reaction: XF => D; APC_PS, Rate Law: compartment_1*v*XF*APC_PS/(k+APC_PS)
v=7.0; k=10.0Reaction: F => XF; XIIIa, Rate Law: compartment_1*v*F*XIIIa/(k+XIIIa)
d_Pg = 0.05Reaction: Pg =>, Rate Law: compartment_1*d_Pg*Pg
PC0 = 60.0; VKH20 = 0.1; d_PC = 0.05Reaction: => PC; VKH2, Rate Law: compartment_1*d_PC*PC0*VKH2/VKH20
c46 = 0.85Reaction: IXa + ATIII_Heparin => IXa_ATIII_Heparin, Rate Law: compartment_1*IXa*ATIII_Heparin/c46
c45 = 0.85Reaction: Xa + ATIII_Heparin => Xa_ATIII_Heparin, Rate Law: compartment_1*Xa*ATIII_Heparin/c45
k=1.0; v=7.0Reaction: XIII => XIIIa; IIa, Rate Law: compartment_1*v*XIII*IIa/(k+IIa)
d_Tmod = 0.05; Tmod0 = 50.0Reaction: => Tmod, Rate Law: compartment_1*Tmod0*d_Tmod
k1=0.7Reaction: VII_TF =>, Rate Law: compartment_1*k1*VII_TF
Fg0 = 8945.5; d_Fg = 0.032Reaction: => Fg, Rate Law: compartment_1*Fg0*d_Fg
k1=20.0Reaction: Va =>, Rate Law: compartment_1*k1*Va
d_XIII = 0.0036Reaction: XIII =>, Rate Law: compartment_1*d_XIII*XIII
k=1.0; v=70.0Reaction: VII_TF => VIIa_TF; Xa, Rate Law: compartment_1*v*VII_TF*Xa/(k+Xa)
k1=0.69Reaction: XIIIa =>, Rate Law: compartment_1*k1*XIIIa
VitaminK_k12 = 0.0587; VitaminK_k21_Vc = 5.08333333333333E-4Reaction: VK => VK_p, Rate Law: compartment_1*(VitaminK_k12*VK-VitaminK_k21_Vc*VK_p)
v=10.0; k=10.0Reaction: XI => XIa; IIa, Rate Law: compartment_1*v*XI*IIa/(k+IIa)
k=500.0; v=9.0Reaction: II => IIa; Xa, Rate Law: compartment_1*v*II*Xa/(k+Xa)
k1=0.1Reaction: D =>, Rate Law: compartment_1*k1*D
v=100.0; k=10.0Reaction: II => IIa; Va_Xa, Rate Law: compartment_1*v*II*Va_Xa/(k+Va_Xa)
Pg0 = 2154.3; d_Pg = 0.05Reaction: => Pg, Rate Law: compartment_1*Pg0*d_Pg
d_PC = 0.05Reaction: PC =>, Rate Law: compartment_1*d_PC*PC
k=500.0; v=500.0Reaction: Fg => FDP; P, Rate Law: compartment_1*v*Fg*P/(k+P)
c=0.1Reaction: VII + TF => VII_TF, Rate Law: compartment_1*VII*TF/c
v=7.0; k=5000.0Reaction: Pg => P; IIa, Rate Law: compartment_1*v*Pg*IIa/(k+IIa)
v=7.0; k=100.0Reaction: XF => D; P, Rate Law: compartment_1*v*XF*P/(k+P)
k=1.0; v=50.0Reaction: VIIIa => ; APC_PS, Rate Law: compartment_1*v*VIIIa*APC_PS/(k+APC_PS)
c=0.01Reaction: IXa + VIIIa => IXa_VIIIa, Rate Law: compartment_1*IXa*VIIIa/c
k1=20.4Reaction: APC =>, Rate Law: compartment_1*k1*APC
Warfarin_ka = 1.0Reaction: A_warf = (-Warfarin_ka)*A_warf, Rate Law: (-Warfarin_ka)*A_warf
d_II = 0.01Reaction: II =>, Rate Law: compartment_1*d_II*II
d_VIII = 0.058; VIII0 = 0.7Reaction: => VIII, Rate Law: compartment_1*VIII0*d_VIII
k=1.0; v=1000.0Reaction: VII_TF => VIIa_TF; TF, Rate Law: compartment_1*v*VII_TF*TF/(k+TF)
k=1.0; v=2.0Reaction: Pg => P; APC_PS, Rate Law: compartment_1*v*Pg*APC_PS/(k+APC_PS)
c44 = 0.119718309859155Reaction: IIa + ATIII_Heparin => IIa_ATIII_Heparin, Rate Law: compartment_1*IIa*ATIII_Heparin/c44
d_Fg = 0.032Reaction: Fg => FDP, Rate Law: compartment_1*d_Fg*Fg
d_IX = 0.029; VKH20 = 0.1; IX0 = 89.6Reaction: => IX; VKH2, Rate Law: compartment_1*d_IX*IX0*VKH2/VKH20
k1=67.4Reaction: IIa => TAT, Rate Law: compartment_1*k1*IIa
Heparin_ke = 0.693Reaction: IIa_ATIII_Heparin =>, Rate Law: compartment_1*Heparin_ke*IIa_ATIII_Heparin
d_Tmod = 0.05Reaction: Tmod =>, Rate Law: compartment_1*d_Tmod*Tmod
d_XII = 0.012Reaction: XII =>, Rate Law: compartment_1*d_XII*XII
d_IX = 0.029Reaction: IX =>, Rate Law: compartment_1*d_IX*IX
k1=0.2Reaction: TAT =>, Rate Law: compartment_1*k1*TAT
k=10000.0; v=5.0Reaction: Pg => P; F, Rate Law: compartment_1*v*Pg*F/(k+F)
XIII0 = 70.3; d_XIII = 0.0036Reaction: => XIII, Rate Law: compartment_1*XIII0*d_XIII

States:

NameDescription
II[Prothrombin]
VIIa TF Xa TFPI[Tissue factor; Coagulation factor X; Coagulation factor VII; Tissue factor pathway inhibitor]
VIII[Coagulation factor VIII]
P[Plasminogen]
TFPI[Tissue factor pathway inhibitor]
A warf[warfarin]
XIII[Coagulation factor XIII A chain; Coagulation factor XIII B chain]
APC PS[Vitamin K-dependent protein C; Vitamin K-dependent protein S]
DPDP
IIa Tmod[Thrombomodulin; Prothrombin]
Xa ATIII Heparin[heparin; Coagulation factor X; Antithrombin-III]
PC[Vitamin K-dependent protein C]
XF[Fibrinogen alpha chain; Fibrinogen gamma chain; Fibrinogen beta chain]
IIa ATIII Heparin[heparin; Antithrombin-III; Prothrombin]
TF[Tissue factor]
VII TF[Tissue factor; Coagulation factor VII]
ATIII Heparin[heparin; Antithrombin-III]
XII[Coagulation factor XII]
XIa[Coagulation factor XI]
Fg[Fibrinogen alpha chain; Fibrinogen gamma chain; Fibrinogen beta chain]
Tmod[Thrombomodulin]
VK p[vitamin K]
DD
VIIIa[Coagulation factor VIII]
Va[Coagulation factor V]
IIa[Prothrombin]
Xa TFPI[Coagulation factor X; Tissue factor pathway inhibitor]
XIIIa[Coagulation factor XIII A chain]
APC[Vitamin K-dependent protein C]
Pg[Plasminogen]
IXa[Coagulation factor IX]
TAT[Prothrombin; Antithrombin-III]
IXa ATIII Heparin[heparin; Coagulation factor IX; Antithrombin-III]
VIIa TF[Tissue factor; Coagulation factor VII]
K[Plasma kallikrein]
F[Fibrinogen alpha chain; Fibrinogen gamma chain; Fibrinogen beta chain]
IX[Coagulation factor IX]

Wajima2009_BloodCoagulation_PTtest: BIOMD0000000339v0.0.1

This model is from the article: A comprehensive model for the humoral coagulation network in humans. Wajima T, Is…

Details

Coagulation is an important process in hemostasis and comprises a complicated interaction of multiple enzymes and proteins. We have developed a mechanistic quantitative model of the coagulation network. The model accurately describes the time courses of coagulation factors following in vivo activation as well as in vitro blood coagulation tests of prothrombin time (PT, often reported as international normalized ratio (INR)) and activated partial thromboplastin time (aPTT). The model predicts the concentration-time and time-effect profiles of warfarin, heparins, and vitamin K in humans. The model can be applied to predict the time courses of coagulation kinetics in clinical situations (e.g., hemophilia) and for biomarker identification during drug development. The model developed in this study is the first quantitative description of the comprehensive coagulation network. link: http://identifiers.org/pubmed/19516255

Parameters:

NameDescription
c=0.5Reaction: VIIa_TF + Xa_TFPI => VIIa_TF_Xa_TFPI, Rate Law: compartment_1*VIIa_TF*Xa_TFPI/c
k1=0.05Reaction: F => FDP, Rate Law: compartment_1*k1*F
v=1.0E-9; k=10.0Reaction: X => Xa; VIIa, Rate Law: compartment_1*v*X*VIIa/(k+VIIa)
v=50000.0; k=1.0E-6Reaction: VIII => VIIIa; IIa, Rate Law: compartment_1*v*VIII*IIa/(k+IIa)
k=1.0; v=1.0Reaction: XF => D; APC_PS, Rate Law: compartment_1*v*XF*APC_PS/(k+APC_PS)
v=7.0; k=10.0Reaction: F => FDP; P, Rate Law: compartment_1*v*F*P/(k+P)
d_Pg = 0.05Reaction: Pg =>, Rate Law: compartment_1*d_Pg*Pg
PC0 = 60.0; VKH20 = 0.1; d_PC = 0.05Reaction: => PC; VKH2, Rate Law: compartment_1*d_PC*PC0*VKH2/VKH20
c46 = 0.85Reaction: IXa + ATIII_Heparin => IXa_ATIII_Heparin, Rate Law: compartment_1*IXa*ATIII_Heparin/c46
v=0.1; k=10.0Reaction: VII => VIIa; IIa, Rate Law: compartment_1*v*VII*IIa/(k+IIa)
k=1.0; v=7.0Reaction: XI => XIa; XIIa, Rate Law: compartment_1*v*XI*XIIa/(k+XIIa)
v=1.0; k=10.0Reaction: VII => VIIa; Xa, Rate Law: compartment_1*v*VII*Xa/(k+Xa)
v=0.2; k=10.0Reaction: VII => VIIa; IXa, Rate Law: compartment_1*v*VII*IXa/(k+IXa)
d_Tmod = 0.05; Tmod0 = 50.0Reaction: => Tmod, Rate Law: compartment_1*Tmod0*d_Tmod
d_VIII = 0.058Reaction: VIII =>, Rate Law: compartment_1*d_VIII*VIII
Fg0 = 8945.5; d_Fg = 0.032Reaction: => Fg, Rate Law: compartment_1*Fg0*d_Fg
k1=20.0Reaction: VIIIa =>, Rate Law: compartment_1*k1*VIIIa
d_XIII = 0.0036Reaction: XIII =>, Rate Law: compartment_1*d_XIII*XIII
k=1.0; v=70.0Reaction: IX => IXa; VIIa_TF, Rate Law: compartment_1*v*IX*VIIa_TF/(k+VIIa_TF)
k=0.1; v=2.0Reaction: X => Xa; IXa_VIIIa, Rate Law: compartment_1*v*X*IXa_VIIIa/(k+IXa_VIIIa)
v=10.0; k=10.0Reaction: XI => XIa; IIa, Rate Law: compartment_1*v*XI*IIa/(k+IIa)
k1=0.1Reaction: D =>, Rate Law: compartment_1*k1*D
k=500.0; v=9.0Reaction: II => IIa; Xa, Rate Law: compartment_1*v*II*Xa/(k+Xa)
v=100.0; k=10.0Reaction: II => IIa; Va_Xa, Rate Law: compartment_1*v*II*Va_Xa/(k+Va_Xa)
Pg0 = 2154.3; d_Pg = 0.05Reaction: => Pg, Rate Law: compartment_1*Pg0*d_Pg
v=0.02; k=10.0Reaction: X => Xa; IXa, Rate Law: compartment_1*v*X*IXa/(k+IXa)
d_PC = 0.05Reaction: PC =>, Rate Law: compartment_1*d_PC*PC
k=500.0; v=500.0Reaction: Fg => FDP; P, Rate Law: compartment_1*v*Fg*P/(k+P)
c=0.1Reaction: VII + TF => VII_TF, Rate Law: compartment_1*VII*TF/c
v=7.0; k=5000.0Reaction: Pg => P; IIa, Rate Law: compartment_1*v*Pg*IIa/(k+IIa)
v=7.0; k=100.0Reaction: XF => D; P, Rate Law: compartment_1*v*XF*P/(k+P)
k=1.0; v=50.0Reaction: VIIIa => ; APC_PS, Rate Law: compartment_1*v*VIIIa*APC_PS/(k+APC_PS)
c=0.01Reaction: IXa + VIIIa => IXa_VIIIa, Rate Law: compartment_1*IXa*VIIIa/c
k1=20.4Reaction: APC =>, Rate Law: compartment_1*k1*APC
Warfarin_ka = 1.0Reaction: A_warf = (-Warfarin_ka)*A_warf, Rate Law: (-Warfarin_ka)*A_warf
d_VIII = 0.058; VIII0 = 0.7Reaction: => VIII, Rate Law: compartment_1*VIII0*d_VIII
c44 = 0.119718309859155Reaction: IIa + ATIII_Heparin => IIa_ATIII_Heparin, Rate Law: compartment_1*IIa*ATIII_Heparin/c44
k=1.0; v=2.0Reaction: Pg => P; APC_PS, Rate Law: compartment_1*v*Pg*APC_PS/(k+APC_PS)
d_Fg = 0.032Reaction: Fg => FDP, Rate Law: compartment_1*d_Fg*Fg
d_IX = 0.029; VKH20 = 0.1; IX0 = 89.6Reaction: => IX; VKH2, Rate Law: compartment_1*d_IX*IX0*VKH2/VKH20
d_XI = 0.1; XI0 = 30.6Reaction: => XI, Rate Law: compartment_1*XI0*d_XI
d_VII = 0.12Reaction: VII =>, Rate Law: compartment_1*d_VII*VII
k1=67.4Reaction: IIa => TAT, Rate Law: compartment_1*k1*IIa
d_Tmod = 0.05Reaction: Tmod =>, Rate Law: compartment_1*d_Tmod*Tmod
v=900.0; k=200.0Reaction: X => Xa; VIIa_TF, Rate Law: compartment_1*v*X*VIIa_TF/(k+VIIa_TF)
VII0 = 10.0; VKH20 = 0.1; d_VII = 0.12Reaction: => VII; VKH2, Rate Law: compartment_1*d_VII*VII0*VKH2/VKH20
d_X = 0.018; VKH20 = 0.1; X0 = 174.3Reaction: => X; VKH2, Rate Law: compartment_1*d_X*X0*VKH2/VKH20
d_X = 0.018Reaction: X =>, Rate Law: compartment_1*d_X*X
d_IX = 0.029Reaction: IX =>, Rate Law: compartment_1*d_IX*IX
k1=0.2Reaction: TAT =>, Rate Law: compartment_1*k1*TAT
k=10000.0; v=5.0Reaction: Pg => P; F, Rate Law: compartment_1*v*Pg*F/(k+F)
XIII0 = 70.3; d_XIII = 0.0036Reaction: => XIII, Rate Law: compartment_1*XIII0*d_XIII

States:

NameDescription
VIIa TF Xa TFPI[Tissue factor; Coagulation factor X; Coagulation factor VII; Tissue factor pathway inhibitor]
VIII[Coagulation factor VIII]
P[Plasminogen]
A warf[warfarin]
APC PS[Vitamin K-dependent protein C; Vitamin K-dependent protein S]
XIII[Coagulation factor XIII A chain; Coagulation factor XIII B chain]
Xa[Coagulation factor X]
FDP[Fibrinogen alpha chain; Fibrinogen gamma chain; Fibrinogen beta chain]
DPDP
IIa Tmod[Thrombomodulin; Prothrombin]
PC[Vitamin K-dependent protein C]
XF[Fibrinogen alpha chain; Fibrinogen gamma chain; Fibrinogen beta chain]
IIa ATIII Heparin[heparin; Antithrombin-III; Prothrombin]
TF[Tissue factor]
XIa[Coagulation factor XI]
Fg[Fibrinogen alpha chain; Fibrinogen gamma chain; Fibrinogen beta chain]
Tmod[Thrombomodulin]
X[Coagulation factor X]
DD
VIIIa[Coagulation factor VIII]
IIa[Prothrombin]
VIIa[Coagulation factor VII]
XI[Coagulation factor XI]
XIIa[Coagulation factor XII]
APC[Vitamin K-dependent protein C]
TAT[Prothrombin; Antithrombin-III]
IXa[Coagulation factor IX]
Pg[Plasminogen]
IXa ATIII Heparin[heparin; Coagulation factor IX; Antithrombin-III]
VII[Coagulation factor VII]
IX[Coagulation factor IX]

Wajima2009_BloodCoagulation_warfarin_heparin: BIOMD0000000340v0.0.1

This model is from the article: A comprehensive model for the humoral coagulation network in humans. Wajima T, Is…

Details

Coagulation is an important process in hemostasis and comprises a complicated interaction of multiple enzymes and proteins. We have developed a mechanistic quantitative model of the coagulation network. The model accurately describes the time courses of coagulation factors following in vivo activation as well as in vitro blood coagulation tests of prothrombin time (PT, often reported as international normalized ratio (INR)) and activated partial thromboplastin time (aPTT). The model predicts the concentration-time and time-effect profiles of warfarin, heparins, and vitamin K in humans. The model can be applied to predict the time courses of coagulation kinetics in clinical situations (e.g., hemophilia) and for biomarker identification during drug development. The model developed in this study is the first quantitative description of the comprehensive coagulation network. link: http://identifiers.org/pubmed/19516255

Parameters:

NameDescription
v=20000.0; k=0.5Reaction: Fg => F; IIa, Rate Law: compartment_1*v*Fg*IIa/(k+IIa)
VKH20 = 0.1; II0 = 1394.4; d_II = 0.01Reaction: => II; VKH2, Rate Law: compartment_1*d_II*II0*VKH2/VKH20
c=0.5Reaction: VIIa_TF + Xa_TFPI => VIIa_TF_Xa_TFPI, Rate Law: compartment_1*VIIa_TF*Xa_TFPI/c
k1=0.05Reaction: F => FDP, Rate Law: compartment_1*k1*F
v=50000.0; k=10.0Reaction: V => Va; IIa, Rate Law: compartment_1*v*V*IIa/(k+IIa)
v=1.0E-9; k=10.0Reaction: X => Xa; VIIa, Rate Law: compartment_1*v*X*VIIa/(k+VIIa)
v=50000.0; k=1.0E-6Reaction: VIII => VIIIa; IIa, Rate Law: compartment_1*v*VIII*IIa/(k+IIa)
v=7.0; k=10.0Reaction: IX => IXa; XIa, Rate Law: compartment_1*v*IX*XIa/(k+XIa)
c46 = 0.85Reaction: IXa + ATIII_Heparin => IXa_ATIII_Heparin, Rate Law: compartment_1*IXa*ATIII_Heparin/c46
v=0.1; k=10.0Reaction: VII => VIIa; IIa, Rate Law: compartment_1*v*VII*IIa/(k+IIa)
c45 = 0.85Reaction: Xa + ATIII_Heparin => Xa_ATIII_Heparin, Rate Law: compartment_1*Xa*ATIII_Heparin/c45
k=1.0; v=7.0Reaction: XIII => XIIIa; IIa, Rate Law: compartment_1*v*XIII*IIa/(k+IIa)
v=1.0; k=10.0Reaction: VII => VIIa; Xa, Rate Law: compartment_1*v*VII*Xa/(k+Xa)
v=0.2; k=10.0Reaction: VII => VIIa; IXa, Rate Law: compartment_1*v*VII*IXa/(k+IXa)
d_VIII = 0.058Reaction: VIII =>, Rate Law: compartment_1*d_VIII*VIII
Fg0 = 8945.5; d_Fg = 0.032Reaction: => Fg, Rate Law: compartment_1*Fg0*d_Fg
k1=20.0Reaction: VIIa_TF_Xa_TFPI =>, Rate Law: compartment_1*k1*VIIa_TF_Xa_TFPI
k=1.0; v=70.0Reaction: IX => IXa; VIIa_TF, Rate Law: compartment_1*v*IX*VIIa_TF/(k+VIIa_TF)
k=0.1; v=2.0Reaction: X => Xa; IXa_VIIIa, Rate Law: compartment_1*v*X*IXa_VIIIa/(k+IXa_VIIIa)
v=10.0; k=10.0Reaction: XI => XIa; IIa, Rate Law: compartment_1*v*XI*IIa/(k+IIa)
k1=0.1Reaction: D =>, Rate Law: compartment_1*k1*D
k=500.0; v=9.0Reaction: II => IIa; Xa, Rate Law: compartment_1*v*II*Xa/(k+Xa)
v=100.0; k=10.0Reaction: II => IIa; Va_Xa, Rate Law: compartment_1*v*II*Va_Xa/(k+Va_Xa)
v=0.02; k=10.0Reaction: X => Xa; IXa, Rate Law: compartment_1*v*X*IXa/(k+IXa)
k=500.0; v=500.0Reaction: Fg => FDP; P, Rate Law: compartment_1*v*Fg*P/(k+P)
c=0.1Reaction: VII + TF => VII_TF, Rate Law: compartment_1*VII*TF/c
heparin_ke = 0.693Reaction: IIa_ATIII_Heparin =>, Rate Law: compartment_1*heparin_ke*IIa_ATIII_Heparin
v=7.0; k=5000.0Reaction: Pg => P; IIa, Rate Law: compartment_1*v*Pg*IIa/(k+IIa)
v=7.0; k=100.0Reaction: XF => D; P, Rate Law: compartment_1*v*XF*P/(k+P)
k1=3.5Reaction: FDP =>, Rate Law: compartment_1*k1*FDP
k=1.0; v=50.0Reaction: VIIIa => ; APC_PS, Rate Law: compartment_1*v*VIIIa*APC_PS/(k+APC_PS)
c=0.01Reaction: IXa + VIIIa => IXa_VIIIa, Rate Law: compartment_1*IXa*VIIIa/c
d_VIII = 0.058; VIII0 = 0.7Reaction: => VIII, Rate Law: compartment_1*VIII0*d_VIII
d_II = 0.01Reaction: II =>, Rate Law: compartment_1*d_II*II
c44 = 0.119718309859155Reaction: IIa + ATIII_Heparin => IIa_ATIII_Heparin, Rate Law: compartment_1*IIa*ATIII_Heparin/c44
d_IX = 0.029; VKH20 = 0.1; IX0 = 89.6Reaction: => IX; VKH2, Rate Law: compartment_1*d_IX*IX0*VKH2/VKH20
d_XI = 0.1; XI0 = 30.6Reaction: => XI, Rate Law: compartment_1*XI0*d_XI
d_V = 0.043; V0 = 26.7Reaction: => V, Rate Law: compartment_1*V0*d_V
d_Fg = 0.032Reaction: Fg => FDP, Rate Law: compartment_1*d_Fg*Fg
d_VII = 0.12Reaction: VII =>, Rate Law: compartment_1*d_VII*VII
k1=67.4Reaction: IIa => TAT, Rate Law: compartment_1*k1*IIa
v=900.0; k=200.0Reaction: X => Xa; VIIa_TF, Rate Law: compartment_1*v*X*VIIa_TF/(k+VIIa_TF)
vitaminK_k21_Vc = 5.08333333333333E-4; vitaminK_k12 = 0.0587Reaction: VK => VK_p, Rate Law: compartment_1*(vitaminK_k12*VK-vitaminK_k21_Vc*VK_p)
VII0 = 10.0; VKH20 = 0.1; d_VII = 0.12Reaction: => VII; VKH2, Rate Law: compartment_1*d_VII*VII0*VKH2/VKH20
d_X = 0.018; VKH20 = 0.1; X0 = 174.3Reaction: => X; VKH2, Rate Law: compartment_1*d_X*X0*VKH2/VKH20
warfarin_ka = 1.0Reaction: A_warf = (-warfarin_ka)*A_warf, Rate Law: (-warfarin_ka)*A_warf
d_X = 0.018Reaction: X =>, Rate Law: compartment_1*d_X*X
Pk0 = 450.0; d_Pk = 0.05Reaction: => Pk, Rate Law: compartment_1*Pk0*d_Pk
d_IX = 0.029Reaction: IX =>, Rate Law: compartment_1*d_IX*IX
d_Pk = 0.05Reaction: Pk =>, Rate Law: compartment_1*d_Pk*Pk
d_V = 0.043Reaction: V =>, Rate Law: compartment_1*d_V*V

States:

NameDescription
F[Fibrinogen alpha chain; Fibrinogen gamma chain; Fibrinogen beta chain]
VIIa TF Xa TFPI[Tissue factor; Coagulation factor X; Coagulation factor VII; Tissue factor pathway inhibitor]
VIII[Coagulation factor VIII]
P[Plasminogen]
V[Coagulation factor V]
A warf[warfarin]
APC PS[Vitamin K-dependent protein C; Vitamin K-dependent protein S]
Xa[Coagulation factor X]
Pk[Plasma kallikrein]
FDP[Fibrinogen alpha chain; Fibrinogen gamma chain; Fibrinogen beta chain]
DPDP
IIa ATIII Heparin[heparin; Antithrombin-III; Prothrombin]
XIa[Coagulation factor XI]
Fg[Fibrinogen alpha chain; Fibrinogen gamma chain; Fibrinogen beta chain]
X[Coagulation factor X]
VK p[vitamin K]
DD
VIIIa[Coagulation factor VIII]
Va[Coagulation factor V]
IIa[Prothrombin]
VIIa[Coagulation factor VII]
XIIIa[Coagulation factor XIII A chain]
XI[Coagulation factor XI]
XIIa[Coagulation factor XII]
IXa ATIII Heparin[heparin; Coagulation factor IX; Antithrombin-III]
IXa[Coagulation factor IX]
VII[Coagulation factor VII]
II[Prothrombin]
IX[Coagulation factor IX]
IXa VIIIa[Coagulation factor IX; Coagulation factor VIII]

Walsh2014 - Inhibition kinetics of DAPT on APP Cleavage: BIOMD0000000617v0.0.1

Walsh2014 - Inhibition kinetics of DAPT on APP CleavageThis model is described in the article: [Are improper kinetic mo…

Details

Reproducibility of biological data is a significant problem in research today. One potential contributor to this, which has received little attention, is the over complication of enzyme kinetic inhibition models. The over complication of inhibitory models stems from the common use of the inhibitory term (1 + [I]/Ki ), an equilibrium binding term that does not distinguish between inhibitor binding and inhibitory effect. Since its initial appearance in the literature, around a century ago, the perceived mechanistic methods used in its production have spurred countless inhibitory equations. These equations are overly complex and are seldom compared to each other, which has destroyed their usefulness resulting in the proliferation and regulatory acceptance of simpler models such as IC50s for drug characterization. However, empirical analysis of inhibitory data recognizing the clear distinctions between inhibitor binding and inhibitory effect can produce simple logical inhibition models. In contrast to the common divergent practice of generating new inhibitory models for every inhibitory situation that presents itself. The empirical approach to inhibition modeling presented here is broadly applicable allowing easy comparison and rational analysis of drug interactions. To demonstrate this, a simple kinetic model of DAPT, a compound that both activates and inhibits γ-secretase is examined using excel. The empirical kinetic method described here provides an improved way of probing disease mechanisms, expanding the investigation of possible therapeutic interventions. link: http://identifiers.org/pubmed/25374788

Parameters:

NameDescription
S = 61.0; V1s = 64.680648010584; K1s = 37.3401755830905Reaction: => v, Rate Law: default_compartment*Compartment_*V1s*S/(S+K1s)
S = 61.0; K3s = 605.01; H2 = 2.69; V2s = 32.4269355627923Reaction: v =>, Rate Law: default_compartment*Compartment_*V2s*S^H2/(S^H2+K3s^H2)
H1 = 1.71; K2s = 126.236082446952; S = 61.0; V1s = 64.680648010584Reaction: v =>, Rate Law: default_compartment*Compartment_*V1s*S^H1/(S^H1+K2s^H1)
H1 = 1.71; K2s = 126.236082446952; S = 61.0; V2s = 32.4269355627923Reaction: => v, Rate Law: default_compartment*Compartment_*V2s*S^H1/(S^H1+K2s^H1)

States:

NameDescription
v[DAPT; Gamma-secretase subunit PEN-2]

Wan2020 - risk estimation and prediction of the transmission of COVID-19 in maninland China excluding Hubei province: BIOMD0000000981v0.0.1

Background: In December 2019, an outbreak of coronavirus disease (later named as COVID-19) was identified in Wuhan, Chin…

Details

BACKGROUND:In December 2019, an outbreak of coronavirus disease (later named as COVID-19) was identified in Wuhan, China and, later on, detected in other parts of China. Our aim is to evaluate the effectiveness of the evolution of interventions and self-protection measures, estimate the risk of partial lifting control measures and predict the epidemic trend of the virus in the mainland of China excluding Hubei province based on the published data and a novel mathematical model. METHODS:A novel COVID-19 transmission dynamic model incorporating the intervention measures implemented in China is proposed. COVID-19 daily data of the mainland of China excluding Hubei province, including the cumulative confirmed cases, the cumulative deaths, newly confirmed cases and the cumulative recovered cases between 20 January and 3 March 2020, were archived from the National Health Commission of China (NHCC). We parameterize the model by using the Markov Chain Monte Carlo (MCMC) method and estimate the control reproduction number (Rc), as well as the effective daily reproduction ratio- Re(t), of the disease transmission in the mainland of China excluding Hubei province. RESULTS:The estimation outcomes indicate that Rc is 3.36 (95% CI: 3.20-3.64) and Re(t) has dropped below 1 since 31 January 2020, which implies that the containment strategies implemented by the Chinese government in the mainland of China are indeed effective and magnificently suppressed COVID-19 transmission. Moreover, our results show that relieving personal protection too early may lead to a prolonged disease transmission period and more people would be infected, and may even cause a second wave of epidemic or outbreaks. By calculating the effective reproduction ratio, we prove that the contact rate should be kept at least less than 30% of the normal level by April, 2020. CONCLUSIONS:To ensure the pandemic ending rapidly, it is necessary to maintain the current integrated restrict interventions and self-protection measures, including travel restriction, quarantine of entry, contact tracing followed by quarantine and isolation and reduction of contact, like wearing masks, keeping social distance, etc. People should be fully aware of the real-time epidemic situation and keep sufficient personal protection until April. If all the above conditions are met, the outbreak is expected to be ended by April in the mainland of China apart from Hubei province. link: http://identifiers.org/pubmed/32831142

Wanant2000_InsulinReceptorModel_A: MODEL1201140005v0.0.1

This a model from the article: Insulin receptor binding kinetics: modeling and simulation studies. Wanant S, Quon MJ…

Details

Biological actions of insulin regulate glucose metabolism and other essential physiological functions. Binding of insulin to its cell surface receptor initiates signal transduction pathways that mediate cellular responses. Thus, it is of great interest to understand the mechanisms underlying insulin receptor binding kinetics. Interestingly, negative cooperative interactions are observed at high insulin concentrations while positive cooperativity may be present at low insulin concentrations. Clearly, insulin receptor binding kinetics cannot be simply explained by a classical bimolecular reaction. Mature insulin receptors have a dimeric structure capable of binding two molecules of insulin. The binding affinity of the receptor for the second insulin molecule is significantly lower than for the first bound insulin molecule. In addition, insulin receptor aggregation occurs in response to ligand binding and aggregation may also influence binding kinetics. In this study, we develop a mathematical model for insulin receptor binding kinetics that explicitly represents the divalent nature of the insulin receptor and incorporates receptor aggregation into the kinetic model. Model parameters are based upon published data where available. Computer simulations with our model are capable of reproducing both negative and positive cooperativity at the appropriate insulin concentrations. This model may be a useful tool for helping to understand the mechanisms underlying insulin receptor binding and the coupling of receptor binding to downstream signaling events. link: http://identifiers.org/pubmed/10882558

Wanant2000_InsulinReceptorModel_B: MODEL1201140006v0.0.1

This a model from the article: Insulin receptor binding kinetics: modeling and simulation studies. Wanant S, Quon MJ…

Details

Biological actions of insulin regulate glucose metabolism and other essential physiological functions. Binding of insulin to its cell surface receptor initiates signal transduction pathways that mediate cellular responses. Thus, it is of great interest to understand the mechanisms underlying insulin receptor binding kinetics. Interestingly, negative cooperative interactions are observed at high insulin concentrations while positive cooperativity may be present at low insulin concentrations. Clearly, insulin receptor binding kinetics cannot be simply explained by a classical bimolecular reaction. Mature insulin receptors have a dimeric structure capable of binding two molecules of insulin. The binding affinity of the receptor for the second insulin molecule is significantly lower than for the first bound insulin molecule. In addition, insulin receptor aggregation occurs in response to ligand binding and aggregation may also influence binding kinetics. In this study, we develop a mathematical model for insulin receptor binding kinetics that explicitly represents the divalent nature of the insulin receptor and incorporates receptor aggregation into the kinetic model. Model parameters are based upon published data where available. Computer simulations with our model are capable of reproducing both negative and positive cooperativity at the appropriate insulin concentrations. This model may be a useful tool for helping to understand the mechanisms underlying insulin receptor binding and the coupling of receptor binding to downstream signaling events. link: http://identifiers.org/pubmed/10882558

Wang1996_Synaptic_Inhibition_Two_Neuron: BIOMD0000000302v0.0.1

This is a model of one presynaptic and one postsynaptic cell, as described in the article: **Gamma oscillation by synap…

Details

Fast neuronal oscillations (gamma, 20-80 Hz) have been observed in the neocortex and hippocampus during behavioral arousal. Using computer simulations, we investigated the hypothesis that such rhythmic activity can emerge in a random network of interconnected GABAergic fast-spiking interneurons. Specific conditions for the population synchronization, on properties of single cells and the circuit, were identified. These include the following: (1) that the amplitude of spike afterhyperpolarization be above the GABAA synaptic reversal potential; (2) that the ratio between the synaptic decay time constant and the oscillation period be sufficiently large; (3) that the effects of heterogeneities be modest because of a steep frequency-current relationship of fast-spiking neurons. Furthermore, using a population coherence measure, based on coincident firings of neural pairs, it is demonstrated that large-scale network synchronization requires a critical (minimal) average number of synaptic contacts per cell, which is not sensitive to the network size. By changing the GABAA synaptic maximal conductance, synaptic decay time constant, or the mean external excitatory drive to the network, the neuronal firing frequencies were gradually and monotonically varied. By contrast, the network synchronization was found to be high only within a frequency band coinciding with the gamma (20-80 Hz) range. We conclude that the GABAA synaptic transmission provides a suitable mechanism for synchronized gamma oscillations in a sparsely connected network of fast-spiking interneurons. In turn, the interneuronal network can presumably maintain subthreshold oscillations in principal cell populations and serve to synchronize discharges of spatially distributed neurons. link: http://identifiers.org/pubmed/8815919

Wang2007 - ATP induced intracellular Calcium Oscillation: BIOMD0000000145v0.0.1

Wang2007 - ATP induced intracellular Calicum Oscillation The model simulate the ATP-induced intracellular Ca2+ oscillati…

Details

A quantitative kinetic model is proposed to simulate the ATP-induced intracellular Ca(2+) oscillations. The quantitative effect of ATP concentration upon the oscillations was successfully simulated. Our simulation results support previous experimental explanations that the Ca(2+) oscillations are mainly due to interaction of Ca(2+) release from the endoplasmic reticulum (ER) and the ATP-dependent Ca(2+) pump back into the ER, and the oscillations are prolonged by extracellular Ca(2+) entry that maintains the constant Ca(2+) supplies to its intracellular stores. The model is also able to simulate the sudden disappearance phenomenon of the Ca(2+) oscillations observed in some cell types by taking into account of the biphasic characteristic of the Ca(2+) release from the endoplasmic reticulum (ER). Moreover, the model simulation results for the Ca(2+) oscillations characteristics such as duration, peak Ca(2+), and average interval, etc., lead to prediction of some possible factors responsible for the variations of Ca(2+) oscillations in different types of cells. link: http://identifiers.org/pubmed/17188305

Parameters:

NameDescription
k11 = 260.0Reaction: => Ca_Cyt, Rate Law: Cytosol*k11
Rer = 0.0; Rcyt1 = 0.0; k8 = 10500.0; Rip3 = 0.0; k9 = 600.0Reaction: Ca_ER => Ca_Cyt, Rate Law: ER*(k8*Rip3*Rer-k9*Rcyt1)
k0 = 0.1Reaction: => Galpha_GTP, Rate Law: Cytosol*k0
k1 = 3.4Reaction: => Galpha_GTP, Rate Law: Cytosol*k1*Galpha_GTP
k7 = 2.0Reaction: IP3 =>, Rate Law: Cytosol*k7*IP3
Cplc_total = 10.0Reaction: PLC = Cplc_total-APLC, Rate Law: missing
k6 = 14.0Reaction: => IP3; APLC, Rate Law: Cytosol*k6*APLC
k3 = 4.5; Rpkc = 0.0Reaction: Galpha_GTP =>, Rate Law: Cytosol*k3*Rpkc*Galpha_GTP
k10 = 3000.0; Rcyt2 = 0.0Reaction: Ca_Cyt =>, Rate Law: Cytosol*k10*Rcyt2
Rgalpha_gtp = 0.0; k4 = 1.2; Rdg = 0.0Reaction: => APLC; PLC, Rate Law: Cytosol*k4*Rgalpha_gtp*Rdg*PLC
k5 = 0.12Reaction: APLC =>, Rate Law: Cytosol*k5*APLC
k2 = 4.0; Raplc = 0.0Reaction: Galpha_GTP =>, Rate Law: Cytosol*k2*Raplc*Galpha_GTP

States:

NameDescription
IP3[1D-myo-inositol 1,4,5-trisphosphate]
APLC[1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase beta-1]
Ca Cyt[calcium(2+)]
DG[diglyceride]
Galpha GTP[GTP; Guanine nucleotide-binding protein subunit alpha-11]
Ca ER[calcium(2+)]
PLC[1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase beta-1]

Wang2008 - Mimicking the inhibitory effect of riluzole on membrane conductance in skeletal fibres: BIOMD0000000693v0.0.1

Wang2008 - Mimicking the inhibitory effect of riluzole on membrane conductance in skeletal fibresThis model is described…

Details

Riluzole is known to be of therapeutic use in the management of amyotrophic lateral sclerosis. In this study, we investigated the effects of riluzole on ion currents in cultured differentiated human skeletal muscle cells (dHSkMCs). Western blotting revealed the protein expression of alpha-subunits for both large-conductance Ca2+-activated K+ (BK(Ca)) channel and Na+ channel (Na(v)1.5) in these cells. Riluzole could reduce the frequency of spontaneous beating in dHSkMCs. In whole-cell configuration, riluzole suppressed voltage-gated Na+ current (I(Na)) in a concentration-dependent manner with an IC50 value of 2.3 microM. Riluzole (10 microM) also effectively increased Ca2+-activated K+ current (I(K(Ca))) which could be reversed by iberiotoxin (200 nM) and paxilline (1 microM), but not by apamin (200 nM). In inside-out patches, when applied to the inside of the cell membrane, riluzole (10 microM) increased BK(Ca)-channel activity with a decrease in mean closed time. Simulation studies also unraveled that both decreased conductance of I(Na) and increased conductance of I(K(Ca)) utilized to mimic riluzole actions in skeletal muscle cells could combine to decrease the amplitude of action potentials and increase the repolarization of action potentials. Taken together, inhibition of I(Na) and stimulation of BK(Ca)-channel activity caused by this drug are partly, if not entirely, responsible for its muscle relaxant actions in clinical setting. link: http://identifiers.org/pubmed/18068197

Parameters:

NameDescription
EK = -70.0; gk_max = 0.42Reaction: IK = gk_max*n^4*(Vm-EK), Rate Law: missing
v_beta_m = 18.0; beta_m_max = 2.081; Em = -42.0Reaction: beta_m = beta_m_max*exp((Em-Vm)/v_beta_m), Rate Law: missing
kpmca = 0.2; alpha = 4.5E-6Reaction: jmem = -(alpha*ICa+kpmca*c), Rate Law: missing
ooinf = 0.013131127960815; tau = 3.69947662338091Reaction: o = (ooinf-o)/tau, Rate Law: (ooinf-o)/tau
EK = -70.0; gKca = 0.5Reaction: IKCa = gKca*o*w*(Vm-EK), Rate Law: missing
v_beta_h = 7.6; beta_h_max = 3.382; Eh = -41.0Reaction: beta_h = beta_h_max/(1+exp((Eh-Vm)/v_beta_h)), Rate Law: missing
kd = 0.18Reaction: w = c^5/(c^5+kd^5), Rate Law: missing
En = -40.0; v_alpha_n = 7.0; alpha_n_max = 0.0229Reaction: alpha_n = alpha_n_max*(Vm-En)/(1-exp((En-Vm)/v_alpha_n)), Rate Law: missing
En = -40.0; beta_n_max = 0.09616; v_beta_n = 40.0Reaction: beta_n = beta_n_max*exp((En-Vm)/v_beta_n), Rate Law: missing
alphad = 8.05558916679958E-5; betad = 0.00696831163375364Reaction: d = (1-d)*alphad-d*betad, Rate Law: (1-d)*alphad-d*betad
gNa_max = 0.9; ENa = 50.0Reaction: INa = gNa_max*m^3*h*(Vm-ENa), Rate Law: missing
pleak = 5.0E-4Reaction: jleak = pleak*(cer-c), Rate Law: missing
kserca = 0.4Reaction: jserca = kserca*c, Rate Law: missing
alpha_m_max = 0.208; v_alpha_m = 10.0; Em = -42.0Reaction: alpha_m = alpha_m_max*(Vm-Em)/(1-exp((Em-Vm)/v_alpha_m)), Rate Law: missing
vcytver = 5.0; fer = 0.01Reaction: cer = (-fer)*vcytver*jer, Rate Law: (-fer)*vcytver*jer
Rs = 15.0Reaction: IT = (Vm-Vt)/Rs, Rate Law: missing
Cm = 0.009Reaction: Vm = (Stimulus-(INa+ICa+IK+IL+IT+IKCa))/Cm, Rate Law: (Stimulus-(INa+ICa+IK+IL+IT+IKCa))/Cm
fcyt = 0.01Reaction: c = fcyt*(jmem+jer), Rate Law: fcyt*(jmem+jer)
gca = -3.75057964796293Reaction: ICa = gca*d^2, Rate Law: missing
alpha_h_max = 0.0156; v_alpha_m = 10.0; Eh = -41.0Reaction: alpha_h = alpha_h_max*exp((Eh-Vm)/v_alpha_m), Rate Law: missing
gL_max = 0.0024; EL = -75.0Reaction: IL = gL_max*(Vm-EL), Rate Law: missing
Stimulus_Start = 5.0; Stimulus_Magnitude = 2.0; Stimulus_Duration = 1.0; Stimulus_Period = 50.0Reaction: Stimulus = piecewise(Stimulus_Magnitude, (time >= Stimulus_Start) && (((time-Stimulus_Start)-floor((time-Stimulus_Start)/Stimulus_Period)*Stimulus_Period) < Stimulus_Duration), 0), Rate Law: missing
Rs = 15.0; Ct = 0.04Reaction: Vt = (Vm-Vt)/(Rs*Ct), Rate Law: (Vm-Vt)/(Rs*Ct)

States:

NameDescription
jmemjmem
nn
cc
jerjer
beta nbeta_n
hh
jleakjleak
cercer
Stimulus[Stimulus]
IKCa[calcium-activated potassium channel activity]
alpha malpha_m
beta mbeta_m
alpha halpha_h
Vm[Membrane Potential]
dd
ITIT
ww
ICaICa
oo
INaINa
IKIK
mm
alpha nalpha_n
jsercajserca
VtVt
ILIL
beta hbeta_h

Wang2008_Neonatal_heartfunction: MODEL7814665196v0.0.1

This a model from the article: Mathematical model of the neonatal mouse ventricular action potential. Wang LJ, Sobie…

Details

Therapies for heart disease are based largely on our understanding of the adult myocardium. The dramatic differences in action potential (AP) shape between neonatal and adult cardiac myocytes, however, indicate that a different set of molecular interactions in neonatal myocytes necessitates different treatment for newborns. Computational modeling is useful for synthesizing data to determine how interactions between components lead to systems-level behavior, but this technique has not been used extensively to study neonatal heart cell function. We created a mathematical model of the neonatal (day 1) mouse myocyte by modifying, on the basis of experimental data, the densities and/or formulations of ion transport mechanisms in an adult cell model. The new model reproduces the characteristic AP shape of neonatal cells, with a brief plateau phase and longer duration than the adult (action potential duration at 80% repolarization = 60.1 vs. 12.6 ms). The simulation results are consistent with experimental data, including 1) decreased density and altered inactivation of transient outward K+ currents, 2) increased delayed rectifier K+ currents, 3) Ca2+ entry through T-type as well as L-type Ca2+ channels, 4) increased Ca2+ influx through Na+/Ca2+ exchange, and 5) Ca2+ transients resulting from transmembrane Ca2+ entry rather than release from the sarcoplasmic reticulum (SR). Simulations performed with the model generated novel predictions, including increased SR Ca2+ leak and elevated intracellular Na+ concentration in neonatal compared with adult myocytes. This new model can therefore be used for testing hypotheses and obtaining a better quantitative understanding of differences between neonatal and adult physiology. link: http://identifiers.org/pubmed/18408122

Wang2009 - PI3K Ras Crosstalk: BIOMD0000000288v0.0.1

This model is from the article: PI3K-dependent cross-talk interactions converge with Ras as quantifiable inputs integr…

Details

Although it is appreciated that canonical signal-transduction pathways represent dominant modes of regulation embedded in larger interaction networks, relatively little has been done to quantify pathway cross-talk in such networks. Through quantitative measurements that systematically canvas an array of stimulation and molecular perturbation conditions, together with computational modeling and analysis, we have elucidated cross-talk mechanisms in the platelet-derived growth factor (PDGF) receptor signaling network, in which phosphoinositide 3-kinase (PI3K) and Ras/extracellular signal-regulated kinase (Erk) pathways are prominently activated. We show that, while PI3K signaling is insulated from cross-talk, PI3K enhances Erk activation at points both upstream and downstream of Ras. The magnitudes of these effects depend strongly on the stimulation conditions, subject to saturation effects in the respective pathways and negative feedback loops. Motivated by those dynamics, a kinetic model of the network was formulated and used to precisely quantify the relative contributions of PI3K-dependent and -independent modes of Ras/Erk activation. link: http://identifiers.org/pubmed/19225459

Parameters:

NameDescription
KGP = 5.09; KGR = 495.0Reaction: eGEF = (KGR*c2+KGP*m3PI)/(1+KGR*c2+KGP*m3PI)*fGEF, Rate Law: missing
k3PI = 1.0Reaction: => m3PI; ePI3K, Rate Law: k3PI*(ePI3K-m3PI)
kappaPI3K = 0.3; alphaPI3K = 80.0Reaction: ePI3K = ((1+kappaPI3K+2*alphaPI3K*c2)-((1+kappaPI3K+2*alphaPI3K*c2)^2-8*alphaPI3K*c2)^0.5)/2, Rate Law: missing
KMx11 = 30.3; KMx22 = 9.59; KMx12 = 18.6; VmaxOVERKMx11 = 1.18; KMyph2 = 7.99; VmaxOVERKMx21 = 0.405; VmaxOVERKMyph1 = 1.65; KMx21 = 13.7; KMyph1 = 23.0Reaction: => y; yp, ypp, x1, x2, Rate Law: (VmaxOVERKMyph1*yp/(1+yp/KMyph1+ypp/KMyph2)-VmaxOVERKMx11*x1*y/(1+y/KMx11+yp/KMx12))-VmaxOVERKMx21*x2*y/(1+y/KMx21+yp/KMx22)
L = 1.0; KDL = 1.5Reaction: c1 = L*sumrc1/(KDL+L), Rate Law: missing
KMy1 = 9.91; KMx11 = 30.3; KMx22 = 9.59; KMy2 = 8.81; VmaxOVERKMx12 = 3.45; KMx12 = 18.6; VmaxOVERKMyph2 = 4.2; KMyph2 = 7.99; VmaxOVERKMx22 = 1.09; KMx21 = 13.7; KMyph1 = 23.0Reaction: => ypp; x1, yp, y, x2, z, zp, Rate Law: (VmaxOVERKMx12*x1*yp/(1+y/KMx11+yp/KMx12)+VmaxOVERKMx22*x2*yp/(1+y/KMx21+yp/KMx22))-VmaxOVERKMyph2*ypp/((1+z/KMy1+zp/KMy2)*(1+yp/KMyph1)+ypp/KMyph2)
KMy1 = 9.91; KMy2 = 8.81; KMzph2 = 31.5; VmaxOVERKMy1 = 6.57; KMzph1 = 8.27; VmaxOVERKMzph1 = 0.167Reaction: => z; eph, zp, zpp, ypp, Rate Law: VmaxOVERKMzph1*eph*zp/(1+zp/KMzph1+zpp/KMzph2)-VmaxOVERKMy1*ypp*z/(1+z/KMy1+zp/KMy2)
kt = 0.005; kxR0 = 0.3; kminusx = 0.007Reaction: => sumrc1; c2, c1, Rate Law: kt*(1-sumrc1)+2*(kminusx*c2-kxR0*c1^2)
KMx11 = 30.3; KMx12 = 18.6; kdx1 = 0.745Reaction: => x1; mRas, y, yp, Rate Law: kdx1*(mRas-x1/(1+y/KMx11+yp/KMx12))
Zf = 0.272; kFBf = 0.976; Kf = 3.76; n = 1.03Reaction: => fGEF; zpp, Rate Law: kFBf*((1-fGEF)/Kf-zpp^n/(Zf^n+zpp^n)*fGEF)
KDL = 1.5; L = 1.0Reaction: r = KDL*sumrc1/(KDL+L), Rate Law: missing
Gamma = 0.1; kRas = 1.0Reaction: => mRas; eGEF, Rate Law: kRas*((1+Gamma)*eGEF-(1+Gamma*eGEF)*mRas)
KMy1 = 9.91; KMy2 = 8.81; KMzph2 = 31.5; VmaxOVERKMy2 = 31.9; VmaxOVERKMzph2 = 0.228; KMzph1 = 8.27Reaction: => zpp; ypp, zp, z, eph, Rate Law: VmaxOVERKMy2*ypp*zp/(1+z/KMy1+zp/KMy2)-VmaxOVERKMzph2*eph*zpp/(1+zp/KMzph1+zpp/KMzph2)
Wph = 0.385; Kph = 4.64; p = 1.98; kFBph = 2.34Reaction: => eph; w, Rate Law: kFBph*(w^p/(Wph^p+w^p)-(eph-1)/Kph)
ke = 0.2; kxR0 = 0.3; kminusx = 0.007Reaction: => c2; c1, Rate Law: kxR0*c1^2-(kminusx+ke)*c2
kdw = 0.0333Reaction: => w; zpp, Rate Law: kdw*(zpp-w)
KMx22 = 9.59; kdx2 = 2.85; Kx2 = 6.77; KMx21 = 13.7Reaction: => x2; m3PI, y, yp, Rate Law: kdx2*((1+Kx2)*m3PI/(1+Kx2*m3PI)-x2/(1+y/KMx21+yp/KMx22))

States:

NameDescription
c2[Platelet-derived growth factor receptor-like protein]
c1[Platelet-derived growth factor D; Platelet-derived growth factor receptor-like protein; Platelet-derived growth factor C; Platelet-derived growth factor receptor-like protein; Platelet-derived growth factor subunit B; Platelet-derived growth factor receptor-like protein; Platelet-derived growth factor subunit A; Platelet-derived growth factor receptor-like protein]
eph[phosphatase activity]
eGEF[Ras-specific guanine nucleotide-releasing factor 2]
w[obsolete transcription activator activity]
x2[Serine/threonine-protein kinase PAK 1]
ypp[Dual specificity mitogen-activated protein kinase kinase 2]
r[Platelet-derived growth factor receptor-like protein]
ePI3K[Phosphatidylinositol 4-phosphate 3-kinase C2 domain-containing subunit beta]
x1[RAF proto-oncogene serine/threonine-protein kinase]
fGEF[Ras-specific guanine nucleotide-releasing factor 2]
zpp[Mitogen-activated protein kinase 3]
m3PI[lipid droplet]
sumrc1[Platelet-derived growth factor receptor-like protein; Platelet-derived growth factor receptor-like protein; Platelet-derived growth factor C; Platelet-derived growth factor receptor-like protein; Platelet-derived growth factor D; Platelet-derived growth factor receptor-like protein; Platelet-derived growth factor subunit B; Platelet-derived growth factor subunit A; Platelet-derived growth factor receptor-like protein]
mRas[Ras-related protein M-Ras]
zp[Mitogen-activated protein kinase 3]
yp[Dual specificity mitogen-activated protein kinase kinase 2]
z[Mitogen-activated protein kinase 3]
y[Dual specificity mitogen-activated protein kinase kinase 2]

Wang2016/1 - oncolytic efficacy of M1 virus-SNTM model: BIOMD0000000780v0.0.1

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

Details

Motivated by the latest findings in a recent medical experiment [19] which identify a naturally occurring alphavirus (M1) as a novel selective killer targeting zinc-finger antiviral protein (ZAP)-deficient cancer cells, we propose a mathematical model to illustrate the growth of normal cells, tumor cells and the M1 virus with limited nutrient. In order to better understand biological mechanisms, we discuss two cases of the model: without competition and with competition. In the first part, the explicit threshold conditions for the persistence of normal cells (or tumor cells) is obtained accompanying with the biological explanations. The second part indicates that when competing with tumor cells, the normal cells will extinct if M1 virus is ignored; Whereas, when M1 virus is considered, the growth trend of normal cells is similar to the one without competition. And by using uniformly strong repeller theorem, the minimum effective dosage of medication is explicitly found which is not reported in [19]. Furthermore, numerical simulations and corresponding biological interpretations are given to support our results. link: http://identifiers.org/pubmed/26976483

Parameters:

NameDescription
u2 = 0.5 1Reaction: S => ; T, Rate Law: tme*u2*S*T
u3 = 0.1 1; r3 = 0.5 1Reaction: => M; T, Rate Law: tme*r3*u3*T*M
r2 = 0.8 1; u2 = 0.4 1Reaction: => T; S, Rate Law: tme*r2*u2*S*T
d = 0.02 1; e1 = 0.01 1Reaction: N =>, Rate Law: tme*(d+e1)*N
b = 0.001 1Reaction: => M, Rate Law: tme*b
u2 = 0.4 1Reaction: S => ; T, Rate Law: tme*u2*S*T
d = 0.02 1; e2 = 0.04 1Reaction: T =>, Rate Law: tme*(d+e2)*T
r1 = 0.8 1; u1 = 0.2 1Reaction: => N; S, Rate Law: tme*r1*u1*S*N
d = 0.02 1; e3 = 0.01 1Reaction: M =>, Rate Law: tme*(d+e3)*M
d = 0.02 1; e2 = 0.008 1Reaction: T =>, Rate Law: tme*(d+e2)*T
a = 0.02 1Reaction: => S, Rate Law: tme*a
d = 0.02 1Reaction: S =>, Rate Law: tme*d*S
u3 = 0.1 1Reaction: T => ; M, Rate Law: tme*u3*T*M
r2 = 0.8 1; u2 = 0.5 1Reaction: => T; S, Rate Law: tme*r2*u2*S*T
u1 = 0.2 1Reaction: S => ; N, Rate Law: tme*u1*S*N

States:

NameDescription
S[Nutrient]
T[malignant cell]
M[Oncolytic Virus]
N[cell]

Wang2016/2 - oncolytic efficacy of M1 virus-SNT model: BIOMD0000000781v0.0.1

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

Details

Motivated by the latest findings in a recent medical experiment [19] which identify a naturally occurring alphavirus (M1) as a novel selective killer targeting zinc-finger antiviral protein (ZAP)-deficient cancer cells, we propose a mathematical model to illustrate the growth of normal cells, tumor cells and the M1 virus with limited nutrient. In order to better understand biological mechanisms, we discuss two cases of the model: without competition and with competition. In the first part, the explicit threshold conditions for the persistence of normal cells (or tumor cells) is obtained accompanying with the biological explanations. The second part indicates that when competing with tumor cells, the normal cells will extinct if M1 virus is ignored; Whereas, when M1 virus is considered, the growth trend of normal cells is similar to the one without competition. And by using uniformly strong repeller theorem, the minimum effective dosage of medication is explicitly found which is not reported in [19]. Furthermore, numerical simulations and corresponding biological interpretations are given to support our results. link: http://identifiers.org/pubmed/26976483

Parameters:

NameDescription
u2 = 0.5 1Reaction: S => ; T, Rate Law: tme*u2*S*T
r1 = 0.8 1; u1 = 0.2 1Reaction: => N; S, Rate Law: tme*r1*u1*S*N
d = 0.02 1; e2 = 0.008 1Reaction: T =>, Rate Law: tme*(d+e2)*T
a = 0.02 1Reaction: => S, Rate Law: tme*a
d = 0.02 1Reaction: S =>, Rate Law: tme*d*S
d = 0.02 1; e1 = 0.01 1Reaction: N =>, Rate Law: tme*(d+e1)*N
r2 = 0.8 1; u2 = 0.5 1Reaction: => T; S, Rate Law: tme*r2*u2*S*T
u1 = 0.2 1Reaction: S => ; N, Rate Law: tme*u1*S*N

States:

NameDescription
S[Nutrient]
T[malignant cell]
N[cell]

Wang2016/3 - oncolytic efficacy of M1 virus-SN model: BIOMD0000000782v0.0.1

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

Details

Motivated by the latest findings in a recent medical experiment [19] which identify a naturally occurring alphavirus (M1) as a novel selective killer targeting zinc-finger antiviral protein (ZAP)-deficient cancer cells, we propose a mathematical model to illustrate the growth of normal cells, tumor cells and the M1 virus with limited nutrient. In order to better understand biological mechanisms, we discuss two cases of the model: without competition and with competition. In the first part, the explicit threshold conditions for the persistence of normal cells (or tumor cells) is obtained accompanying with the biological explanations. The second part indicates that when competing with tumor cells, the normal cells will extinct if M1 virus is ignored; Whereas, when M1 virus is considered, the growth trend of normal cells is similar to the one without competition. And by using uniformly strong repeller theorem, the minimum effective dosage of medication is explicitly found which is not reported in [19]. Furthermore, numerical simulations and corresponding biological interpretations are given to support our results. link: http://identifiers.org/pubmed/26976483

Parameters:

NameDescription
r1 = 0.8 1; u1 = 0.2 1Reaction: => N; S, Rate Law: tme*r1*u1*S*N
d = 0.02 1; e1 = 0.01 1Reaction: N =>, Rate Law: tme*(d+e1)*N
a = 0.02 1Reaction: => S, Rate Law: tme*a
d = 0.02 1Reaction: S =>, Rate Law: tme*d*S
u1 = 0.2 1Reaction: S => ; N, Rate Law: tme*u1*S*N

States:

NameDescription
S[Nutrient]
N[cell]

Wang2019 - A mathematical model of oncolytic virotherapy with time delay: BIOMD0000000772v0.0.1

A mathematical model describing oncolytic virotherapy with incorporation the viral lytic cycle and the virus-specific CT…

Details

Oncolytic virotherapy is an emerging treatment modality which uses replication-competent viruses to destroy cancers without causing harm to normal tissues. By the development of molecular biotechnology, many effective viruses are adapted or engineered to make them cancer-specific, such as measles, adenovirus, herpes simplex virus and M1 virus. A successful design of virus needs a full understanding about how viral and host parameters influence the tumor load. In this paper, we propose a mathematical model on the oncolytic virotherapy incorporating viral lytic cycle and virus-specific CTL response. Thresholds for viral treatment and virus-specific CTL response are obtained. Different protocols are given depending on the thresholds. Our results also support that immune suppressive drug can enhance the oncolytic effect of virus as reported in recent literature. link: http://identifiers.org/pubmed/31137188

Parameters:

NameDescription
b = 4.48E-4Reaction: x + y => I, Rate Law: compartment*b*x*y
c = 0.02Reaction: => z; y, z, Rate Law: compartment*c*y*z
k = 2139.0; r = 0.206Reaction: => x; x, y, Rate Law: compartment*r*x*(1-(x+y)/k)
a = 1.0Reaction: y =>, Rate Law: compartment*a*y
p = 0.01Reaction: y => ; z, Rate Law: compartment*p*y*z
tau = 0.0; b = 4.48E-4; n = 0.01Reaction: I => y; x, y, Rate Law: compartment*b*exp((-n)*tau)*x*y
d = 0.5Reaction: z =>, Rate Law: compartment*d*z

States:

NameDescription
I[latency-replication decision]
x[cell]
z[cytotoxic T cell]
y[infected cell]

Wang2019 - A mathematical model of oncolytic virotherapy with time delay: BIOMD0000000902v0.0.1

A mathematical model describing oncolytic virotherapy with incorporation the viral lytic cycle and the virus-specific CT…

Details

Oncolytic virotherapy is an emerging treatment modality which uses replication-competent viruses to destroy cancers without causing harm to normal tissues. By the development of molecular biotechnology, many effective viruses are adapted or engineered to make them cancer-specific, such as measles, adenovirus, herpes simplex virus and M1 virus. A successful design of virus needs a full understanding about how viral and host parameters influence the tumor load. In this paper, we propose a mathematical model on the oncolytic virotherapy incorporating viral lytic cycle and virus-specific CTL response. Thresholds for viral treatment and virus-specific CTL response are obtained. Different protocols are given depending on the thresholds. Our results also support that immune suppressive drug can enhance the oncolytic effect of virus as reported in recent literature. link: http://identifiers.org/pubmed/31137188

Parameters:

NameDescription
b = 4.48E-4Reaction: x + y => I, Rate Law: compartment*b*x*y
c = 0.02Reaction: => z; y, z, Rate Law: compartment*c*y*z
k = 2139.0; r = 0.206Reaction: => x; x, y, Rate Law: compartment*r*x*(1-(x+y)/k)
a = 1.0Reaction: y =>, Rate Law: compartment*a*y
p = 0.01Reaction: y => ; z, Rate Law: compartment*p*y*z
tau = 0.0; b = 4.48E-4; n = 0.01Reaction: I => y; x, y, Rate Law: compartment*b*exp((-n)*tau)*x*y
d = 0.5Reaction: z =>, Rate Law: compartment*d*z

States:

NameDescription
I[latency-replication decision]
x[cell]
z[cytotoxic T cell]
y[infected cell]

Warren2009_CalciumWavePropagation: MODEL1006230018v0.0.1

This a model from the article: Mathematical modelling of calcium wave propagation in mammalian airway epithelium: evid…

Details

Airway epithelium has been shown to exhibit intracellular calcium waves after mechanical stimulation. Two classes of mechanism have been proposed to explain calcium wave propagation: diffusion through gap junctions of the intracellular messenger inositol 1,4,5-trisphosphate (IP3), and diffusion of paracrine extracellular messengers such as ATP. We have used single cell recordings of airway epithelium to parameterize a model of an airway epithelial cell. This was then incorporated into a spatial model of a cell culture where both mechanisms for calcium wave propagation are possible. It is shown that a decreasing return on the radius of Ca2+ wave propagation is achieved as the amount of ATP released from the stimulated cell increases. It is therefore shown that for a Ca2+ wave to propagate large distances, a significant fraction of the intracellular ATP pool would be required to be released. Further to this, the radial distribution of maximal calcium response from the stimulated cell does not produce the same flat profile of maximal calcium response seen in experiential studies. This suggests that an additional mechanism is important in Ca2+ wave propagation, such as regenerative release of ATP from cells downstream of the stimulated cell. link: http://identifiers.org/pubmed/19700517

Watanabe2018_Simple markov model: MODEL1803120004v0.0.1

Simple Markov model. There are 3 disease states: Healthy, Sick, and Dead, where the Dead state is terminal. The yearly…

Details

Disease modelers have been modeling progression of diseases for several decades using such tools as Markov models or microsimulation. However, they need to address a serious challenge; many models they create are not reproducible. Moreover, there is no proper practice that ensures reproducible models, since modelers rely on loose guidelines that change periodically, rather than well-defined machine-readable standards. The Systems Biology Markup Language (SBML) is one such standard that allows exchange of models between different software tools. Recently, the SBML Arrays package has been developed, which extends the standard to allow handling simulation of populations. This paper demonstrates through several abstract examples how microsimulation disease models can be encoded using the SBML Arrays package, enabling reproducible disease modeling. link: http://identifiers.org/doi/10.1177/0037549718793214

Watanabe2018_State Transition Model with Treatment and Costs: MODEL1803120006v0.0.1

Model with functions depending on Age, Male, BP (Blood Pressure). There are 3 disease states: Healthy, Sick, and Dead,…

Details

Disease modelers have been modeling progression of diseases for several decades using such tools as Markov models or microsimulation. However, they need to address a serious challenge; many models they create are not reproducible. Moreover, there is no proper practice that ensures reproducible models, since modelers rely on loose guidelines that change periodically, rather than well-defined machine-readable standards. The Systems Biology Markup Language (SBML) is one such standard that allows exchange of models between different software tools. Recently, the SBML Arrays package has been developed, which extends the standard to allow handling simulation of populations. This paper demonstrates through several abstract examples how microsimulation disease models can be encoded using the SBML Arrays package, enabling reproducible disease modeling. link: http://identifiers.org/doi/10.1177/0037549718793214

Waugh2006 - Diabetic Wound Healing - Macrophage Dynamics: BIOMD0000000679v0.0.1

This a model from the article: Macrophage dynamics in diabetic wound dealing. Waugh HV, Sherratt JA. Bull Math Biol…

Details

Wound healing in diabetes is a complex process, characterised by a chronic inflammation phase. The exact mechanism by which this occurs is not fully understood, and whilst several treatments for healing diabetic wounds exist, very little research has been conducted towards the causes of the extended inflammation phase. We describe a mathematical model which offers a possible explanation for diabetic wound healing in terms of the distribution of macrophage phenotypes being altered in the diabetic patient compared to normal wound repair. As a consequence of this, we put forward a suggestion for treatment based on rectifying the macrophage phenotype imbalance. link: http://identifiers.org/pubmed/16794927

Parameters:

NameDescription
d1 = 0.2 1/(0.0115741*ms)Reaction: phi_I =>, Rate Law: COMpartment*d1*phi_I
d2 = 9.1 1/(0.0115741*ms)Reaction: T =>, Rate Law: COMpartment*d2*T
k4 = 0.07 0.0864*µg/sReaction: => T; phi_I, Rate Law: COMpartment*k4*phi_I
k3 = 0.002 0.001*m³; k2 = 0.693 1/(0.0115741*ms); k1 = 0.05 1Reaction: => phi_I; phi_R, Rate Law: COMpartment*k1*k2*phi_I*(1-k3*(phi_I+phi_R))
alpha = 0.8 1Reaction: => phi_I; K_T, Rate Law: COMpartment*alpha*K_T
k3 = 0.002 0.001*m³; d1 = 0.2 1/(0.0115741*ms); k2 = 0.693 1/(0.0115741*ms); alpha = 0.8 1; k1 = 0.05 1Reaction: phi_R = ((1-alpha)*K_T+k1*k2*phi_R*(1-k3*(phi_I+phi_R)))-d1*phi_R, Rate Law: ((1-alpha)*K_T+k1*k2*phi_R*(1-k3*(phi_I+phi_R)))-d1*phi_R
tau4 = 1.75 1/(0.0115741*m³*s); tau2 = 21.94 0.0864*mm³/(g²*s); tau1 = -2.47 8.64e-11*m^6/(g³*s); tau3 = 6.41 1/(11.5741*Mg*s)Reaction: K_T = tau1*T^3+tau2*T^2+tau3*T+tau4, Rate Law: missing
k4 = 0.07 0.0864*µg/s; d2 = 9.1 1/(0.0115741*ms)Reaction: T = k4*phi_I-d2*T, Rate Law: k4*phi_I-d2*T

States:

NameDescription
T[TGF-beta]
phi I[macrophage; inflammatory macrophage]
phi R[macrophage; Wound Repair]
K T[monocyte; monocyte migration into blood stream; monocyte]

Waugh2006 - Diabetic Wound Healing - TGF-B Dynamics: BIOMD0000000680v0.0.1

This a model from the article: Macrophage dynamics in diabetic wound dealing. Waugh HV, Sherratt JA. Bull Math Biol…

Details

Wound healing in diabetes is a complex process, characterised by a chronic inflammation phase. The exact mechanism by which this occurs is not fully understood, and whilst several treatments for healing diabetic wounds exist, very little research has been conducted towards the causes of the extended inflammation phase. We describe a mathematical model which offers a possible explanation for diabetic wound healing in terms of the distribution of macrophage phenotypes being altered in the diabetic patient compared to normal wound repair. As a consequence of this, we put forward a suggestion for treatment based on rectifying the macrophage phenotype imbalance. link: http://identifiers.org/pubmed/16794927

Parameters:

NameDescription
k3 = 0.002 0.001*m³; d1 = 0.2 1/(0.0115741*ms); k2 = 0.693 1/(0.0115741*ms); alpha = 0.8 1; k1 = 0.05 1Reaction: phi_I = (alpha*K_T+k1*k2*phi_I*(1-k3*(phi_I+phi_R)))-d1*phi_I, Rate Law: (alpha*K_T+k1*k2*phi_I*(1-k3*(phi_I+phi_R)))-d1*phi_I
tau4 = 1.75 1/(0.0115741*m³*s); tau2 = 21.94 0.0864*mm³/(g²*s); tau1 = -2.47 8.64e-11*m^6/(g³*s); tau3 = 6.41 1/(11.5741*Mg*s)Reaction: K_T = tau1*T^3+tau2*T^2+tau3*T+tau4, Rate Law: missing
k4 = 0.07 0.0864*µg/s; d2 = 9.1 1/(0.0115741*ms)Reaction: T = k4*phi_I-d2*T, Rate Law: k4*phi_I-d2*T

States:

NameDescription
T[TGF-beta]
phi I[inflammatory macrophage; macrophage]
phi R[macrophage; Wound Repair]
K T[monocyte; monocyte migration into blood stream; monocyte]

Waugh2006 - Diabetic Wound Healing - Treated and Untreated Macrophage Dynamics: BIOMD0000000681v0.0.1

This a model from the article: Macrophage dynamics in diabetic wound dealing. Waugh HV, Sherratt JA. Bull Math Biol…

Details

Wound healing in diabetes is a complex process, characterised by a chronic inflammation phase. The exact mechanism by which this occurs is not fully understood, and whilst several treatments for healing diabetic wounds exist, very little research has been conducted towards the causes of the extended inflammation phase. We describe a mathematical model which offers a possible explanation for diabetic wound healing in terms of the distribution of macrophage phenotypes being altered in the diabetic patient compared to normal wound repair. As a consequence of this, we put forward a suggestion for treatment based on rectifying the macrophage phenotype imbalance. link: http://identifiers.org/pubmed/16794927

Parameters:

NameDescription
k3 = 0.002 0.001*m³; d1 = 0.2 1/(0.0115741*ms); k2 = 0.693 1/(0.0115741*ms); alpha = 0.8 1; k1 = 0.05 1Reaction: phi_R = ((1-alpha)*K_T+k1*k2*phi_R*(1-k3*(phi_I+phi_R)))-d1*phi_R, Rate Law: ((1-alpha)*K_T+k1*k2*phi_R*(1-k3*(phi_I+phi_R)))-d1*phi_R
tau4 = 1.75 1/(0.0115741*m³*s); tau2 = 21.94 0.0864*mm³/(g²*s); tau1 = -2.47 8.64e-11*m^6/(g³*s); tau3 = 6.41 1/(11.5741*Mg*s)Reaction: K_T = tau1*T^3+tau2*T^2+tau3*T+tau4, Rate Law: missing
k4 = 0.07 0.0864*µg/s; d2 = 9.1 1/(0.0115741*ms)Reaction: T = k4*phi_I-d2*T, Rate Law: k4*phi_I-d2*T

States:

NameDescription
T[TGF-beta]
phi I[macrophage; inflammatory macrophage]
phi R[macrophage; Wound Repair]
K T[monocyte; monocyte migration into blood stream; monocyte]

Waugh2006_WoundHealing_Diabetic_ModelA: MODEL1006230106v0.0.1

This a model from the article: Modeling the effects of treating diabetic wounds with engineered skin substitutes. Wa…

Details

In this paper, a novel mathematical model of wound healing in both normal and diabetic cases is presented, focusing upon the effects of adding two currently available commercial engineered skin substitute therapies to the wound (Apligraf) and Dermagraft). Our work extends a previously developed model, which considers inflammatory and repair macrophage dynamics in normal and diabetic wound healing. Here, we extend the model to include equations for platelet-derived growth factor concentration, fibroblast density, collagen density, and hyaluronan concentration. This enables us to examine the variation of these components in both normal and diabetic wound healing cases, and to model the treatment protocols of these therapies. Within the context of our model, we find that the key component to successful healing in diabetic wounds is hyaluronan and that the therapies work by increasing the amount of hyaluronan available in the wound environment. The time-to-healing results correlate with those observed in clinical trials and the model goes some way to establishing an understanding of why diabetic wounds do not heal, and how these treatments affect the diabetic wound environment to promote wound closure. link: http://identifiers.org/pubmed/17650100

Waugh2006_WoundHealing_Diabetic_ModelB: MODEL1006230003v0.0.1

This a model from the article: Modeling the effects of treating diabetic wounds with engineered skin substitutes. Wa…

Details

In this paper, a novel mathematical model of wound healing in both normal and diabetic cases is presented, focusing upon the effects of adding two currently available commercial engineered skin substitute therapies to the wound (Apligraf) and Dermagraft). Our work extends a previously developed model, which considers inflammatory and repair macrophage dynamics in normal and diabetic wound healing. Here, we extend the model to include equations for platelet-derived growth factor concentration, fibroblast density, collagen density, and hyaluronan concentration. This enables us to examine the variation of these components in both normal and diabetic wound healing cases, and to model the treatment protocols of these therapies. Within the context of our model, we find that the key component to successful healing in diabetic wounds is hyaluronan and that the therapies work by increasing the amount of hyaluronan available in the wound environment. The time-to-healing results correlate with those observed in clinical trials and the model goes some way to establishing an understanding of why diabetic wounds do not heal, and how these treatments affect the diabetic wound environment to promote wound closure. link: http://identifiers.org/pubmed/17650100

Waugh2006_WoundHealing_Diabetic_ModelC: MODEL1006230099v0.0.1

This a model from the article: Modeling the effects of treating diabetic wounds with engineered skin substitutes. Wa…

Details

In this paper, a novel mathematical model of wound healing in both normal and diabetic cases is presented, focusing upon the effects of adding two currently available commercial engineered skin substitute therapies to the wound (Apligraf) and Dermagraft). Our work extends a previously developed model, which considers inflammatory and repair macrophage dynamics in normal and diabetic wound healing. Here, we extend the model to include equations for platelet-derived growth factor concentration, fibroblast density, collagen density, and hyaluronan concentration. This enables us to examine the variation of these components in both normal and diabetic wound healing cases, and to model the treatment protocols of these therapies. Within the context of our model, we find that the key component to successful healing in diabetic wounds is hyaluronan and that the therapies work by increasing the amount of hyaluronan available in the wound environment. The time-to-healing results correlate with those observed in clinical trials and the model goes some way to establishing an understanding of why diabetic wounds do not heal, and how these treatments affect the diabetic wound environment to promote wound closure. link: http://identifiers.org/pubmed/17650100

Waugh2006_WoundHealing_Diabetic_ModelD: MODEL1006230102v0.0.1

This a model from the article: Modeling the effects of treating diabetic wounds with engineered skin substitutes. Wa…

Details

In this paper, a novel mathematical model of wound healing in both normal and diabetic cases is presented, focusing upon the effects of adding two currently available commercial engineered skin substitute therapies to the wound (Apligraf) and Dermagraft). Our work extends a previously developed model, which considers inflammatory and repair macrophage dynamics in normal and diabetic wound healing. Here, we extend the model to include equations for platelet-derived growth factor concentration, fibroblast density, collagen density, and hyaluronan concentration. This enables us to examine the variation of these components in both normal and diabetic wound healing cases, and to model the treatment protocols of these therapies. Within the context of our model, we find that the key component to successful healing in diabetic wounds is hyaluronan and that the therapies work by increasing the amount of hyaluronan available in the wound environment. The time-to-healing results correlate with those observed in clinical trials and the model goes some way to establishing an understanding of why diabetic wounds do not heal, and how these treatments affect the diabetic wound environment to promote wound closure. link: http://identifiers.org/pubmed/17650100

Webb2002 - Fas/FasL mediated tumor T-cell interaction: BIOMD0000000661v0.0.1

Webb2002 - Fas/FasL mediated tumor T-cell interactionThis deterministic model of immunological surveillance involving tu…

Details

One proposed mechanism of tumour escape from immune surveillance is tumour up-regulation of the cell surface ligand FasL, which can lead to apoptosis of Fas receptor (Fas) positive lymphocytes. Based upon this 'counterattack', we have developed a mathematical model involving tumour cell-lymphocyte interaction, cell surface expression of Fas/FasL, and their secreted soluble forms. The model predicts that (a) the production of soluble forms of Fas and FasL will lead to the down-regulation of the immune response; (b) matrix metalloproteinase (MMP) inactivation should lead to increased membrane FasL and result in a higher rate of Fas-mediated apoptosis for lymphocytes than for tumour cells. Recent studies on cancer patients lend support for these predictions. The clinical implications are two-fold. Firstly, the use of broad spectrum MMP inhibitors as anti-angiogenic agents may be compromised by their adverse effect on tumour FasL up-regulation. Also, Fas/FasL interactions may have an impact on the outcome of numerous ongoing immunotherapeutic trials since the final common pathway of all these approaches is the transduction of death signals within the tumour cell. link: http://identifiers.org/pubmed/12208611

Parameters:

NameDescription
k2 = 0.006Reaction: => LT; T, m, Rate Law: compartment*k2*T*m
k3 = 5.9413Reaction: => SL; Lm, m, LT, T, Rate Law: k3*(Lm*m+LT*T)
k9 = 8.73E9Reaction: SL => ; Rm, m, RT, T, Rate Law: k9*(Rm*m+RT*T)*SL
k6 = 2244.0Reaction: => Rm, Rate Law: Tumour_cell*k6
k10 = 3110.0Reaction: => Lm, Rate Law: Tumour_cell*k10
k5 = 2.52E-9Reaction: LT => ; m, T, Rm, Rate Law: k5*m*T*LT*Rm
k1 = 8.38E-10Reaction: T => ; m, Lm, RT, Rate Law: k1*m*T*Lm*RT
k4 = 0.35; k3 = 5.9413Reaction: Lm =>, Rate Law: Tumour_cell*(k3+k4)*Lm
k7 = 0.35Reaction: RT =>, Rate Law: T_Lymphocyte_cell*k7*RT
k8 = 1.92E10Reaction: Rm => ; SL, Rate Law: k8*Rm*SL
k11 = 13.9Reaction: SL =>, Rate Law: compartment*k11*SL

States:

NameDescription
RT[Tumor necrosis factor receptor superfamily member 6; Cellular Membrane]
T[Effector T-Lymphocyte]
Rm[Tumor necrosis factor receptor superfamily member 6; Cellular Membrane]
LT[glycoprotein; Cellular Membrane; Tumor necrosis factor ligand superfamily member 6]
m[Neoplastic Cell]
SL[tumor necrosis factor ligand superfamily member 6 isoform FasL soluble form]
Lm[glycoprotein; Cellular Membrane; Tumor necrosis factor ligand superfamily member 6]

Wegner2012_TGFbetaSignalling_FeedbackLoops: BIOMD0000000410v0.0.1

This model is from the article: Dynamics and feedback loops in the transforming growth factor β signaling pathway. W…

Details

Transforming growth factor β (TGF-β) ligands activate a signaling cascade with multiple cell context dependent outcomes. Disruption or disturbance leads to variant clinical disorders. To develop strategies for disease intervention, delineation of the pathway in further detail is required. Current theoretical models of this pathway describe production and degradation of signal mediating proteins and signal transduction from the cell surface into the nucleus, whereas feedback loops have not exhaustively been included. In this study we present a mathematical model to determine the relevance of feedback regulators (Arkadia, Smad7, Smurf1, Smurf2, SnoN and Ski) on TGF-β target gene expression and the potential to initiate stable oscillations within a realistic parameter space. We employed massive sampling of the parameters space to pinpoint crucial players for potential oscillations as well as transcriptional product levels. We identified Smad7 and Smurf2 with the highest impact on the dynamics. Based on these findings, we conducted preliminary time course experiments. link: http://identifiers.org/pubmed/22284904

Parameters:

NameDescription
k1=0.1; k2=0.01Reaction: _25 => _174, Rate Law: k1*_25-k2*_174
v=0.00146Reaction: => _75, Rate Law: _1*v
k1=0.05Reaction: species_1 => ; _174, Rate Law: _1*k1*species_1*(1+_174)
k1=0.1; k2=0.2Reaction: species_2 => _11, Rate Law: k1*species_2-k2*_11
k=1000.0; km=0.0318Reaction: _99 => _129; _96, Rate Law: _1*k*_96*_99/(km+_99)
k2=0.01; k1=8.69Reaction: _174 + _96 => _198, Rate Law: _1*(k1*_174*_96-k2*_198)
k1=2.9; k2=0.2Reaction: _15 + _25 => _27, Rate Law: _3*(k1*_15*_25-k2*_27)
Km=0.53; V=3.51Reaction: _19 => _21, Rate Law: _3*V*_19/(Km+_19)
k1=0.2; k2=0.2Reaction: _13 + species_28 => species_29, Rate Law: _3*(k1*_13*species_28-k2*species_29)
k1=1.0; k2=0.01Reaction: _27 => _181, Rate Law: k1*_27-k2*_181
k1=0.1; k2=0.1Reaction: _99 + species_7 => species_12, Rate Law: _1*(k1*_99*species_7-k2*species_12)
k1=1900.0Reaction: _181 + _96 =>, Rate Law: _1*k1*_181*_96
k2=0.336; k1=0.156Reaction: _99 => _21, Rate Law: k1*_99-k2*_21
k1=0.027778Reaction: _96 =>, Rate Law: _1*k1*_96
v=0.01183Reaction: => _147, Rate Law: _1*v
V=2.34; Km=40.0Reaction: _9 => species_19, Rate Law: _3*V*_9/(Km+_9)
k1=0.065Reaction: _101 =>, Rate Law: _1*k1*_101
k2=1.8056; k1=0.2333Reaction: _15 => species_1, Rate Law: k1*_15-k2*species_1
k1=255.068Reaction: _5 + _19 => _9, Rate Law: _3*k1*_5*_19^2
v=0.0156Reaction: => _99, Rate Law: _1*v
k1=0.1; k=1.0E-4Reaction: => _174; species_30, species_16, species_23, Rate Law: _1*(k+k1*species_30)/(1+species_16+species_23)
k2=0.156; k1=0.156Reaction: _147 => _5, Rate Law: k1*_147-k2*_5
k2=0.05288; k1=1.0Reaction: _5 + _11 => species_16, Rate Law: _3*(k1*_5*_11-k2*species_16)
k1=1.0; k2=1.0Reaction: species_20 + _11 => species_14, Rate Law: k1*species_20^3*_11^3-k2*species_14
V=0.53; Km=3.51Reaction: _129 => _99, Rate Law: _1*V*_129/(Km+_129)
k2=1.6; k1=1.6Reaction: _9 + species_22 => species_11, Rate Law: k1*_9*species_22^2-k2*species_11
k1=0.031; k=1.0E-4Reaction: => _101; species_30, Rate Law: _1*(k+k1*species_30)
k1=0.16Reaction: _153 => _9, Rate Law: k1*_153
k1=0.0492Reaction: species_27 => _5 + species_17, Rate Law: _3*k1*species_27
k1=0.2Reaction: _21 + _15 =>, Rate Law: _3*k1*_21*_15
k1=0.232Reaction: _15 + species_13 => _19, Rate Law: k1*_15^3*species_13
k1=0.0285; k=2.28E-4Reaction: => species_1; species_30, Rate Law: _1*(k+k1*species_30)
k2=0.102; k1=0.463Reaction: _9 + species_28 => species_30, Rate Law: _3*(k1*_9*species_28-k2*species_30)
k1=0.1Reaction: _174 => ; species_15, Rate Law: _1*k1*_174*(1+species_15)
k1=1.0; k2=0.1Reaction: _101 + species_3 => species_5, Rate Law: _1*(k1*_101*species_3-k2*species_5)
k=2.0E-5; k1=5.5E-4Reaction: => species_2; species_30, Rate Law: _1*(k+k1*species_30)
k1=16.6Reaction: _129 => _19, Rate Law: k1*_129
k1=0.1266Reaction: _147 =>, Rate Law: _1*k1*_147
k1=0.03333; k2=0.03333Reaction: _79 + _84 => _96, Rate Law: _1*(k1*_79*_84^2-k2*_96)
k1=1000.0Reaction: _147 + _129 => _153, Rate Law: _1*k1*_147*_129^2
k=3.51; km=0.53Reaction: _105 => _129 + _101; _96, Rate Law: _1*k*_96*_105/(km+_105)

States:

NameDescription
153[Phosphoprotein; Mothers against decapentaplegic homolog 4; Mothers against decapentaplegic homolog 2]
species 27[Smad3 protein; Mothers against decapentaplegic homolog 4]
101[Smad anchor for receptor activation]
species 1[E3 ubiquitin-protein ligase SMURF2]
25[Mothers against decapentaplegic homolog 7]
species 28freePromoters
species 16[SnoN protein; Mothers against decapentaplegic homolog 4]
11[SnoN protein]
84[TGF-beta receptor type-1]
13[Phosphoprotein; SnoN protein; Mothers against decapentaplegic homolog 4; Mothers against decapentaplegic homolog 2]
15[E3 ubiquitin-protein ligase SMURF2]
75[TGF-beta receptor type-2]
27[E3 ubiquitin-protein ligase SMURF2; Mothers against decapentaplegic homolog 7]
species 21[Phosphoprotein; SnoN protein; Mothers against decapentaplegic homolog 4]
species 17[Smad3 protein]
species 25[E3 ubiquitin-protein ligase SMURF2; Mothers against decapentaplegic homolog 7]
species 29inactivePromoters
species 30[CCO:U0000003]
species 2[SnoN protein]
174[Mothers against decapentaplegic homolog 7]
21[Mothers against decapentaplegic homolog 2]
129[Mothers against decapentaplegic homolog 2; Phosphoprotein]
96[reactive oxygen species]
147[Mothers against decapentaplegic homolog 4]
181[E3 ubiquitin-protein ligase SMURF2; Mothers against decapentaplegic homolog 7]
species 24[E3 ubiquitin-protein ligase SMURF2]
species 22[Ski oncogene]
198[Transforming growth factor beta-1; TGF-beta receptor type-1; TGF-beta receptor type-2; Mothers against decapentaplegic homolog 7]
19[Phosphoprotein; Mothers against decapentaplegic homolog 2]
99[Mothers against decapentaplegic homolog 2]
species 7[Ski oncogene]
9[Phosphoprotein; Mothers against decapentaplegic homolog 4; Mothers against decapentaplegic homolog 2]
species 26[Phosphoprotein; Ski oncogene; Smad3 protein; Mothers against decapentaplegic homolog 4]
105[Smad anchor for receptor activation; Mothers against decapentaplegic homolog 2]

Wei2011_MLCactivationPathway_EndothelialPermeability: MODEL1102210000v0.0.1

This model is from the article: An Integrated Mathematical Model of Thrombin-, Histamine-and VEGF-Mediated Signalling…

Details

BACKGROUND: Endothelial permeability is involved in injury, inflammation, diabetes and cancer. It is partly regulated by the thrombin-, histamine-, and VEGF-mediated myosin-light-chain (MLC) activation pathways. While these pathways have been investigated, questions such as temporal effects and the dynamics of multi-mediator regulation remain to be fully studied. Mathematical modeling of these pathways facilitates such studies. Based on the published ordinary differential equation models of the pathway components, we developed an integrated model of thrombin-, histamine-, and VEGF-mediated MLC activation pathways. RESULTS: Our model was validated against experimental data for calcium release and thrombin-, histamine-, and VEGF-mediated MLC activation. The simulated effects of PAR-1, Rho GTPase, ROCK, VEGF and VEGFR2 over-expression on MLC activation, and the collective modulation by thrombin and histamine are consistent with experimental findings. Our model was used to predict enhanced MLC activation by CPI-17 over-expression and by synergistic action of thrombin and VEGF at low mediator levels. These may have impact in endothelial permeability and metastasis in cancer patients with blood coagulation. CONCLUSION: Our model was validated against a number of experimental findings and the observed synergistic effects of low concentrations of thrombin and histamine in mediating the activation of MLC. It can be used to predict the effects of altered pathway components, collective actions of multiple mediators and the potential impact to various diseases. Similar to the published models of other pathways, our model can potentially be used to identify important disease genes through sensitivity analysis of signalling components. link: http://identifiers.org/pubmed/21756365

Wei2017 - tumor, T cell and cytokine interaction: BIOMD0000000778v0.0.1

The paper describes a model of tumor-immune interaction. Created by COPASI 4.25 (Build 207) This model is describe…

Details

Immunotherapy is one of the most recent approaches for controlling and curing malignant tumors. In this paper, we consider a mathematical model of periodically pulsed immunotherapy using CD4+ T cells and an antitumor cytokine. Mathematical analyses are performed to determine the threshold of a successful treatment. The interindividual variability is explored by one-, two-, and three-parameter bifurcation diagrams for a nontreatment case. Numerical simulation conducted in this paper shows that (i) the tumor can be regulated by administering CD4+ T cells alone in a patient with a strong immune system or who has been diagnosed at an early stage, (ii) immunotherapy with a large amount of an antitumor cytokine can boost the immune system to remit or even to suppress tumor cells completely, and (iii) through polytherapy the tumor can be kept at a smaller size with reduced dosages. link: http://identifiers.org/pubmed/29250133

Parameters:

NameDescription
d = 0.1 1/d; m = 50.0 1Reaction: T => ; C, Rate Law: tme*d*T*C/(m+T)
l2 = 0.0 1/dReaction: => C, Rate Law: tme*l2
r = 0.01 1/d; K = 1000.0 1Reaction: => T, Rate Law: tme*r*T*(1-T/K)
k = 1000.0 1; beta = 0.1 1/dReaction: => I; T, Rate Law: tme*beta*T*I/(k+T)
u = 50.0 1/dReaction: C =>, Rate Law: tme*u*C
b = 100.0 1; alpha = 0.01 1/dReaction: => C; T, I, Rate Law: tme*alpha*T*I/(b+T)
a = 0.03 1/dReaction: I =>, Rate Law: tme*a*I
l1 = 0.0 1/dReaction: => I, Rate Law: tme*l1

States:

NameDescription
I[CD4-positive helper T cell]
T[malignant cell]
C[Cytokine]

Wei2019 - Mathematical modeling of tumor growth The MCF-7 breast cancer cell line: MODEL1909090002v0.0.1

This is a mathematical model describing MCF-7 cancer cell growth with interaction between tumor cells, estradiol (hormon…

Details

Breast cancer is the second most commonly diagnosed cancer in women worldwide. MCF-7 cell line is an extensively studied human breast cancer cell line. This cell line expresses estrogen receptors, and the growth of MCF-7 cells is hormone dependent. In this study, a mathematical model, which governs MCF-7 cell growth with interaction among tumor cells, estradiol, natural killer (NK) cells, cytotoxic T lymphocytes (CTLs) or CD8+ T cells, and white blood cells (WBCs), is proposed. Experimental data are used to determine functional forms and parameter values. Breast tumor growth is then studied using the mathematical model. The results obtained from numerical simulation are compared with those from clinical and experimental studies. The system has three coexisting stable equilibria representing the tumor free state, a microscopic tumor, and a large tumor. Numerical simulation shows that an immune system is able to eliminate or control a tumor with a restricted initial size. A healthy immune system is able to effectively eliminate a small tumor or produces long-term dormancy. An immune system with WBC count at the low parts of the normal ranges or with temporary low NK cell count is able to eliminate a smaller tumor. The cytotoxicity of CTLs plays an important role in immune surveillance. The association between the circulating estradiol level and cancer risk is not significant. link: http://identifiers.org/doi/10.3934/mbe.2019325

Weimann2004_CircadianOscillator: BIOMD0000000170v0.0.1

The model reproduces the time profile of the species as depicted in Fig 3A of the paper. Model successfully tested on Ma…

Details

The suprachiasmatic nucleus governs daily variations of physiology and behavior in mammals. Within single neurons, interlocked transcriptional/translational feedback loops generate circadian rhythms on the molecular level. We present a mathematical model that reflects the essential features of the mammalian circadian oscillator to characterize the differential roles of negative and positive feedback loops. The oscillations that are obtained have a 24-h period and are robust toward parameter variations even when the positive feedback is replaced by a constantly expressed activator. This demonstrates the crucial role of the negative feedback for rhythm generation. Moreover, it explains the rhythmic phenotype of Rev-erbalpha-/- mutant mice, where a positive feedback is missing. The interplay of negative and positive feedback reveals a complex dynamics. In particular, the model explains the unexpected rescue of circadian oscillations in Per2Brdm1/Cry2-/- double-mutant mice (Per2Brdm1 single-mutant mice are arrhythmic). Here, a decrease of positive feedback strength associated with mutating the Per2 gene is compensated by the Cry2-/- mutation that simultaneously decreases the negative feedback strength. Finally, this model leads us to a testable prediction of a molecular and behavioral phenotype: circadian oscillations should be rescued when arrhythmic Per2Brdm1 mutant mice are crossed with Rev- erbalpha -/- mutant mice. link: http://identifiers.org/pubmed/15347590

Parameters:

NameDescription
k2d = 0.05 hr_invReaction: y2 =>, Rate Law: Cytoplasm*k2d*y2
trans_per2_cry = 0.0 nM_per_hourReaction: => y1, Rate Law: Cytoplasm*trans_per2_cry
k2t = 0.24 hr_invReaction: y2 => y3, Rate Law: Cytoplasm*k2t*y2
k6t = 0.06 hr_invReaction: y6 => y5, Rate Law: Nucleus*k6t*y6
k6d = 0.12 hr_invReaction: y6 =>, Rate Law: Nucleus*k6d*y6
k5t = 0.45 hr_invReaction: y5 => y6, Rate Law: Cytoplasm*k5t*y5
k3d = 0.12 hr_invReaction: y3 =>, Rate Law: Nucleus*k3d*y3
k6a = 0.09 hr_invReaction: y6 => y7, Rate Law: Nucleus*k6a*y6
k5b = 0.24 hr_invReaction: => y5; y4, Rate Law: Cytoplasm*k5b*y4
trans_Bmal1 = 0.0 nM_per_hourReaction: => y4, Rate Law: Cytoplasm*trans_Bmal1
k5d = 0.06 hr_invReaction: y5 =>, Rate Law: Cytoplasm*k5d*y5
k7a = 0.003 hr_invReaction: y7 => y6, Rate Law: Nucleus*k7a*y7
k4d = 0.75 hr_invReaction: y4 =>, Rate Law: Cytoplasm*k4d*y4
k3t = 0.02 hr_invReaction: y3 => y2, Rate Law: Nucleus*k3t*y3
q = 2.0 dimensionless; k2b = 0.3 nM_inv_hr_invReaction: => y2; y1, Rate Law: Cytoplasm*k2b*y1^q
k7d = 0.09 hr_invReaction: y7 =>, Rate Law: Nucleus*k7d*y7
k1d = 0.12 hr_invReaction: y1 =>, Rate Law: Cytoplasm*k1d*y1

States:

NameDescription
y3[Period circadian protein homolog 2; Cryptochrome-1]
y1[Period circadian protein homolog 2; Cryptochrome-1; messenger RNA; RNA]
y4[messenger RNA; RNA; Aryl hydrocarbon receptor nuclear translocator-like protein 1]
y7[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
y2[Period circadian protein homolog 2; Cryptochrome-1]
y6[Aryl hydrocarbon receptor nuclear translocator-like protein 1]
y5[Aryl hydrocarbon receptor nuclear translocator-like protein 1]

Weinstein2000_OMCD: MODEL1006230037v0.0.1

This a model from the article: A mathematical model of the outer medullary collecting duct of the rat. Weinstein AM.…

Details

A mathematical model of the outer medullary collecting duct (OMCD) has been developed, consisting of alpha-intercalated cells and a paracellular pathway, and which includes Na(+), K(+), Cl(-), HCO(3)(-), CO(2), H(2)CO(3), phosphate, ammonia, and urea. Proton secretion across the luminal cell membrane is mediated by both H(+)-ATPase and H-K-ATPase, with fluxes through the H-K-ATPase given by a previously developed kinetic model (Weinstein AM. Am J Physiol Renal Physiol 274: F856-F867, 1998). The flux across each ATPase is substantial, and variation in abundance of either pump can be used to control OMCD proton secretion. In comparison with the H(+)-ATPase, flux through the H-K-ATPase is relatively insensitive to changes in lumen pH, so as luminal acidification proceeds, proton secretion shifts toward this pathway. Peritubular HCO(3)(-) exit is via a conductive pathway and via the Cl(-)/HCO(3)(-) exchanger, AE1. To represent AE1, a kinetic model has been developed based on transport studies obtained at 38 degrees C in red blood cells. (Gasbjerg PK, Knauf PA, and Brahm J. J Gen Physiol 108: 565-575, 1996; Knauf PA, Gasbjerg PK, and Brahm J. J Gen Physiol 108: 577-589, 1996). Model calculations indicate that if all of the chloride entry via AE1 recycles across a peritubular chloride channel and if this channel is anything other than highly selective for chloride, then it should conduct a substantial fraction of the bicarbonate exit. Since both luminal membrane proton pumps are sensitive to small changes in cytosolic pH, variation in density of either AE1 or peritubular anion conductance can modulate OMCD proton secretory rate. With respect to the OMCD in situ, available buffer is predicted to be abundant, including delivered HCO(3)(-) and HPO(4)(2-), as well as peritubular NH(3). Thus, buffer availability is unlikely to exert a regulatory role in total proton secretion by this tubule segment. link: http://identifiers.org/pubmed/10894785

Weis2014 - Data driven Mammalian Cell Cycle Model: BIOMD0000000723v0.0.1

This a model from the article: A Data-Driven, Mathematical Model of Mammalian Cell Cycle Regulation. Michael C. Weis…

Details

Few of >150 published cell cycle modeling efforts use significant levels of data for tuning and validation. This reflects the difficultly to generate correlated quantitative data, and it points out a critical uncertainty in modeling efforts. To develop a data-driven model of cell cycle regulation, we used contiguous, dynamic measurements over two time scales (minutes and hours) calculated from static multiparametric cytometry data. The approach provided expression profiles of cyclin A2, cyclin B1, and phospho-S10-histone H3. The model was built by integrating and modifying two previously published models such that the model outputs for cyclins A and B fit cyclin expression measurements and the activation of B cyclin/Cdk1 coincided with phosphorylation of histone H3. The model depends on Cdh1-regulated cyclin degradation during G1, regulation of B cyclin/Cdk1 activity by cyclin A/Cdk via Wee1, and transcriptional control of the mitotic cyclins that reflects some of the current literature. We introduced autocatalytic transcription of E2F, E2F regulated transcription of cyclin B, Cdc20/Cdh1 mediated E2F degradation, enhanced transcription of mitotic cyclins during late S/early G2 phase, and the sustained synthesis of cyclin B during mitosis. These features produced a model with good correlation between state variable output and real measurements. Since the method of data generation is extensible, this model can be continually modified based on new correlated, quantitative data. link: http://identifiers.org/pubmed/24824602

Parameters:

NameDescription
v50 = 2622.5416344645Reaction: pE2F + Rb => pE2FRB, Rate Law: cell*v50
v30 = 176.595160106273Reaction: pE2FRB => ppRB + pE2F, Rate Law: cell*v30
v44 = 4495.61597904523Reaction: ppRB => Rb, Rate Law: cell*v44
a1frac = 0.081283; kdia = 196.0783Reaction: => actCycACdk1; TriA, Rate Law: cell*a1frac*TriA*kdia
v43 = 35.2584572674058Reaction: Rb => ppRB, Rate Law: cell*v43
v51 = 1.89195334Reaction: pE2FRB => E2FRB, Rate Law: cell*v51
ka20 = 292.669; Ja20 = 0.005Reaction: => Cdc20A; APCP, Cdc20T, Rate Law: cell*ka20*APCP*(Cdc20T-Cdc20A)/((Ja20+Cdc20T)-Cdc20A)
kde2fcdc20 = 881.75Reaction: E2F => ; E2F, Cdc20A, Rate Law: cell*kde2fcdc20*E2F*Cdc20A
k10 = 88.175Reaction: actCycD =>, Rate Law: cell*k10*actCycD
k24 = 1763.5; freeCK1 = 0.55403292Reaction: actCycD => TriD; actCycD, Rate Law: cell*k24*actCycD*freeCK1
kdia = 196.0783Reaction: TriA =>, Rate Law: cell*kdia*TriA
ke2f = 4.2324Reaction: => E2F; mass, E2F, Rate Law: cell*ke2f*mass*E2F
kd20 = 17.635Reaction: Cdc20A =>, Rate Law: cell*kd20*Cdc20A
k17p = 2.64525Reaction: => DRG; ERG, Rate Law: cell*k17p*ERG
v45 = 9.7440429Reaction: E2FRB => E2F + Rb, Rate Law: cell*v45
TriE = 0.026647; Vdi = 306.292436560171Reaction: => actCycE, Rate Law: cell*Vdi*TriE
Vwee = 17866.3670696676Reaction: actCycB =>, Rate Law: cell*Vwee*actCycB
kasa = 19733.57; freeCK1 = 0.55403292Reaction: actCycACdk1 => ; actCycACdk1, Rate Law: cell*kasa*actCycACdk1*freeCK1
Vdb = 282.537036Reaction: actCycB =>, Rate Law: cell*Vdb*actCycB
v47 = 4.897369999Reaction: pE2F => E2F, Rate Law: cell*v47
a1frac = 0.081283; Vdi = 306.292436560171Reaction: => actCycACdk1; TriA, Rate Law: cell*a1frac*TriA*Vdi
v48 = 1352.4191649675Reaction: E2F + Rb => E2FRB, Rate Law: cell*v48
v49 = 18.9195334Reaction: pE2FRB => pE2F + Rb, Rate Law: cell*v49
Vsb = 7.16092423585105; k2=2.0Reaction: => actCycB + cycB; mass, Rate Law: cell*Vsb*mass*k2
JaAPC = 0.01; kaAPC = 2.33401Reaction: => APCP; actCycB, Rate Law: cell*kaAPC*actCycB*(1-APCP)/((JaAPC+1)-APCP)
ksep = 1.562461; k2=2.0Reaction: => actCycE + cycE; mass, Rate Law: cell*ksep*mass*k2
k15 = 5.2905; J15 = 0.1Reaction: => ERG; DRG, Rate Law: cell*k15/(1+(DRG/J15)^2)
J20 = 100.0; ka20 = 292.669Reaction: => Cdc20T; actCycB, Rate Law: cell*ka20*actCycB/(J20+actCycB)
u = 0.693937Reaction: => mass; mass, Rate Law: cell*u*mass
a1frac = 0.081283; Vdi = 306.292436560171; kdia = 196.0783Reaction: => actCycACdk2; TriA, Rate Law: cell*(1-a1frac)*(Vdi+kdia)*TriA
k18 = 176.35Reaction: DRG =>, Rate Law: cell*k18*DRG
ki20 = 17.635; Ji20 = 0.005Reaction: Cdc20A =>, Rate Law: cell*ki20*Cdc20A/(Ji20+Cdc20A)
v29 = 90.9510176402072Reaction: E2FRB => ppRB + E2F, Rate Law: cell*v29
V25 = 63.8899881606308Reaction: => actCycB; cycB, Rate Law: cell*V25*cycB
a1frac = 0.081283; ksappp = 20.28025; TFAB = 1.39800750094916E-6; ksap = 16.75325; ksapp = 0.10581Reaction: => actCycACdk1; E2F, mass, Rate Law: cell*a1frac*(ksap+ksapp*E2F+ksappp*TFAB)*mass*2
Vda = 985.702423Reaction: actCycACdk1 =>, Rate Law: cell*Vda*actCycACdk1
k9 = 45.851Reaction: => actCycD; DRG, Rate Law: cell*k9*DRG
ksepp = 8.8175; k2=2.0Reaction: => actCycE + cycE; E2F, mass, Rate Law: cell*ksepp*E2F*mass*k2
v52 = 0.00715073488283791Reaction: E2FRB => pE2FRB, Rate Law: cell*v52
Vdi = 306.292436560171Reaction: TriD => actCycD; TriD, Rate Law: cell*Vdi*TriD
k17 = 2645.25; J17 = 0.3Reaction: => DRG, Rate Law: cell*k17*(DRG/J17)^2/(1+(DRG/J17)^2)
kdie = 196.0783; TriE = 0.026647Reaction: => actCycE, Rate Law: cell*kdie*TriE
Vsi = 390.9926Reaction: => CKI, Rate Law: cell*Vsi
kde2fcdh1 = 1.7635Reaction: E2F => ; Cdh1, E2F, Rate Law: cell*kde2fcdh1*Cdh1*E2F
k16 = 44.0875Reaction: ERG =>, Rate Law: cell*k16*ERG
Jih1 = 0.01; kih1pp = 17635.0; kih1ppp = 1763.5Reaction: Cdh1 => ; actCycACdk1, actCycACdk2, actCycB, Rate Law: cell*(kih1pp*(actCycACdk1+actCycACdk2)+kih1ppp*actCycB)*Cdh1/(Jih1+Cdh1)
Vde = 45.5696612502715Reaction: actCycE =>, Rate Law: cell*Vde*actCycE
kah1p = 155.8708; Jah1 = 0.15; kah1pp = 176350.0Reaction: => Cdh1; Cdc20A, Rate Law: cell*(kah1p+kah1pp*Cdc20A)*(1-Cdh1)/((Jah1+1)-Cdh1)
JiAPC = 0.001; kiAPC = 3.862259Reaction: APCP =>, Rate Law: cell*kiAPC*APCP/(JiAPC+APCP)
v46 = 0.0185337512814656Reaction: E2F => pE2F, Rate Law: cell*v46
freeCK1 = 0.55403292; kase = 19733.57Reaction: actCycE => ; actCycE, Rate Law: cell*kase*actCycE*freeCK1
ksappp = 20.28025; TFAB = 1.39800750094916E-6; ksap = 16.75325; k0=1.0; ksapp = 0.10581Reaction: => cycA; E2F, mass, Rate Law: cell*k0*(ksap+ksapp*E2F+ksappp*TFAB)*mass*2
k24r = 176.35Reaction: TriD => actCycD; TriD, Rate Law: cell*k24r*TriD

States:

NameDescription
preMPF[Cyclin-A2; Cyclin-dependent kinase 1; G2/mitotic-specific cyclin-B1]
ERG[Transcriptional regulator ERG]
E2F[CCO:42550]
DRG[Transcriptional regulator ERG]
Cdc20T[Cell division cycle protein 20 homolog]
mass[Mass]
ppRB[Retinoblastoma-associated protein]
actCycB[G2/mitotic-specific cyclin-B3]
actCycE[G1/S-specific cyclin-E1; G1/S-specific cyclin-E2]
Rb[Retinoblastoma-associated protein]
cycA[Cyclin-A2]
CKI[Cyclin-dependent kinase inhibitor 1B]
TriA[Cyclin-A2]
Cdc20A[Cell division cycle protein 20 homolog]
pE2FRB[CCO:42550; Retinoblastoma-associated protein]
APCP[anaphase-promoting complex (human); anaphase-promoting complex]
actCycACdk2[Cyclin-A2; Cyclin-dependent kinase 2]
pE2F[CCO:42550]
actCycACdk1[Cyclin-A2; Cyclin-dependent kinase 1]
E2FRB[CCO:42550; Rb-E2F complex]
cycB[G2/mitotic-specific cyclin-B3]
cycE[G1/S-specific cyclin-E2; G1/S-specific cyclin-E1]
Cdh1[Cadherin-1]
actCycD[G1/S-specific cyclin-D2; G1/S-specific cyclin-D1]
TriD[G1/S-specific cyclin-D2]

Weitz2020 - SIR model of COVID-19 transmission with shielding: BIOMD0000000963v0.0.1

The COVID-19 pandemic has precipitated a global crisis, with more than 1,430,000 confirmed cases and more than 85,000 co…

Details

The COVID-19 pandemic has precipitated a global crisis, with more than 1,430,000 confirmed cases and more than 85,000 confirmed deaths globally as of 9 April 20201-4. Mitigation and suppression of new infections have emerged as the two predominant public health control strategies5. Both strategies focus on reducing new infections by limiting human-to-human interactions, which could be both socially and economically unsustainable in the long term. We have developed and analyzed an epidemiological intervention model that leverages serological tests6,7 to identify and deploy recovered individuals8 as focal points for sustaining safer interactions via interaction substitution, developing what we term &#39;shield immunity&#39; at the population scale. The objective of a shield immunity strategy is to help to sustain the interactions necessary for the functioning of essential goods and services9 while reducing the probability of transmission. Our shield immunity approach could substantively reduce the length and reduce the overall burden of the current outbreak, and can work synergistically with social distancing. The present model highlights the value of serological testing as part of intervention strategies, in addition to its well-recognized roles in estimating prevalence10,11 and in the potential development of plasma-based therapies12-15. link: http://identifiers.org/pubmed/32382154

Werner2005_IkappaB_kinase: MODEL1008110000v0.0.1

This is the model described in the article: Stimulus specificity of gene expression programs determined by temporal co…

Details

A small number of mammalian signaling pathways mediate a myriad of distinct physiological responses to diverse cellular stimuli. Temporal control of the signaling module that contains IkappaB kinase (IKK), its substrate inhibitor of NF-kappaB (IkappaB), and the key inflammatory transcription factor NF-kappaB can allow for selective gene activation. We have demonstrated that different inflammatory stimuli induce distinct IKK profiles, and we examined the underlying molecular mechanisms. Although tumor necrosis factor-alpha (TNFalpha)-induced IKK activity was rapidly attenuated by negative feedback, lipopolysaccharide (LPS) signaling and LPS-specific gene expression programs were dependent on a cytokine-mediated positive feedback mechanism. Thus, the distinct biological responses to LPS and TNFalpha depend on signaling pathway-specific mechanisms that regulate the temporal profile of IKK activity. link: http://identifiers.org/pubmed/16166517

West2019 - Cellular interactions constrain tumor growth: BIOMD0000000820v0.0.1

These selections of models are described in the paper: Cellular interactions constrain tumor growth by Jeffrey West an…

Details

A tumor is made up of a heterogeneous collection of cell types, all competing on a fitness landscape mediated by microenvironmental conditions that dictate their interactions. Despite the fact that much is known about cell signaling, cellular cooperation, and the functional constraints that affect cellular behavior, the specifics of how these constraints (and the range over which they act) affect the macroscopic tumor growth laws that govern total volume, mass, and carrying capacity remain poorly understood. We develop a statistical mechanics approach that focuses on the total number of possible states each cell can occupy and show how different assumptions on correlations of these states give rise to the many different macroscopic tumor growth laws used in the literature. Although it is widely understood that molecular and cellular heterogeneity within a tumor is a driver of growth, here we emphasize that focusing on the functional coupling of states at the cellular level is what determines macroscopic growth characteristics. link: http://identifiers.org/pubmed/30674661

Parameters:

NameDescription
gamma = 0.666666666666667; a = 4.0Reaction: => tumor_at_Power_growth, Rate Law: compartment*a*tumor_at_Power_growth^gamma
alpha_1_variable = 0.0; alpha_0_variable = 1.0Reaction: => tumor_at_Exp_Lin_growth, Rate Law: compartment*(alpha_0_variable*tumor_at_Exp_Lin_growth+alpha_1_variable)
K = 100.0; nu = 0.3; alpha = 1.0Reaction: => tumor_at_Gen__logistic_growth, Rate Law: compartment*alpha*tumor_at_Gen__logistic_growth*(1-(tumor_at_Gen__logistic_growth/K)^nu)
K = 100.0; alpha = 1.0Reaction: => tumor_at_Gomp__growth, Rate Law: compartment*(alpha*tumor_at_Gomp__growth*ln(K)-alpha*tumor_at_Gomp__growth*ln(tumor_at_Gomp__growth))
gamma = 0.666666666666667; a = 4.0; b = 0.2Reaction: => tumor_at_Von_Bert__growth, Rate Law: compartment*(a*tumor_at_Von_Bert__growth^gamma-b*tumor_at_Von_Bert__growth)
alpha_0 = 1.0Reaction: => tumor_at_Exp_growth, Rate Law: compartment*alpha_0*tumor_at_Exp_growth

States:

NameDescription
tumor at Logistic growth[Tumor Size]
tumor at Exp Lin growth[Tumor Size]
tumor at Von Bert growth[Tumor Size]
tumor at Gomp growth[Tumor Size]
tumor at Exp growth[Tumor Size]
tumor at Gen logistic growth[Tumor Size]
tumor at Power growth[Tumor Size; power-law modular rate law]

Westerhoff2020 - systems biology model of the coronavirus pandemic 2020: BIOMD0000000988v0.0.1

Using standard systems biology methodologies a 14-compartment dynamic model was developed for the Corona virus epidemic.…

Details

Using standard systems biology methodologies a 14-compartment dynamic model was developed for the Corona virus epidemic. The model predicts that: (i) it will be impossible to limit lockdown intensity such that sufficient herd immunity develops for this epidemic to die down, (ii) the death toll from the SARS-CoV-2 virus decreases very strongly with increasing intensity of the lockdown, but (iii) the duration of the epidemic increases at first with that intensity and then decreases again, such that (iv) it may be best to begin with selecting a lockdown intensity beyond the intensity that leads to the maximum duration, (v) an intermittent lockdown strategy should also work and might be more acceptable socially and economically, (vi) an initially intensive but adaptive lockdown strategy should be most efficient, both in terms of its low number of casualties and shorter duration, (vii) such an adaptive lockdown strategy offers the advantage of being robust to unexpected imports of the virus, e.g. due to international travel, (viii) the eradication strategy may still be superior as it leads to even fewer deaths and a shorter period of economic downturn, but should have the adaptive strategy as backup in case of unexpected infection imports, (ix) earlier detection of infections is the most effective way in which the epidemic can be controlled, whilst waiting for vaccines. link: http://identifiers.org/pubmed/32532983

Westermark2003_Pancreatic_GlycOsc_basic: BIOMD0000000225v0.0.1

This is the basic model described in eq. 1 of the article: A model of phosphofructokinase and glycolytic oscillations…

Details

We have constructed a model of the upper part of the glycolysis in the pancreatic beta-cell. The model comprises the enzymatic reactions from glucokinase to glyceraldehyde-3-phosphate dehydrogenase (GAPD). Our results show, for a substantial part of the parameter space, an oscillatory behavior of the glycolysis for a large range of glucose concentrations. We show how the occurrence of oscillations depends on glucokinase, aldolase and/or GAPD activities, and how the oscillation period depends on the phosphofructokinase activity. We propose that the ratio of glucokinase and aldolase and/or GAPD activities are adequate as characteristics of the glucose responsiveness, rather than only the glucokinase activity. We also propose that the rapid equilibrium between different oligomeric forms of phosphofructokinase may reduce the oscillation period sensitivity to phosphofructokinase activity. Methodologically, we show that a satisfying description of phosphofructokinase kinetics can be achieved using the irreversible Hill equation with allosteric modifiers. We emphasize the use of parameter ranges rather than fixed values, and the use of operationally well-defined parameters in order for this methodology to be feasible. The theoretical results presented in this study apply to the study of insulin secretion mechanisms, since glycolytic oscillations have been proposed as a cause of oscillations in the ATP/ADP ratio which is linked to insulin secretion. link: http://identifiers.org/pubmed/12829470

Parameters:

NameDescription
Sfba = 0.005 mM; Vpfk = NaN mM per sec; hx = 2.5 dimensionless; Xpfk = 0.01 mM; hpfk = 2.5 dimensionless; Spfk = 4.0 mM; alpha = 5.0 dimensionless; hact = 1.0 dimensionlessReaction: G6P_F6P => FBP; F6P, Rate Law: comp*Vpfk*(F6P/Spfk)^(hpfk-(hpfk-hact)*FBP/Sfba/(1+FBP/Sfba))/((F6P/Spfk)^(hpfk-(hpfk-hact)*FBP/Sfba/(1+FBP/Sfba))+(1+(FBP/Xpfk)^hx)/(1+alpha^(hpfk-(hpfk-hact)*FBP/Sfba/(1+FBP/Sfba))*(FBP/Xpfk)^hx))
Sfba = 0.005 mM; Vfba = NaN mM per secReaction: FBP => G3P, Rate Law: comp*Vfba*FBP/Sfba/(FBP/Sfba+1)
KeqGPI = 0.3 dimensionlessReaction: F6P = G6P_F6P*KeqGPI/(1+KeqGPI), Rate Law: missing
hGK = 1.7 dimensionless; Vgk = NaN mM per sec; Sgk = 8.0 mMReaction: GLC => G6P_F6P, Rate Law: comp*Vgk*(GLC/Sgk)^hGK/(1+(GLC/Sgk)^hGK)

States:

NameDescription
G6P F6P[D-Fructose 6-phosphate; dTDP-4-dehydro-beta-L-rhamnose; keto-D-fructose 6-phosphate; keto-D-fructose 6-phosphate; alpha-D-glucose 6-phosphate]
FBP[keto-D-fructose 1,6-bisphosphate; D-Fructose 1,6-bisphosphate]
GLC[glucose; C00293]
F6P[keto-D-fructose 6-phosphate; D-Fructose 6-phosphate]
G3P[D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]

Westermark2003_Pancreatic_GlycOsc_extended: BIOMD0000000236v0.0.1

This is the extended model described in eq. 2 of the article: A model of phosphofructokinase and glycolytic oscillatio…

Details

We have constructed a model of the upper part of the glycolysis in the pancreatic beta-cell. The model comprises the enzymatic reactions from glucokinase to glyceraldehyde-3-phosphate dehydrogenase (GAPD). Our results show, for a substantial part of the parameter space, an oscillatory behavior of the glycolysis for a large range of glucose concentrations. We show how the occurrence of oscillations depends on glucokinase, aldolase and/or GAPD activities, and how the oscillation period depends on the phosphofructokinase activity. We propose that the ratio of glucokinase and aldolase and/or GAPD activities are adequate as characteristics of the glucose responsiveness, rather than only the glucokinase activity. We also propose that the rapid equilibrium between different oligomeric forms of phosphofructokinase may reduce the oscillation period sensitivity to phosphofructokinase activity. Methodologically, we show that a satisfying description of phosphofructokinase kinetics can be achieved using the irreversible Hill equation with allosteric modifiers. We emphasize the use of parameter ranges rather than fixed values, and the use of operationally well-defined parameters in order for this methodology to be feasible. The theoretical results presented in this study apply to the study of insulin secretion mechanisms, since glycolytic oscillations have been proposed as a cause of oscillations in the ATP/ADP ratio which is linked to insulin secretion. link: http://identifiers.org/pubmed/12829470

Parameters:

NameDescription
Sfba = 0.005 mM; Vpfk = NaN mM per sec; hx = 2.5 dimensionless; Xpfk = 0.01 mM; hpfk = 2.5 dimensionless; Spfk = 4.0 mM; alpha = 5.0 dimensionless; hact = 1.0 dimensionlessReaction: G6P_F6P => FBP; F6P, Rate Law: cell*Vpfk*(F6P/Spfk)^(hpfk-(hpfk-hact)*FBP/Sfba/(1+FBP/Sfba))/((F6P/Spfk)^(hpfk-(hpfk-hact)*FBP/Sfba/(1+FBP/Sfba))+(1+(FBP/Xpfk)^hx)/(1+alpha^(hpfk-(hpfk-hact)*FBP/Sfba/(1+FBP/Sfba))*(FBP/Xpfk)^hx))
Vgapdh = NaN mM per sec; Sgapdh = 0.005 mMReaction: DHAP_G3P => ; G3P, Rate Law: cell*Vgapdh*G3P/(Sgapdh+G3P)
KeqTPI = 0.045455 dimensionlessReaction: G3P = DHAP_G3P*KeqTPI/(1+KeqTPI), Rate Law: missing
Sfba = 0.005 mM; KeqFBA = 0.1 mM; Vfba = NaN mM per sec; Pfba = 0.5 mM; Qfba = 0.275 mMReaction: FBP => DHAP_G3P; G3P, DHAP, Rate Law: cell*Vfba*(FBP/Sfba-G3P*DHAP/(Pfba*Qfba*KeqFBA))/(1+FBP/Sfba+DHAP/Qfba+G3P*DHAP/(Pfba*Qfba))
hGK = 1.7 dimensionless; Vgk = NaN mM per sec; Sgk = 8.0 mMReaction: GLC => G6P_F6P, Rate Law: cell*Vgk*(GLC/Sgk)^hGK/(1+(GLC/Sgk)^hGK)
KeqGPI = 0.3 dimensionlessReaction: F6P = G6P_F6P*KeqGPI/(1+KeqGPI), Rate Law: missing

States:

NameDescription
G6P F6P[D-Glucose 6-phosphate; D-Fructose 6-phosphate; D-glucopyranose 6-phosphate; D-glucose 6-phosphate; keto-D-fructose 6-phosphate]
DHAP G3P[dihydroxyacetone phosphate; D-glyceraldehyde 3-phosphate; Glycerone phosphate; D-Glyceraldehyde 3-phosphate; dihydroxyacetone phosphate]
FBP[keto-D-fructose 1,6-bisphosphate; D-Fructose 1,6-bisphosphate]
GLC[glucose; C00293]
F6P[keto-D-fructose 6-phosphate; D-Fructose 6-phosphate]
DHAP[dihydroxyacetone phosphate; Glycerone phosphate]
G3P[D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]

Whitcomb2004_Bicarbonate_Pancreas: BIOMD0000000327v0.0.1

**A mathematical model of the pancreatic duct cell generating high bicarbonate concentrations in pancreatic juice** Dav…

Details

OBJECTIVE: To develop a simple, physiologically based mathematical model of pancreatic duct cell secretion using experimentally derived parameters that generates pancreatic fluid bicarbonate concentrations of >140 mM after CFTR activation. METHODS: A new mathematical model was developed simulating a duct cell within a proximal pancreatic duct and included a sodium-2-bicarbonate cotransporter (NBC) and sodium-potassium pump (NaK pump) on a chloride-impermeable basolateral membrane, CFTR on the luminal membrane with 0.2 to 1 bicarbonate to chloride permeability ratio. Chloride-bicarbonate antiporters (Cl/HCO3 AP) were added or subtracted from the basolateral (APb) and luminal (APl) membranes. The model was integrated over time using XPPAUT. RESULTS: This model predicts robust, NaK pump-dependent bicarbonate secretion with opening of the CFTR, generates and maintains pancreatic fluid secretion with bicarbonate concentrations >140 mM, and returns to basal levels with CFTR closure. Limiting CFTR permeability to bicarbonate, as seen in some CFTR mutations, markedly inhibited pancreatic bicarbonate and fluid secretion. CONCLUSIONS: A simple CFTR-dependent duct cell model can explain active, high-volume, high-concentration bicarbonate secretion in pancreatic juice that reproduces the experimental findings. This model may also provide insight into why CFTR mutations that predominantly affect bicarbonate permeability predispose to pancreatic dysfunction in humans. link: http://identifiers.org/pubmed/15257112

Parameters:

NameDescription
jccftr = NaN mmol per sec per m^2; zeta = 50.0 m^2 per LReaction: cl => ci, Rate Law: cell*zeta*jccftr
jac = 0.025 mmol per sec per m^2; zeta = 50.0 m^2 per LReaction: => cl, Rate Law: lumen*zeta*jac
zeta = 50.0 m^2 per L; jbcftr = NaN mmol per sec per m^2Reaction: bl => bi, Rate Law: cell*zeta*jbcftr
jnak = NaN mmol per sec per m^2; zeta = 50.0 m^2 per LReaction: ni => nb, Rate Law: cell*zeta*jnak
jnaleak = NaN mmol per sec per m^2; zeta = 50.0 m^2 per LReaction: ni => nb, Rate Law: cell*zeta*jnaleak
jac = 0.025 mmol per sec per m^2; zeta = 50.0 m^2 per L; rat = 0.25 dimensionlessReaction: => bl, Rate Law: lumen*zeta*jac*rat
japbl = NaN mmol per sec per m^2; zeta = 50.0 m^2 per LReaction: bb + ci => bi + cb, Rate Law: cell*zeta*japbl
zeta = 50.0 m^2 per L; jlum = NaN L per sec per m^2Reaction: bl =>, Rate Law: lumen*zeta*jlum*bl
japl = NaN mmol per sec per m^2; zeta = 50.0 m^2 per LReaction: bb + nb => bi + ni, Rate Law: cell*zeta*japl
zeta = 50.0 m^2 per L; buf = 0.1 L per sec per m^2; bi0 = 15.0 mMReaction: => bi, Rate Law: cell*zeta*buf*(bi0-bi)

States:

NameDescription
bl[hydrogencarbonate]
cl[chloride]
ci[chloride]
bi[hydrogencarbonate]
bb[hydrogencarbonate]
ni[sodium(1+)]
cb[chloride]
nb[sodium(1+)]

Widiastuti2010 - Genome-scale metabolic network Zymomonas mobilis (iZM363): MODEL1507180057v0.0.1

Widiastuti2010 - Genome-scale metabolic network Zymomonas mobilis (iZM363)This model is described in the article: [Geno…

Details

Bioethanol has been recognized as a potential alternative energy source. Among various ethanol-producing microbes, Zymomonas mobilis has acquired special attention due to its higher ethanol yield and tolerance. However, cellular metabolism in Z. mobilis remains unclear, hindering its practical application for bioethanol production. To elucidate such physiological characteristics, we reconstructed and validated a genome-scale metabolic network (iZM363) of Z. mobilis ATCC31821 (ZM4) based on its annotated genome and biochemical information. The phenotypic behaviors and metabolic states predicted by our genome-scale model were highly consistent with the experimental observations of Z. mobilis ZM4 strain growing on glucose as well as NMR-measured intracellular fluxes of an engineered strain utilizing glucose, fructose, and xylose. Subsequent comparative analysis with Escherichia coli and Saccharomyces cerevisiae as well as gene essentiality and flux coupling analyses have also confirmed the functional role of pdc and adh genes in the ethanologenic activity of Z. mobilis, thus leading to better understanding of this natural ethanol producer. In future, the current model could be employed to identify potential cell engineering targets, thereby enhancing the productivity of ethanol in Z. mobilis. link: http://identifiers.org/pubmed/20967753