SBMLBioModels: P - Q

P


Pitkanen2014 - Metabolic reconstruction of Aspergillus niger using CoReCo: MODEL1302010017v0.0.1

Pitkanen2014 - Metabolic reconstruction of Aspergillus niger using CoReCoThis model was reconstructed with the CoReCo me…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Aspergillus oryzae using CoReCo: MODEL1302010008v0.0.1

Pitkanen2014 - Metabolic reconstruction of Aspergillus oryzae using CoReCoThis model was reconstructed with the CoReCo m…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Aspergillus terreus using CoReCo: MODEL1302010015v0.0.1

Pitkanen2014 - Metabolic reconstruction of Aspergillus terreus using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Batrachochytrium dendrobatidis using CoReCo: MODEL1302010042v0.0.1

Pitkanen2014 - Metabolic reconstruction of Batrachochytrium dendrobatidis using CoReCoThis model was reconstructed with…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Botrytis cinerea using CoReCo: MODEL1302010027v0.0.1

Pitkanen2014 - Metabolic reconstruction of Botrytis cinerea using CoReCoThis model was reconstructed with the CoReCo met…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Candida albicans using CoReCo: MODEL1302010046v0.0.1

Pitkanen2014 - Metabolic reconstruction of Candida albicans using CoReCoThis model was reconstructed with the CoReCo met…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Candida glabrata using CoReCo: MODEL1302010028v0.0.1

Pitkanen2014 - Metabolic reconstruction of Candida glabrata using CoReCoThis model was reconstructed with the CoReCo met…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Candida lusitaniae using CoReCo: MODEL1302010037v0.0.1

Pitkanen2014 - Metabolic reconstruction of Candida lusitaniae using CoReCoThis model was reconstructed with the CoReCo m…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Candida tropicalis using CoReCo: MODEL1302010003v0.0.1

Pitkanen2014 - Metabolic reconstruction of Candida tropicalis using CoReCoThis model was reconstructed with the CoReCo m…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Chaetomium globosum using CoReCo: MODEL1302010002v0.0.1

Pitkanen2014 - Metabolic reconstruction of Chaetomium globosum using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Coccidioides immitis using CoReCo: MODEL1302010007v0.0.1

Pitkanen2014 - Metabolic reconstruction of Coccidioides immitis using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Coprinus cinereus using CoReCo: MODEL1302010006v0.0.1

Pitkanen2014 - Metabolic reconstruction of Coprinus cinereus using CoReCoThis model was reconstructed with the CoReCo me…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Cryptococcus neoformans using CoReCo: MODEL1302010039v0.0.1

Pitkanen2014 - Metabolic reconstruction of Cryptococcus neoformans using CoReCoThis model was reconstructed with the CoR…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Debaryomyces hansenii using CoReCo: MODEL1302010023v0.0.1

Pitkanen2014 - Metabolic reconstruction of Debaryomyces hansenii using CoReCoThis model was reconstructed with the CoReC…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Encephalitozoon cuniculi using CoReCo: MODEL1302010030v0.0.1

Pitkanen2014 - Metabolic reconstruction of Encephalitozoon cuniculi using CoReCoThis model was reconstructed with the Co…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Fusarium graminearum using CoReCo: MODEL1302010026v0.0.1

Pitkanen2014 - Metabolic reconstruction of Fusarium graminearum using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Fusarium oxysporum using CoReCo: MODEL1302010014v0.0.1

Pitkanen2014 - Metabolic reconstruction of Fusarium oxysporum using CoReCoThis model was reconstructed with the CoReCo m…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Fusarium verticillioides using CoReCo: MODEL1302010001v0.0.1

Pitkanen2014 - Metabolic reconstruction of Fusarium verticillioides using CoReCoThis model was reconstructed with the Co…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Histoplasma capsulatum using CoReCo: MODEL1302010022v0.0.1

Pitkanen2014 - Metabolic reconstruction of Histoplasma capsulatum using CoReCoThis model was reconstructed with the CoRe…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Kluyveromyces lactis using CoReCo: MODEL1302010011v0.0.1

Pitkanen2014 - Metabolic reconstruction of Kluyveromyces lactis using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Laccaria bicolor using CoReCo: MODEL1302010041v0.0.1

Pitkanen2014 - Metabolic reconstruction of Laccaria bicolor using CoReCoThis model was reconstructed with the CoReCo met…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Lodderomyces elongisporus using CoReCo: MODEL1302010033v0.0.1

Pitkanen2014 - Metabolic reconstruction of Lodderomyces elongisporus using CoReCoThis model was reconstructed with the C…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Magnaporthe grisea using CoReCo: MODEL1302010009v0.0.1

Pitkanen2014 - Metabolic reconstruction of Magnaporthe grisea using CoReCoThis model was reconstructed with the CoReCo m…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Mycosphaerella graminicola using CoReCo: MODEL1302010031v0.0.1

Pitkanen2014 - Metabolic reconstruction of Mycosphaerella graminicola using CoReCoThis model was reconstructed with the…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Nectria haematococca using CoReCo: MODEL1302010020v0.0.1

Pitkanen2014 - Metabolic reconstruction of Nectria haematococca using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Neosartorya fischeri using CoReCo: MODEL1302010047v0.0.1

Pitkanen2014 - Metabolic reconstruction of Neosartorya fischeri using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Neurospora crassa using CoReCo: MODEL1302010040v0.0.1

Pitkanen2014 - Metabolic reconstruction of Neurospora crassa using CoReCoThis model was reconstructed with the CoReCo me…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Phaeosphaeria nodorum using CoReCo: MODEL1302010000v0.0.1

Pitkanen2014 - Metabolic reconstruction of Phaeosphaeria nodorum using CoReCoThis model was reconstructed with the CoReC…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Phanerochaete chrysosporium using CoReCo: MODEL1302010025v0.0.1

Pitkanen2014 - Metabolic reconstruction of Phanerochaete chrysosporium using CoReCoThis model was reconstructed with the…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Phycomyces blakesleeanus using CoReCo: MODEL1302010010v0.0.1

Pitkanen2014 - Metabolic reconstruction of Phycomyces blakesleeanus using CoReCoThis model was reconstructed with the Co…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Pichia guilliermondii using CoReCo: MODEL1302010004v0.0.1

Pitkanen2014 - Metabolic reconstruction of Pichia guilliermondii using CoReCoThis model was reconstructed with the CoReC…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Pichia pastoris using CoReCo: MODEL1302010048v0.0.1

Pitkanen2014 - Metabolic reconstruction of Pichia pastoris using CoReCoThis model was reconstructed with the CoReCo meth…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Pichia stipitis using CoReCo: MODEL1302010043v0.0.1

Pitkanen2014 - Metabolic reconstruction of Pichia stipitis using CoReCoThis model was reconstructed with the CoReCo meth…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Postia placenta using CoReCo: MODEL1302010032v0.0.1

Pitkanen2014 - Metabolic reconstruction of Postia placenta using CoReCoThis model was reconstructed with the CoReCo meth…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Puccinia graminis using CoReCo: MODEL1302010045v0.0.1

Pitkanen2014 - Metabolic reconstruction of Puccinia graminis using CoReCoThis model was reconstructed with the CoReCo me…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Rhizopus oryzae using CoReCo: MODEL1302010018v0.0.1

Pitkanen2014 - Metabolic reconstruction of Rhizopus oryzae using CoReCoThis model was reconstructed with the CoReCo meth…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Saccharomyces cerevisiae using CoReCo: MODEL1302010029v0.0.1

Pitkanen2014 - Metabolic reconstruction of Saccharomyces cerevisiae using CoReCoThis model was reconstructed with the Co…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Schizosaccharomyces japonicus using CoReCo: MODEL1302010021v0.0.1

Pitkanen2014 - Metabolic reconstruction of Schizosaccharomyces japonicus using CoReCoThis model was reconstructed with t…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Schizosaccharomyces pombe using CoReCo: MODEL1302010035v0.0.1

Pitkanen2014 - Metabolic reconstruction of Schizosaccharomyces pombe using CoReCoThis model was reconstructed with the C…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Sclerotinia sclerotiorum using CoReCo: MODEL1302010034v0.0.1

Pitkanen2014 - Metabolic reconstruction of Sclerotinia sclerotiorum using CoReCoThis model was reconstructed with the Co…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Sporobolomyces roseus using CoReCo: MODEL1302010036v0.0.1

Pitkanen2014 - Metabolic reconstruction of Sporobolomyces roseus using CoReCoThis model was reconstructed with the CoReC…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Trichoderma reesei using CoReCo: MODEL1302010019v0.0.1

Pitkanen2014 - Metabolic reconstruction of Trichoderma reesei using CoReCoThis model was reconstructed with the CoReCo m…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Uncinocarpus reesii using CoReCo: MODEL1302010044v0.0.1

Pitkanen2014 - Metabolic reconstruction of Uncinocarpus reesii using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Ustilago maydis using CoReCo: MODEL1302010016v0.0.1

Pitkanen2014 - Metabolic reconstruction of Ustilago maydis using CoReCoThis model was reconstructed with the CoReCo meth…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Pitkanen2014 - Metabolic reconstruction of Yarrowia lipolytica using CoReCo: MODEL1302010013v0.0.1

Pitkanen2014 - Metabolic reconstruction of Yarrowia lipolytica using CoReCoThis model was reconstructed with the CoReCo…

Details

We introduce a novel computational approach, CoReCo, for comparative metabolic reconstruction and provide genome-scale metabolic network models for 49 important fungal species. Leveraging on the exponential growth in sequenced genome availability, our method reconstructs genome-scale gapless metabolic networks simultaneously for a large number of species by integrating sequence data in a probabilistic framework. High reconstruction accuracy is demonstrated by comparisons to the well-curated Saccharomyces cerevisiae consensus model and large-scale knock-out experiments. Our comparative approach is particularly useful in scenarios where the quality of available sequence data is lacking, and when reconstructing evolutionary distant species. Moreover, the reconstructed networks are fully carbon mapped, allowing their use in 13C flux analysis. We demonstrate the functionality and usability of the reconstructed fungal models with computational steady-state biomass production experiment, as these fungi include some of the most important production organisms in industrial biotechnology. In contrast to many existing reconstruction techniques, only minimal manual effort is required before the reconstructed models are usable in flux balance experiments. CoReCo is available at http://esaskar.github.io/CoReCo/. link: http://identifiers.org/pubmed/24516375

Plant1981_BurstingNerveCells: BIOMD0000000304v0.0.1

This a model from the article: Bifurcation and resonance in a model for bursting nerve cells. Plant RE J Math Biol…

Details

In this paper we consider a model for the phenomenon of bursting in nerve cells. Experimental evidence indicates that this phenomenon is due to the interaction of multiple conductances with very different kinetics, and the model incorporates this evidence. As a parameter is varied the model undergoes a transition between two oscillatory waveforms; a corresponding transition is observed experimentally. After establishing the periodicity of the subcritical oscillatory solution, the nature of the transition is studied. It is found to be a resonance bifurcation, with the solution branching at the critical point to another periodic solution of the same period. Using this result a comparison is made between the model and experimental observations. The model is found to predict and allow an interpretation of these observations. link: http://identifiers.org/pubmed/7252375

Parameters:

NameDescription
tau_n = NaN; n_infinity = NaNReaction: n1 = (n_infinity-n1)/tau_n, Rate Law: (n_infinity-n1)/tau_n
tau_h = NaN; h_infinity = NaNReaction: h1 = (h_infinity-h1)/tau_h, Rate Law: (h_infinity-h1)/tau_h
f = 3.0E-4; V_Ca = 140.0; K_c = 0.0085Reaction: c = f*(K_c*x1*(V_Ca-V_membrane)-c), Rate Law: f*(K_c*x1*(V_Ca-V_membrane)-c)
i_Na = NaN; i_K_Ca = NaN; i_K = NaN; i_Ca = NaN; i_L = NaNReaction: V_membrane = i_Na+i_Ca+i_K+i_K_Ca+i_L, Rate Law: i_Na+i_Ca+i_K+i_K_Ca+i_L
x_infinity = NaN; tau_x = 235.0Reaction: x1 = (x_infinity-x1)/tau_x, Rate Law: (x_infinity-x1)/tau_x

States:

NameDescription
h1[sodium(1+)]
x1[calcium(2+)]
c[calcium(2+)]
V membrane[membrane potential]
n1[potassium(1+)]

Plata2010_P_falciparum_iTH366: MODEL1007060000v0.0.1

This is the genome-scale metabolic network of Plasmodium falciparum described in the article: Reconstruction and flux-…

Details

Genome-scale metabolic reconstructions can serve as important tools for hypothesis generation and high-throughput data integration. Here, we present a metabolic network reconstruction and flux-balance analysis (FBA) of Plasmodium falciparum, the primary agent of malaria. The compartmentalized metabolic network accounts for 1001 reactions and 616 metabolites. Enzyme-gene associations were established for 366 genes and 75% of all enzymatic reactions. Compared with other microbes, the P. falciparum metabolic network contains a relatively high number of essential genes, suggesting little redundancy of the parasite metabolism. The model was able to reproduce phenotypes of experimental gene knockout and drug inhibition assays with up to 90% accuracy. Moreover, using constraints based on gene-expression data, the model was able to predict the direction of concentration changes for external metabolites with 70% accuracy. Using FBA of the reconstructed network, we identified 40 enzymatic drug targets (i.e. in silico essential genes), with no or very low sequence identity to human proteins. To demonstrate that the model can be used to make clinically relevant predictions, we experimentally tested one of the identified drug targets, nicotinate mononucleotide adenylyltransferase, using a recently discovered small-molecule inhibitor. link: http://identifiers.org/pubmed/20823846

Platelet PAR1 with SOCE: MODEL1807190001v0.0.1

Mathematical model of platelet intracellular signaling network

Details

Blood platelets need to undergo activation to carry out their function of stopping bleeding. Different activation degrees lead to a stepped hierarchy of responses: ability to aggregate, granule release, and, in a fraction of platelets, phosphatidylserine (PS) exposure. This suggests the existence of decision-making mechanisms in the platelet intracellular signaling network. To identify and investigate them, we developed a computational model of PAR1-stimulated platelet signal transduction that included a minimal set of major players in the calcium signaling network. The model comprised three intracellular compartments: cytosol, dense tubular system (DTS) and mitochondria and extracellular space. Computer simulations showed that the stable resting state of platelets is maintained via a balance between calcium pumps and leaks through the DTS and plasma membranes. Stimulation of PAR1 induced oscillations in the cytosolic calcium concentrations, in good agreement with experimental observations. Further increase in the agonist level activated the mitochondrial uniporter leading to calcium uptake by mitochondria, which caused the collapse of mitochondrial membrane potential in a fraction of platelets leading to the PS exposure. The formation of this subpopulation was shown to be a stochastic process determined by the small number of activated PAR1 receptors and by heterogeneity in the number of ion pumps. These results demonstrate how a gradual increase of the activation degree can be converted into a stepped response hierarchy ultimately leading to formation of two distinct subpopulations from an initially homogeneous population. link: http://identifiers.org/pubmed/25627921

Pokhilko1998 - Intrinsic Activation Kinetics (Dimensional Model): MODEL1808210003v0.0.1

Mathematical model of intrinsic pathway activation consisting of XIIa, kallikrein and HMWKa.

Details

A mathematical model of contact activation of blood coagulation was developed and analysed. The model variables are concentrations of factor XIIa, kallikrein and activated high-molecular-weight kininogen. Concentrations of active factors were shown to depend on the activating signal value in a hysteretic manner. Within a range of relatively small signals, two (activated and non-activated) stable states coexist (bistability). Signals of the natural environment (surfaces of endothelial and blood cells) seem to be in the range of bistability; therefore, contact activation that persists for a short time can induce a transition of the system to the activated state, and, correspondingly, the formation of a clot. The system cannot return to the initial state, which is characterized by low activation levels, until the activating signals decrease significantly below those present in the circulation. link: http://identifiers.org/doi/10.1006/jtbi.1997.0584

Pokhilko2010_CircClock: BIOMD0000000273v0.0.1

This a model from the article: Data assimilation constrains new connections and components in a complex, eukaryotic…

Details

Circadian clocks generate 24-h rhythms that are entrained by the day/night cycle. Clock circuits include several light inputs and interlocked feedback loops, with complex dynamics. Multiple biological components can contribute to each part of the circuit in higher organisms. Mechanistic models with morning, evening and central feedback loops have provided a heuristic framework for the clock in plants, but were based on transcriptional control. Here, we model observed, post-transcriptional and post-translational regulation and constrain many parameter values based on experimental data. The model's feedback circuit is revised and now includes PSEUDO-RESPONSE REGULATOR 7 (PRR7) and ZEITLUPE. The revised model matches data in varying environments and mutants, and gains robustness to parameter variation. Our results suggest that the activation of important morning-expressed genes follows their release from a night inhibitor (NI). Experiments inspired by the new model support the predicted NI function and show that the PRR5 gene contributes to the NI. The multiple PRR genes of Arabidopsis uncouple events in the late night from light-driven responses in the day, increasing the flexibility of rhythmic regulation. link: http://identifiers.org/pubmed/20865009

Parameters:

NameDescription
m6 = 0.25; m7 = 0.5; D = 0.5; p5 = 1.0; m8 = 0.1; L = 0.5Reaction: cT => ; cZG, cZTL, Rate Law: def*((m6*L+m7*D)*cT*(p5*cZTL+cZG)+m8*cT)/def
g9 = 0.3; n7 = 0.2; i = 3.0; g8 = 0.14; q3 = 2.9; h = 2.0; n4 = 0.0; L = 0.5Reaction: => cP9_m; cL, cP, cT, Rate Law: def*(L*q3*cP+(n4*L+n7*cL^i/(cL^i+g9^i))*g8^h/(cT^h+g8^h))/def
m16 = 0.5Reaction: cNI_m =>, Rate Law: def*m16*cNI_m/def
p10 = 0.36Reaction: => cNI; cNI_m, Rate Law: def*p10*cNI_m/def
m4 = 0.2Reaction: cLm =>, Rate Law: def*m4*cLm/def
m11 = 1.0; L = 0.5Reaction: cP =>, Rate Law: def*m11*cP*L/def
D = 0.5; m26 = 0.14; m25 = 0.28; L = 0.5Reaction: cTm =>, Rate Law: def*(m25*L+m26*D)*cTm/def
D = 0.5; p2 = 0.27; p1 = 0.4; L = 0.5Reaction: => cL; cL_m, Rate Law: def*cL_m*(p1*L+p2*D)/def
m10 = 0.3Reaction: cY =>, Rate Law: def*m10*cY/def
m9 = 1.0Reaction: cY_m =>, Rate Law: def*m9*cY_m/def
p4 = 0.268Reaction: => cT; cT_m, Rate Law: def*p4*cT_m/def
m21 = 0.2Reaction: cZG =>, Rate Law: def*m21*cZG/def
m24 = 0.405; D = 0.5; m17 = 0.3; L = 0.5Reaction: cNI =>, Rate Law: def*(m17*L+m24*D)*cNI/def
m18 = 1.0Reaction: cG_m =>, Rate Law: def*m18*cG_m/def
D = 0.5; p7 = 0.3Reaction: => cP, Rate Law: def*p7*D*(1-cP)/def
D = 0.5; m2 = 0.24; m1 = 0.54; L = 0.5Reaction: cL_m =>, Rate Law: def*(m1*L+m2*D)*cL_m/def
p6 = 0.44Reaction: => cY; cY_m, Rate Law: def*p6*cY_m/def
g3 = 0.4; m3 = 0.2; p3 = 0.1; c = 3.0Reaction: cL =>, Rate Law: def*(m3*cL+p3*cL^c/(cL^c+g3^c))/def
m13 = 0.32; m22 = 2.0; D = 0.5; L = 0.5Reaction: cP9 =>, Rate Law: def*(m13*L+m22*D)*cP9/def
n1 = 1.8; n0 = 0.4; g2 = 0.28; g1 = 0.1; q1 = 0.8; a = 2.0; b = 3.0; L = 0.5Reaction: => cL_m; cNI, cP, cP7, cP9, cTm, Rate Law: def*(n0*L+L*q1*cP+n1*cTm^b/(cTm^b+g2^b))*g1^a/((cP9+cP7+cNI)^a+g1^a)/def
p8 = 0.7Reaction: => cP9; cP9_m, Rate Law: def*p8*cP9_m/def
g4 = 0.91; n2 = 0.7; g5 = 0.3; e = 2.0; n3 = 0.06; d = 2.5Reaction: => cT_m; cL, cY, Rate Law: def*(n2*cY^d/(cY^d+g4^d)+n3)*g5^e/(cL^e+g5^e)/def
n8 = 0.42; g11 = 0.7; j = 3.0; n9 = 0.26; k = 3.0; g10 = 0.7Reaction: => cP7_m; cL, cLm, cP9, Rate Law: def*(n8*(cLm+cL)^j/((cLm+cL)^j+g10^j)+n9*cP9^k/(cP9^k+g11^k))/def
D = 0.5; p13 = 0.4; p12 = 30.0; L = 0.5Reaction: cG + cZTL => cZG, Rate Law: def*(p12*L*cZTL*cG-p13*D*cZG)/def
m12 = 1.0Reaction: cP9_m =>, Rate Law: def*m12*cP9_m/def
p14 = 0.45Reaction: => cZTL, Rate Law: def*p14/def
g3 = 0.4; p3 = 0.1; c = 3.0Reaction: => cLm; cL, Rate Law: def*p3*cL^c/(cL^c+g3^c)/def
m5 = 0.3Reaction: cT_m =>, Rate Law: def*m5*cT_m/def
m20 = 1.2Reaction: cZTL =>, Rate Law: def*m20*cZTL/def
g16 = 0.2; n5 = 3.4; D = 0.5; q2 = 0.5; s = 3.0; g7 = 0.18; n6 = 1.25; L = 0.5; g = 2.0Reaction: => cY_m; cL, cP, cT, Rate Law: def*(L*q2*cP+(n5*L+n6*D)*g7^s/(cT^s+g7^s)*g16^g/(cL^g+g16^g))/def
p15 = 0.05; f = 3.0; g6 = 0.3Reaction: => cTm; cT, Rate Law: def*p15*cT^f/(cT^f+g6^f)/def
p11 = 0.23Reaction: => cG; cG_m, Rate Law: def*p11*cG_m/def
m19 = 0.2Reaction: cG =>, Rate Law: def*m19*cG/def
m14 = 0.28Reaction: cP7_m =>, Rate Law: def*m14*cP7_m/def
p9 = 0.4Reaction: => cP7; cP7_m, Rate Law: def*p9*cP7_m/def
n10 = 0.18; g12 = 0.5; m = 2.0; n11 = 0.71; g13 = 0.6; l = 2.0Reaction: => cNI_m; cLm, cP7, Rate Law: def*(n10*cLm^l/(cLm^l+g12^l)+n11*cP7^m/(cP7^m+g13^m))/def
g14 = 0.17; g15 = 0.4; n = 1.0; q4 = 0.6; o = 2.0; n12 = 2.3; L = 0.5Reaction: => cG_m; cL, cP, cT, Rate Law: def*(L*q4*cP+n12*L*g15^o/(cL^o+g15^o)*g14^n/(cT^n+g14^n))/def
D = 0.5; m15 = 0.31; L = 0.5; m23 = 1.0Reaction: cP7 =>, Rate Law: def*(m15*L+m23*D)*cP7/def

States:

NameDescription
cL m[messenger RNA]
cNI[inhibitor]
cG[Protein GIGANTEA]
cP9[Two-component response regulator-like APRR9]
cP9 m[messenger RNA]
cZTL[Adagio protein 1]
cP7 m[messenger RNA]
cNI m[inhibitor; messenger RNA]
cG m[messenger RNA]
cY[protein]
cY m[messenger RNA; RNA]
cT m[messenger RNA]
cPcP
cLm[Protein CCA1; Protein LHY; protein modification]
cP7[Two-component response regulator-like APRR7]
cT[Two-component response regulator-like APRR1]
cZG[Protein GIGANTEA; Adagio protein 1]
cTm[Two-component response regulator-like APRR1; protein modification]
cL[Protein CCA1; Protein LHY]

Pokhilko2012_CircClock_RepressilatorFeedbackloop: BIOMD0000000412v0.0.1

This model is from the article: The clock gene circuit in Arabidopsis includes a repressilator with additional feedb…

Details

Circadian clocks synchronise biological processes with the day/night cycle, using molecular mechanisms that include interlocked, transcriptional feedback loops. Recent experiments identified the evening complex (EC) as a repressor that can be essential for gene expression rhythms in plants. Integrating the EC components in this role significantly alters our mechanistic, mathematical model of the clock gene circuit. Negative autoregulation of the EC genes constitutes the clock's evening loop, replacing the hypothetical component Y. The EC explains our earlier conjecture that the morning gene Pseudo-Response Regulator 9 was repressed by an evening gene, previously identified with Timing Of CAB Expression1 (TOC1). Our computational analysis suggests that TOC1 is a repressor of the morning genes Late Elongated Hypocotyl and Circadian Clock Associated1 rather than an activator as first conceived. This removes the necessity for the unknown component X (or TOC1mod) from previous clock models. As well as matching timeseries and phase-response data, the model provides a new conceptual framework for the plant clock that includes a three-component repressilator circuit in its complex structure. link: http://identifiers.org/pubmed/22395476

Parameters:

NameDescription
twilightPeriod = 0.05 3600*s; n12 = 12.5; q2 = 1.56; e = 2.0; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; g15 = 0.4; g14 = 0.004; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: s42 => cG_m; cEC, cL, cP, Rate Law: def*((((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))*q2*cP+n12*g14/(cEC+g14)*g15^e/(cL^e+g15^e))
p16 = 0.62Reaction: s31 => cE3; cE3_m, Rate Law: def*p16*cE3_m/def
twilightPeriod = 0.05 3600*s; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; m27 = 0.1; p15 = 3.0; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: cCOP1n => s40, Rate Law: def*m27*cCOP1n*(1+p15*(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod))))
twilightPeriod = 0.05 3600*s; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; m11 = 1.0; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: cP => s8, Rate Law: def*m11*cP*(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))
m16 = 0.5Reaction: cNI_m => s18, Rate Law: def*m16*cNI_m/def
twilightPeriod = 0.05 3600*s; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; m33 = 13.0; lightOffset = 0.0 3600*s; m31 = 0.3; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: cCOP1d => s41, Rate Law: def*m31*(1+m33*(1-(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))))*cCOP1d
p12 = 3.4; lightOffset = 0.0 3600*s; phase = 0.0 3600*s; twilightPeriod = 0.05 3600*s; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; p13 = 0.1; lightAmplitude = 1.0 3600*sReaction: cG + cZTL => cZG, Rate Law: def*(p12*(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))*cZTL*cG-p13*(1-(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod))))*cZG)
p8 = 0.6Reaction: s11 => cP9; cP9_m, Rate Law: def*p8*cP9_m/def
n2 = 0.64; g5 = 0.15; g4 = 0.01; e = 2.0Reaction: s21 => cT_m; cEC, cL, Rate Law: def*n2*g4/(cEC+g4)*g5^e/(cL^e+g5^e)/def
g16 = 0.3; e = 2.0; n3 = 0.29Reaction: s29 => cE3_m; cL, Rate Law: def*n3*g16^e/(cL^e+g16^e)/def
p23 = 0.37Reaction: s27 => cE4; cE4_m, Rate Law: def*p23*cE4_m/def
twilightPeriod = 0.05 3600*s; p2 = 0.27; cyclePeriod = 24.0 3600*s; p1 = 0.13; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: s3 => cL; cL_m, Rate Law: def*cL_m*(p1*(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))+p2)
p3 = 0.1; c = 2.0; g3 = 0.6Reaction: s5 => cLm; cL, Rate Law: def*p3*cL^c/(cL^c+g3^c)/def
n5 = 0.23Reaction: s38 => cCOP1c, Rate Law: def*n5/def
p11 = 0.51Reaction: s44 => cG; cG_m, Rate Law: def*p11*cG_m/def
p17 = 4.8Reaction: cE3 + cG => cEG, Rate Law: def*p17*cE3*cG/def
p27 = 0.8Reaction: s36 => cLUX; cLUX_m, Rate Law: def*p27*cLUX_m/def
m20 = 0.6Reaction: cZTL => s47, Rate Law: def*m20*cZTL/def
twilightPeriod = 0.05 3600*s; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; m24 = 0.1; phase = 0.0 3600*s; m17 = 0.5; lightAmplitude = 1.0 3600*sReaction: cNI => s20, Rate Law: def*(m17+m24*(1-(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))))*cNI
m14 = 0.4Reaction: cP7_m => s14, Rate Law: def*m14*cP7_m/def
p9 = 0.8Reaction: s15 => cP7; cP7_m, Rate Law: def*p9*cP7_m/def
m12 = 1.0Reaction: cP9_m => s10, Rate Law: def*m12*cP9_m/def
m19 = 0.2; p26 = 0.3; p28 = 2.0; m30 = 3.0; m29 = 5.0; p25 = 8.0; m37 = 0.8; p29 = 0.1; m36 = 0.1; p17 = 4.8; p21 = 1.0Reaction: cE3n => s33; cCOP1d, cCOP1n, cE4, cG, cLUX, Rate Law: def*(((m29*cE3n*cCOP1n+m30*cE3n*cCOP1d+p25*cE4*cE3n)-p21*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n))+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/def
m5 = 0.3Reaction: cT_m => s22, Rate Law: def*m5*cT_m/def
twilightPeriod = 0.05 3600*s; a = 2.0; cyclePeriod = 24.0 3600*s; n1 = 2.6; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; g1 = 0.1; q1 = 1.2; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: s1 => cL_m; cNI, cP, cP7, cP9, cT, Rate Law: def*((((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))*q1*cP+n1*g1^a/((cP9+cP7+cNI+cT)^a+g1^a))
m21 = 0.08Reaction: cZG => s48, Rate Law: def*m21*cZG/def
m26 = 0.5Reaction: cE3_m => s30, Rate Law: def*m26*cE3_m/def
m3 = 0.2; p3 = 0.1; c = 2.0; g3 = 0.6Reaction: cL => s4, Rate Law: def*(m3*cL+p3*cL^c/(cL^c+g3^c))/def
p4 = 0.56Reaction: s23 => cT; cT_m, Rate Law: def*p4*cT_m/def
p25 = 8.0; m37 = 0.8; m36 = 0.1; p26 = 0.3; m39 = 0.3; p21 = 1.0Reaction: cLUX => s37; cCOP1d, cCOP1n, cE3n, cE4, Rate Law: def*(m39*cLUX+p26*cLUX*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n))/def
m4 = 0.2Reaction: cLm => s6, Rate Law: def*m4*cLm/def
twilightPeriod = 0.05 3600*s; g9 = 0.3; n7 = 0.2; e = 2.0; cyclePeriod = 24.0 3600*s; q3 = 2.8; g8 = 0.01; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*s; n4 = 0.07Reaction: s9 => cP9_m; cEC, cL, cP, Rate Law: def*((((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))*q3*cP+(n4+n7*cL^e/(cL^e+g9^e))*g8/(cEC+g8))
p14 = 0.14Reaction: s46 => cZTL, Rate Law: def*p14/def
m32 = 0.2; m19 = 0.2; m10 = 1.0; m36 = 0.1; p17 = 4.8; d = 2.0; p24 = 10.0; lightOffset = 0.0 3600*s; p18 = 4.0; g7 = 0.6; m37 = 0.8; m9 = 1.1; phase = 0.0 3600*s; twilightPeriod = 0.05 3600*s; cyclePeriod = 24.0 3600*s; p29 = 0.1; p31 = 0.1; photoPeriod = 12.0 3600*s; p28 = 2.0; lightAmplitude = 1.0 3600*sReaction: cEC => s51; cCOP1d, cCOP1n, cE3n, cEG, cG, Rate Law: def*(m36*cCOP1n*cEC+m37*cCOP1d*cEC+m32*cEC*(1+p24*(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))*(p28*cG/(p29+m19+p17*cE3n)+(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m9*cCOP1n+m10*cCOP1d+p31))^d/((p28*cG/(p29+m19+p17*cE3n)+(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m9*cCOP1n+m10*cCOP1d+p31))^d+g7^d)))
m34 = 0.6Reaction: cE4_m => s26, Rate Law: def*m34*cE4_m/def
p25 = 8.0; m37 = 0.8; m36 = 0.1; p26 = 0.3; p21 = 1.0; m35 = 0.3Reaction: cE4 => s28; cCOP1d, cCOP1n, cE3n, cLUX, Rate Law: def*((m35*cE4+p25*cE4*cE3n)-p21*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n))/def
n10 = 0.4; g12 = 0.2; n11 = 0.6; b = 2.0; e = 2.0; g13 = 1.0Reaction: s17 => cNI_m; cLm, cP7, Rate Law: def*(n10*cLm^e/(cLm^e+g12^e)+n11*cP7^b/(cP7^b+g13^b))/def
twilightPeriod = 0.05 3600*s; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; m23 = 1.8; lightOffset = 0.0 3600*s; m15 = 0.7; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: cP7 => s16, Rate Law: def*(m15+m23*(1-(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))))*cP7
twilightPeriod = 0.05 3600*s; m22 = 0.1; cyclePeriod = 24.0 3600*s; m13 = 0.32; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: cP9 => s12, Rate Law: def*(m13+m22*(1-(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))))*cP9
m18 = 3.4Reaction: cG_m => s43, Rate Law: def*m18*cG_m/def
m19 = 0.2; p29 = 0.1; p17 = 4.8; p28 = 2.0Reaction: cG => s45; cE3n, Rate Law: def*((m19*cG+p28*cG)-p29*p28*cG/(p29+m19+p17*cE3n))/def
twilightPeriod = 0.05 3600*s; p5 = 4.0; cyclePeriod = 24.0 3600*s; m8 = 0.4; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; m6 = 0.3; m7 = 0.7; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: cT => s24; cZG, cZTL, Rate Law: def*((m6+m7*(1-(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))))*cT*(p5*cZTL+cZG)+m8*cT)
twilightPeriod = 0.05 3600*s; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; p7 = 0.3; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: s7 => cP, Rate Law: def*p7*(1-(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod))))*(1-cP)
m9 = 1.1Reaction: cE3 => s32; cCOP1c, Rate Law: def*m9*cE3*cCOP1c/def
twilightPeriod = 0.05 3600*s; n14 = 0.1; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; n6 = 20.0; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: cCOP1n => cCOP1d; cP, Rate Law: def*(n6*(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod)))*cP*cCOP1n+n14*cCOP1n)
twilightPeriod = 0.05 3600*s; m2 = 0.24; cyclePeriod = 24.0 3600*s; photoPeriod = 12.0 3600*s; lightOffset = 0.0 3600*s; m1 = 0.54; phase = 0.0 3600*s; lightAmplitude = 1.0 3600*sReaction: cL_m => s2, Rate Law: def*(m2+(m1-m2)*(((lightOffset+0.5*lightAmplitude*(1+tanh(cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))/twilightPeriod)))-0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-photoPeriod)/twilightPeriod)))+0.5*lightAmplitude*(1+tanh((cyclePeriod*((time+phase)/cyclePeriod-floor(floor(time+phase)/cyclePeriod))-cyclePeriod)/twilightPeriod))))*cL_m
m19 = 0.2; p18 = 4.0; p28 = 2.0; m10 = 1.0; p29 = 0.1; m9 = 1.1; p17 = 4.8; p31 = 0.1Reaction: cEG => s49; cCOP1c, cCOP1d, cCOP1n, cE3n, cG, Rate Law: def*((m9*cEG*cCOP1c+p18*cEG)-p31*(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m9*cCOP1n+m10*cCOP1d+p31))/def
p10 = 0.54Reaction: s19 => cNI; cNI_m, Rate Law: def*p10*cNI_m/def
p20 = 0.1; p19 = 1.0Reaction: cE3 => cE3n, Rate Law: def*(p19*cE3-p20*cE3n)/def
p25 = 8.0; m37 = 0.8; m36 = 0.1; p26 = 0.3; p21 = 1.0Reaction: s50 => cEC; cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*p26*cLUX*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n)/def
n13 = 1.3; g2 = 0.01; e = 2.0; g6 = 0.3Reaction: s25 => cE4_m; cEC, cL, Rate Law: def*n13*g2/(cEC+g2)*g6^e/(cL^e+g6^e)/def
g11 = 0.7; n9 = 0.2; f = 2.0; g10 = 0.5; n8 = 0.5; e = 2.0Reaction: s13 => cP7_m; cL, cLm, cP9, Rate Law: def*(n8*(cLm+cL)^e/((cLm+cL)^e+g10^e)+n9*cP9^f/(cP9^f+g11^f))/def

States:

NameDescription
cE4[Protein EARLY FLOWERING 4]
cNI[Two-component response regulator-like APRR5]
cLUX[Homeodomain-like superfamily protein]
s5s5
s40s40
cP9 m[Two-component response regulator-like APRR9; messenger RNA]
s37s37
s44s44
s31s31
cNI m[Two-component response regulator-like APRR3; messenger RNA]
cEG[Protein GIGANTEA; Protein EARLY FLOWERING 3]
s10s10
s34s34
s38s38
s36s36
s6s6
s46s46
s11s11
cP[obsolete protein]
s45s45
cZG[Protein GIGANTEA; Adagio protein 1]
s1s1
cG[Protein GIGANTEA]
cE3[Protein EARLY FLOWERING 3]
s17s17
s41s41
cP7 m[Two-component response regulator-like APRR7; messenger RNA]
s25s25
s2s2
s49s49
s33s33
cCOP1c[E3 ubiquitin-protein ligase COP1; cytoplasm]
cT m[Two-component response regulator-like APRR1; messenger RNA]
s28s28
cLm[Protein LHY; Protein CCA1; CCO:U0000010]
cL[Protein LHY; Protein CCA1]
s35s35
s24s24
cP9[Two-component response regulator-like APRR9]
s7s7
cZTL[Adagio protein 1]
s43s43
cCOP1n[E3 ubiquitin-protein ligase COP1; nucleus]
s47s47
cE4 m[Protein EARLY FLOWERING 4; messenger RNA]
cG m[Protein GIGANTEA; messenger RNA]
s32s32
s22s22
cCOP1d[E3 ubiquitin-protein ligase COP1; nucleus]
cE3n[Protein EARLY FLOWERING 3; nucleus]
s51s51
cP7[Two-component response regulator-like APRR7]
s3s3
cE3 m[Protein EARLY FLOWERING 3; messenger RNA]
s48s48
cEC[Protein EARLY FLOWERING 3; Protein EARLY FLOWERING 4; Homeodomain-like superfamily protein]
cL m[Protein CCA1; Protein LHY; messenger RNA]
s12s12
s4s4
cLUX m[Homeodomain-like superfamily protein; messenger RNA]
s30s30
s26s26
s42s42
s39s39
cT[Two-component response regulator-like APRR1]
s29s29
s27s27

Pokhilko2013 - TOC1 signalling in Arabidopsis circadian clock: BIOMD0000000445v0.0.1

Pokhilko2013 - TOC1 signalling in Arabidopsis circadian clockIn this model, Pokhilko et al. has incorporated the negat…

Details

24-hour biological clocks are intimately connected to the cellular signalling network, which complicates the analysis of clock mechanisms. The transcriptional regulator TOC1 (TIMING OF CAB EXPRESSION 1) is a founding component of the gene circuit in the plant circadian clock. Recent results show that TOC1 suppresses transcription of multiple target genes within the clock circuit, far beyond its previously-described regulation of the morning transcription factors LHY (LATE ELONGATED HYPOCOTYL) and CCA1 (CIRCADIAN CLOCK ASSOCIATED 1). It is unclear how this pervasive effect of TOC1 affects the dynamics of the clock and its outputs. TOC1 also appears to function in a nested feedback loop that includes signalling by the plant hormone Abscisic Acid (ABA), which is upregulated by abiotic stresses, such as drought. ABA treatments both alter TOC1 levels and affect the clock's timing behaviour. Conversely, the clock rhythmically modulates physiological processes induced by ABA, such as the closing of stomata in the leaf epidermis. In order to understand the dynamics of the clock and its outputs under changing environmental conditions, the reciprocal interactions between the clock and other signalling pathways must be integrated.We extended the mathematical model of the plant clock gene circuit by incorporating the repression of multiple clock genes by TOC1, observed experimentally. The revised model more accurately matches the data on the clock's molecular profiles and timing behaviour, explaining the clock's responses in TOC1 over-expression and toc1 mutant plants. A simplified representation of ABA signalling allowed us to investigate the interactions of ABA and circadian pathways. Increased ABA levels lengthen the free-running period of the clock, consistent with the experimental data. Adding stomatal closure to the model, as a key ABA- and clock-regulated downstream process allowed to describe TOC1 effects on the rhythmic gating of stomatal closure.The integrated model of the circadian clock circuit and ABA-regulated environmental sensing allowed us to explain multiple experimental observations on the timing and stomatal responses to genetic and environmental perturbations. These results crystallise a new role of TOC1 as an environmental sensor, which both affects the pace of the central oscillator and modulates the kinetics of downstream processes. link: http://identifiers.org/pubmed/23506153

Parameters:

NameDescription
p17 = 17.0Reaction: cE3 + cG => cEG; cE3, cG, Rate Law: def*p17*cE3*cG/def
n13 = 2.0; parameter_7 = 2.0; parameter_3 = 0.4; g2 = 0.01; e = 2.0; g6 = 0.3Reaction: => cLUX_m; cT, cEC, cL, cEC, cL, cT, Rate Law: def*parameter_3^parameter_7/(parameter_3^parameter_7+cT^parameter_7)*n13*g2/(cEC+g2)*g6^e/(cL^e+g6^e)/def
p16 = 0.62Reaction: => cE3; cE3_m, cE3_m, Rate Law: def*p16*cE3_m/def
m16 = 0.5Reaction: cNI_m => ; cNI_m, Rate Law: def*m16*cNI_m/def
parameter_14 = 0.5; n2 = 0.35; g5 = 0.2; parameter_11 = 2.0; g4 = 0.006; e = 2.0Reaction: => cT_m; cL, species_3, cEC, cEC, cL, species_3, Rate Law: def*n2/(1+(cL/(g5*(1+(species_3/parameter_14)^parameter_11)))^e)*g4/(cEC+g4)/def
m29 = 0.3Reaction: species_4 => ; species_4, Rate Law: default*m29*species_4/def
m7 = 0.1; p5 = 1.0; m6 = 0.2; m8 = 0.5; L = 0.5Reaction: cT => ; cZTL, cZG, cT, cZG, cZTL, Rate Law: def*((m6+m7*(1-L))*cT*(p5*cZTL+cZG)+m8*cT)
parameter_29 = 1.0; parameter_28 = 0.2; parameter_9 = 2.0; parameter_18 = 1.0; parameter_16 = 0.2Reaction: => species_2; species_1, species_1, Rate Law: default*parameter_28*parameter_16^parameter_9/((0.5*((parameter_29+species_1+parameter_18)-((parameter_29+species_1+parameter_18)^2-4*parameter_29*species_1)^(1/2)))^parameter_9+parameter_16^parameter_9)/def
m11 = 1.0; L = 0.5Reaction: cP => ; cP, Rate Law: def*m11*cP*L
m33 = 13.0; m31 = 0.1; L = 0.5Reaction: cCOP1d => ; cCOP1d, Rate Law: def*m31*(1+m33*(1-L))*cCOP1d
p8 = 0.6Reaction: => cP9; cP9_m, cP9_m, Rate Law: def*p8*cP9_m/def
p17 = 17.0; p29 = 0.1; m19 = 0.9; p28 = 2.0Reaction: cG => ; cE3n, cE3n, cG, Rate Law: def*((m19*cG+p28*cG)-p29*p28*cG/(p29+m19+p17*cE3n))/def
m30 = 1.0Reaction: species_3 => ; species_2, species_2, species_3, Rate Law: default*m30*species_3*species_2/def
parameter_7 = 2.0; g12 = 0.1; n10 = 0.3; e = 2.0; n11 = 0.6; b = 2.0; parameter_12 = 0.6; g13 = 1.0Reaction: => cNI_m; cT, cLm, cP7, cLm, cP7, cT, Rate Law: def*parameter_12^parameter_7/(parameter_12^parameter_7+cT^parameter_7)*(n10*cLm^e/(cLm^e+g12^e)+n11*cP7^b/(cP7^b+g13^b))/def
m32 = 0.2; parameter_26 = 0.1; m19 = 0.9; m10 = 0.1; p29 = 0.1; L = 0.5; d = 2.0; p17 = 17.0; p24 = 11.0; p18 = 4.0; p28 = 2.0; m9 = 0.2; g7 = 1.0Reaction: cEC => ; cCOP1n, cCOP1d, cG, cE3n, cEG, cCOP1d, cCOP1n, cE3n, cEC, cEG, cG, Rate Law: def*(m10*cCOP1n*cEC+m9*cCOP1d*cEC+m32*cEC*(1+p24*L*(p28*cG/(p29+m19+p17*cE3n)+(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m10*cCOP1n+m9*cCOP1d+parameter_26))^d/((p28*cG/(p29+m19+p17*cE3n)+(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m10*cCOP1n+m9*cCOP1d+parameter_26))^d+g7^d)))
g16 = 0.3; e = 2.0; n3 = 0.29Reaction: => cE3_m; cL, cL, Rate Law: def*n3*g16^e/(cL^e+g16^e)/def
p3 = 0.1; c = 2.0; g3 = 0.6Reaction: => cLm; cL, cL, Rate Law: def*p3*cL^c/(cL^c+g3^c)/def
p23 = 0.37Reaction: => cE4; cE4_m, cE4_m, Rate Law: def*p23*cE4_m/def
g14 = 0.02; parameter_7 = 2.0; q2 = 1.56; g15 = 0.4; e = 2.0; n12 = 9.0; parameter_1 = 0.6; L = 0.5Reaction: => cG_m; cT, cP, cEC, cL, cEC, cL, cP, cT, Rate Law: def*parameter_1^parameter_7/(parameter_1^parameter_7+cT^parameter_7)*(L*q2*cP+n12*g14/(cEC+g14)*g15^e/(cL^e+g15^e))
p27 = 0.8Reaction: => cLUX; cLUX_m, cLUX_m, Rate Law: def*p27*cLUX_m/def
m20 = 0.6Reaction: cZTL => ; cZTL, Rate Law: def*m20*cZTL/def
p4 = 0.5Reaction: => cT; cT_m, cT_m, Rate Law: def*p4*cT_m/def
p17 = 17.0; p26 = 0.3; m19 = 0.9; p28 = 2.0; m10 = 0.1; p29 = 0.1; m9 = 0.2; p21 = 1.0; p25 = 2.0Reaction: cE3n => ; cCOP1n, cCOP1d, cE4, cLUX, cG, cE3n, cCOP1d, cCOP1n, cE3n, cE4, cG, cLUX, Rate Law: def*(((m10*cE3n*cCOP1n+m9*cE3n*cCOP1d+p25*cE4*cE3n)-p21*p25*cE4*cE3n/(p26*cLUX+p21+m9*cCOP1d+m10*cCOP1n))+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/def
m14 = 0.4Reaction: cP7_m => ; cP7_m, Rate Law: def*m14*cP7_m/def
p9 = 0.8Reaction: => cP7; cP7_m, cP7_m, Rate Law: def*p9*cP7_m/def
m12 = 1.0Reaction: cP9_m => ; cP9_m, Rate Law: def*m12*cP9_m/def
n4 = 0.04; n7 = 0.1; g9 = 0.3; parameter_7 = 2.0; e = 2.0; g8 = 0.04; parameter_2 = 0.4; q3 = 3.0; L = 0.5Reaction: => cP9_m; cP, cL, cEC, cT, cEC, cL, cP, cT, Rate Law: def*parameter_2^parameter_7/(parameter_2^parameter_7+cT^parameter_7)*(L*q3*cP+(n4+n7*cL^e/(cL^e+g9^e))*g8/(cEC+g8))
m5 = 0.3Reaction: cT_m => ; cT_m, Rate Law: def*m5*cT_m/def
p11 = 0.5Reaction: => cG; cG_m, cG_m, Rate Law: def*p11*cG_m/def
m37 = 0.4Reaction: species_1 => ; species_1, Rate Law: default*m37*species_1/def
m21 = 0.08Reaction: cZG => ; cZG, Rate Law: def*m21*cZG/def
m24 = 0.5; m17 = 0.5; L = 0.5Reaction: cNI => ; cNI, Rate Law: def*(m17+m24*(1-L))*cNI
p1 = 0.13; p2 = 0.27; L = 0.5Reaction: => cL; cL_m, cL_m, Rate Law: def*cL_m*(p1*L+p2)
m10 = 0.1; m9 = 0.2; p26 = 0.3; p21 = 1.0; m35 = 0.3; p25 = 2.0Reaction: cE4 => ; cE3n, cLUX, cCOP1d, cCOP1n, cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*((m35*cE4+p25*cE4*cE3n)-p21*p25*cE4*cE3n/(p26*cLUX+p21+m9*cCOP1d+m10*cCOP1n))/def
m26 = 0.5Reaction: cE3_m => ; cE3_m, Rate Law: def*m26*cE3_m/def
m3 = 0.2; p3 = 0.1; c = 2.0; g3 = 0.6Reaction: cL => ; cL, Rate Law: def*(m3*cL+p3*cL^c/(cL^c+g3^c))/def
parameter_27 = 0.1Reaction: => species_3, Rate Law: default*parameter_27/def
m23 = 0.5; m15 = 0.7; L = 0.5Reaction: cP7 => ; cP7, Rate Law: def*(m15+m23*(1-L))*cP7
p12 = 10.0; L = 0.5; p13 = 0.1Reaction: cG + cZTL => cZG; cG, cZG, cZTL, Rate Law: def*(p12*L*cZTL*cG-p13*(1-L)*cZG)
p6 = 0.2Reaction: cCOP1c => cCOP1n; cCOP1c, Rate Law: def*p6*cCOP1c/def
m4 = 0.2Reaction: cLm => ; cLm, Rate Law: def*m4*cLm/def
p14 = 0.14Reaction: => cZTL, Rate Law: def*p14/def
m34 = 0.6Reaction: cLUX_m => ; cLUX_m, Rate Law: def*m34*cLUX_m/def
m10 = 0.1; m9 = 0.2; p26 = 0.3; p21 = 1.0; p25 = 2.0Reaction: => cEC; cLUX, cE4, cE3n, cCOP1d, cCOP1n, cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*p26*cLUX*p25*cE4*cE3n/(p26*cLUX+p21+m9*cCOP1d+m10*cCOP1n)/def
m10 = 0.1; m9 = 0.2; m36 = 0.3; p26 = 0.3; p21 = 1.0; p25 = 2.0Reaction: cLUX => ; cE4, cE3n, cCOP1d, cCOP1n, cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*(m36*cLUX+p26*cLUX*p25*cE4*cE3n/(p26*cLUX+p21+m9*cCOP1d+m10*cCOP1n))/def
g11 = 0.7; parameter_7 = 2.0; g10 = 0.5; n9 = 0.6; e = 2.0; f = 2.0; n8 = 0.5; parameter_6 = 0.1Reaction: => cP7_m; cLm, cL, cP9, cT, cL, cLm, cP9, cT, Rate Law: def*parameter_6^parameter_7/(parameter_6^parameter_7+cT^parameter_7)*(n8*(cLm+cL)^e/((cLm+cL)^e+g10^e)+n9*cP9^f/(cP9^f+g11^f))/def
parameter_13 = 0.3; parameter_7 = 2.0; parameter_24 = 0.5; e = 2.0; parameter_17 = 0.1Reaction: => species_1; cT, cL, cL, cT, Rate Law: default*parameter_13^parameter_7/(parameter_13^parameter_7+cT^parameter_7)*parameter_24*cL^e/(cL^e+parameter_17^e)/def
m18 = 3.4Reaction: cG_m => ; cG_m, Rate Law: def*m18*cG_m/def
m9 = 0.2Reaction: cE3 => ; cCOP1c, cCOP1c, cE3, Rate Law: def*m9*cE3*cCOP1c/def
m27 = 0.1; p15 = 2.0; L = 0.5Reaction: cCOP1c => ; cCOP1c, Rate Law: def*m27*cCOP1c*(1+p15*L)
m13 = 0.32; m22 = 0.1; L = 0.5Reaction: cP9 => ; cP9, Rate Law: def*(m13+m22*(1-L))*cP9
m2 = 0.24; m1 = 0.54; L = 0.5Reaction: cL_m => ; cL_m, Rate Law: def*(m2+(m1-m2)*L)*cL_m
parameter_20 = 0.2Reaction: species_2 => ; species_2, Rate Law: default*parameter_20*species_2/def
a = 2.0; n1 = 2.6; g1 = 0.1; q1 = 1.0; L = 0.5Reaction: => cL_m; cP, cP9, cP7, cNI, cT, cNI, cP, cP7, cP9, cT, Rate Law: def*(L*q1*cP+n1*g1^a/((cP9+cP7+cNI+cT)^a+g1^a))
n14 = 0.1; n6 = 20.0; L = 0.5Reaction: cCOP1n => cCOP1d; cP, cCOP1n, cP, Rate Law: def*(n6*L*cP*cCOP1n+n14*cCOP1n)
n5 = 0.4Reaction: => cCOP1c, Rate Law: def*n5/def
parameter_10 = 2.0; parameter_21 = 0.5; parameter_15 = 0.3; parameter_25 = 0.2; L = 0.5Reaction: => species_4; species_4, species_3, species_3, species_4, Rate Law: default*(parameter_25+parameter_21*L)*(1-species_4)*parameter_15^parameter_10/(parameter_15^parameter_10+species_3^parameter_10)/def
parameter_7 = 2.0; parameter_8 = 2.0; e = 2.0; g6 = 0.3; parameter_4 = 0.03; parameter_5 = 0.4Reaction: => cE4_m; cT, cEC, cL, cEC, cL, cT, Rate Law: def*parameter_5^parameter_7/(parameter_5^parameter_7+cT^parameter_7)*parameter_8*parameter_4/(cEC+parameter_4)*g6^e/(cL^e+g6^e)/def
p17 = 17.0; parameter_26 = 0.1; p18 = 4.0; m19 = 0.9; p28 = 2.0; m10 = 0.1; p29 = 0.1; m9 = 0.2Reaction: cEG => ; cCOP1c, cE3n, cG, cCOP1n, cCOP1d, cCOP1c, cCOP1d, cCOP1n, cE3n, cEG, cG, Rate Law: def*((m10*cEG*cCOP1c+p18*cEG)-parameter_26*(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m10*cCOP1n+m9*cCOP1d+parameter_26))/def
p10 = 0.54Reaction: => cNI; cNI_m, cNI_m, Rate Law: def*p10*cNI_m/def
p20 = 0.1; p19 = 1.0Reaction: cE3 => cE3n; cE3, cE3n, Rate Law: def*(p19*cE3-p20*cE3n)/def
p7 = 0.3; L = 0.5Reaction: => cP; cP, Rate Law: def*p7*(1-L)*(1-cP)

States:

NameDescription
cE4[Protein EARLY FLOWERING 4]
cNI[Two-component response regulator-like APRR5]
cLUX[Homeodomain-like superfamily protein]
cP9[Two-component response regulator-like APRR9]
cP9 m[Two-component response regulator-like APRR9; messenger RNA]
cZTL[Adagio protein 1]
species 1[Magnesium-chelatase subunit ChlH, chloroplastic; messenger RNA]
species 4cs
cCOP1n[E3 ubiquitin-protein ligase COP1; nucleus]
cNI m[Two-component response regulator-like APRR3; messenger RNA]
cEG[Protein GIGANTEA; Protein EARLY FLOWERING 3]
cG m[Protein GIGANTEA; messenger RNA]
cE4 m[Protein EARLY FLOWERING 4; messenger RNA]
cCOP1d[E3 ubiquitin-protein ligase COP1; nucleus]
cP[GO:0003575]
cE3n[Protein EARLY FLOWERING 3; nucleus]
cP7[Two-component response regulator-like APRR7]
cZG[Protein GIGANTEA; Adagio protein 1]
cE3 m[Protein EARLY FLOWERING 3; messenger RNA]
species 2[Protein phosphatase 2C 16]
cL m[Protein CCA1; Protein LHY; messenger RNA]
cG[Protein GIGANTEA]
cE3[Protein EARLY FLOWERING 3]
cEC[Protein EARLY FLOWERING 3; Protein EARLY FLOWERING 4; Homeodomain-like superfamily protein]
cP7 m[Two-component response regulator-like APRR7; messenger RNA]
cLUX m[Homeodomain-like superfamily protein; messenger RNA]
cT m[Two-component response regulator-like APRR1; messenger RNA]
cCOP1c[E3 ubiquitin-protein ligase COP1; cytoplasm]
species 3[Serine/threonine-protein kinase SRK2E]
cLm[Protein LHY; Protein CCA1; CCO:U0000010]
cT[Two-component response regulator-like APRR1]
cL[Protein LHY; Protein CCA1]

Poliquin2013 - Energy Deregulations in Parkinson's Disease: MODEL1410060000v0.0.1

Poliquin2013 - Energy Deregulations in Parkinson's DiseaseEncoded non-curated model. Issues: - Fluxes, reactions, param…

Details

Parkinson's disease (PD) is a multifactorial disease known to result from a variety of factors. Although age is the principal risk factor, other etiological mechanisms have been identified, including gene mutations and exposure to toxins. Deregulation of energy metabolism, mostly through the loss of complex I efficiency, is involved in disease progression in both the genetic and sporadic forms of the disease. In this study, we investigated energy deregulation in the cerebral tissue of animal models (genetic and toxin induced) of PD using an approach that combines metabolomics and mathematical modelling. In a first step, quantitative measurements of energy-related metabolites in mouse brain slices revealed most affected pathways. A genetic model of PD, the Park2 knockout, was compared to the effect of CCCP, a mitochondrial uncoupler [corrected]. Model simulated and experimental results revealed a significant and sustained decrease in ATP after CCCP exposure, but not in the genetic mice model. In support to data analysis, a mathematical model of the relevant metabolic pathways was developed and calibrated onto experimental data. In this work, we show that a short-term stress response in nucleotide scavenging is most probably induced by the toxin exposure. In turn, the robustness of energy-related pathways in the model explains how genetic perturbations, at least in young animals, are not sufficient to induce significant changes at the metabolite level. link: http://identifiers.org/pubmed/23935941

Pomerening2005- Model of the Xenopus Cdc2/APC System: MODEL2005150001v0.0.1

The cell-cycle oscillator includes an essential negative-feedback loop: Cdc2 activates the anaphase-promoting complex (A…

Details

The cell-cycle oscillator includes an essential negative-feedback loop: Cdc2 activates the anaphase-promoting complex (APC), which leads to cyclin destruction and Cdc2 inactivation. Under some circumstances, a negative-feedback loop is sufficient to generate sustained oscillations. However, the Cdc2/APC system also includes positive-feedback loops, whose functional importance we now assess. We show that short-circuiting positive feedback makes the oscillations in Cdc2 activity faster, less temporally abrupt, and damped. This compromises the activation of cyclin destruction and interferes with mitotic exit and DNA replication. This work demonstrates a systems-level role for positive-feedback loops in the embryonic cell cycle and provides an example of how oscillations can emerge out of combinations of subcircuits whose individual behaviors are not oscillatory. This work also underscores the fundamental similarity of cell-cycle oscillations in embryos to repetitive action potentials in pacemaker neurons, with both systems relying on a combination of negative and positive-feedback loops. link: http://identifiers.org/pubmed/16122424

Poolman2004_CalvinCycle: BIOMD0000000013v0.0.1

This a model from the article: Applications of metabolic modelling to plant metabolism. Poolman MG ,Assmus HE, F…

Details

In this paper some of the general concepts underpinning the computer modelling of metabolic systems are introduced. The difference between kinetic and structural modelling is emphasized, and the more important techniques from both, along with the physiological implications, are described. These approaches are then illustrated by descriptions of other work, in which they have been applied to models of the Calvin cycle, sucrose metabolism in sugar cane, and starch metabolism in potatoes. link: http://identifiers.org/pubmed/15073223

Parameters:

NameDescription
PGI_v=5.0E8; q14=2.3Reaction: F6P_ch => G6P_ch, Rate Law: PGI_v*chloroplast*(F6P_ch-G6P_ch/q14)
q15=0.058; PGM_v=5.0E8Reaction: G6P_ch => G1P_ch, Rate Law: PGM_v*chloroplast*(G6P_ch-G1P_ch/q15)
F_TKL_v=5.0E8; q7=0.084Reaction: GAP_ch + F6P_ch => X5P_ch + E4P_ch, Rate Law: chloroplast*F_TKL_v*(F6P_ch*GAP_ch-E4P_ch*X5P_ch/q7)
Light_on = 1.0; FBPase_ch_KiF6P=0.7; FBPase_ch_km=0.03; FBPase_ch_KiPi=12.0; FBPase_ch_vm=200.0Reaction: FBP_ch => F6P_ch + Pi_ch, Rate Law: Light_on*FBPase_ch_vm*FBP_ch*chloroplast/(FBP_ch+FBPase_ch_km*(1+F6P_ch/FBPase_ch_KiF6P+Pi_ch/FBPase_ch_KiPi))
q3=1.6E7; Light_on = 1.0; G3Pdh_v=5.0E8Reaction: x_NADPH_ch + BPGA_ch + x_Proton_ch => x_NADP_ch + GAP_ch + Pi_ch, Rate Law: Light_on*G3Pdh_v*chloroplast*(BPGA_ch*x_NADPH_ch*x_Proton_ch-x_NADP_ch*GAP_ch*Pi_ch/q3)
q10=0.85; G_TKL_v=5.0E8Reaction: S7P_ch + GAP_ch => R5P_ch + X5P_ch, Rate Law: chloroplast*G_TKL_v*(GAP_ch*S7P_ch-X5P_ch*R5P_ch/q10)
StPase_Vm=40.0; StPase_kiG1P=0.05; StPase_km=0.1Reaction: x_Starch_ch + Pi_ch => G1P_ch, Rate Law: StPase_Vm*Pi_ch*chloroplast/(Pi_ch+StPase_km*(1+G1P_ch/StPase_kiG1P))
Ru5Pk_ch_KiPi=4.0; Ru5Pk_ch_KiADP1=2.5; Light_on = 1.0; Ru5Pk_ch_KiADP2=0.4; Ru5Pk_ch_vm=10000.0; Ru5Pk_ch_KiPGA=2.0; Ru5Pk_ch_km1=0.05; Ru5Pk_ch_KiRuBP=0.7; Ru5Pk_ch_km2=0.05Reaction: Ru5P_ch + ATP_ch => RuBP_ch + ADP_ch; PGA_ch, Pi_ch, Rate Law: Light_on*Ru5Pk_ch_vm*Ru5P_ch*chloroplast*ATP_ch/((Ru5P_ch+Ru5Pk_ch_km1*(1+PGA_ch/Ru5Pk_ch_KiPGA+RuBP_ch/Ru5Pk_ch_KiRuBP+Pi_ch/Ru5Pk_ch_KiPi))*(ATP_ch*(1+ADP_ch/Ru5Pk_ch_KiADP1)+Ru5Pk_ch_km2*(1+ADP_ch/Ru5Pk_ch_KiADP2)))
q4=22.0; TPI_v=5.0E8Reaction: GAP_ch => DHAP_ch, Rate Law: chloroplast*TPI_v*(GAP_ch-DHAP_ch/q4)
R5Piso_v=5.0E8; q11=0.4Reaction: R5P_ch => Ru5P_ch, Rate Law: R5Piso_v*chloroplast*(R5P_ch-Ru5P_ch/q11)
Light_on = 1.0; Rbco_KiFBP=0.04; Rbco_KiNADPH=0.07; Rbco_KiPGA=0.84; Rbco_vm=340.0; Rbco_KiSBP=0.075; Rbco_km=0.02; Rbco_KiPi=0.9Reaction: RuBP_ch + x_CO2 => PGA_ch; FBP_ch, SBP_ch, Pi_ch, x_NADPH_ch, Rate Law: Light_on*Rbco_vm*RuBP_ch*chloroplast/(RuBP_ch+Rbco_km*(1+PGA_ch/Rbco_KiPGA+FBP_ch/Rbco_KiFBP+SBP_ch/Rbco_KiSBP+Pi_ch/Rbco_KiPi+x_NADPH_ch/Rbco_KiNADPH))
TP_Piap_vm=250.0; TP_Piap_kPGA_ch=0.25; TP_Piap_kDHAP_ch=0.077; TP_Piap_kPi_ch=0.63; TP_Piap_kGAP_ch=0.075; TP_Piap_kPi_cyt=0.74Reaction: x_Pi_cyt + GAP_ch => x_GAP_cyt + Pi_ch; PGA_ch, DHAP_ch, Rate Law: TP_Piap_vm*GAP_ch*chloroplast/(TP_Piap_kGAP_ch*(1+(1+TP_Piap_kPi_cyt/x_Pi_cyt)*(Pi_ch/TP_Piap_kPi_ch+PGA_ch/TP_Piap_kPGA_ch+DHAP_ch/TP_Piap_kDHAP_ch+GAP_ch/TP_Piap_kGAP_ch)))
E_Aldo_v=5.0E8; q8=13.0Reaction: DHAP_ch + E4P_ch => SBP_ch, Rate Law: chloroplast*E_Aldo_v*(E4P_ch*DHAP_ch-SBP_ch/q8)
q12=0.67; X5Pepi_v=5.0E8Reaction: X5P_ch => Ru5P_ch, Rate Law: chloroplast*X5Pepi_v*(X5P_ch-Ru5P_ch/q12)
Light_on = 1.0; SBPase_ch_km=0.013; SBPase_ch_vm=40.0; SBPase_ch_KiPi=12.0Reaction: SBP_ch => Pi_ch + S7P_ch, Rate Law: Light_on*SBPase_ch_vm*SBP_ch*chloroplast/(SBP_ch+SBPase_ch_km*(1+Pi_ch/SBPase_ch_KiPi))
q2=3.1E-4; PGK_v=5.0E8; Light_on = 1.0Reaction: PGA_ch + ATP_ch => BPGA_ch + ADP_ch, Rate Law: Light_on*PGK_v*chloroplast*(PGA_ch*ATP_ch-BPGA_ch*ADP_ch/q2)
stsyn_ch_km1=0.08; stsyn_ch_Ki=10.0; stsyn_ch_ka2=0.02; stsyn_ch_ka1=0.1; stsyn_ch_ka3=0.02; StSyn_vm=40.0; stsyn_ch_km2=0.08Reaction: ATP_ch + G1P_ch => x_Starch_ch + ADP_ch + Pi_ch; PGA_ch, F6P_ch, FBP_ch, Rate Law: StSyn_vm*G1P_ch*ATP_ch*chloroplast/((G1P_ch+stsyn_ch_km1)*(1+ADP_ch/stsyn_ch_Ki)*(ATP_ch+stsyn_ch_km2)+stsyn_ch_km2*Pi_ch/(stsyn_ch_ka1*PGA_ch)+stsyn_ch_ka2*F6P_ch+stsyn_ch_ka3*FBP_ch)
q5=7.1; F_Aldo_v=5.0E8Reaction: GAP_ch + DHAP_ch => FBP_ch, Rate Law: F_Aldo_v*chloroplast*(DHAP_ch*GAP_ch-FBP_ch/q5)
TP_Piap_vm=250.0; PGA_xpMult=0.75; TP_Piap_kPGA_ch=0.25; TP_Piap_kDHAP_ch=0.077; TP_Piap_kPi_ch=0.63; TP_Piap_kGAP_ch=0.075; TP_Piap_kPi_cyt=0.74Reaction: x_Pi_cyt + PGA_ch => x_PGA_cyt + Pi_ch; DHAP_ch, GAP_ch, Rate Law: PGA_xpMult*TP_Piap_vm*PGA_ch*chloroplast/(TP_Piap_kPGA_ch*(1+(1+TP_Piap_kPi_cyt/x_Pi_cyt)*(Pi_ch/TP_Piap_kPi_ch+PGA_ch/TP_Piap_kPGA_ch+DHAP_ch/TP_Piap_kDHAP_ch+GAP_ch/TP_Piap_kGAP_ch)))
LR_kmPi=0.3; Light_on = 1.0; LR_kmADP=0.014; LR_vm=3500.0Reaction: Pi_ch + ADP_ch => ATP_ch, Rate Law: Light_on*LR_vm*ADP_ch*Pi_ch*chloroplast/((ADP_ch+LR_kmADP)*(Pi_ch+LR_kmPi))

States:

NameDescription
E4P ch[D-erythrose 4-phosphate; D-Erythrose 4-phosphate]
DHAP ch[dihydroxyacetone phosphate; Glycerone phosphate]
PGA ch[3-Phospho-D-glycerate]
x NADPH ch[NADPH; NADPH]
x PGA cyt[3-Phospho-D-glycerate]
x DHAP cyt[dihydroxyacetone phosphate; Glycerone phosphate]
R5P ch[aldehydo-D-ribose 5-phosphate; D-Ribose 5-phosphate]
ADP ch[ADP; ADP]
FBP ch[beta-D-fructofuranose 1,6-bisphosphate; beta-D-Fructose 1,6-bisphosphate]
Pi ch[phosphate(3-); Orthophosphate]
S7P ch[sedoheptulose 7-phosphate; Sedoheptulose 7-phosphate]
Ru5P ch[D-ribulose 5-phosphate; D-Ribulose 5-phosphate]
x Pi cyt[phosphate(3-); Orthophosphate]
GAP ch[glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]
RuBP ch[D-Ribulose 1,5-bisphosphate]
ATP ch[ATP; ATP]
x Starch ch[Starch]
BPGA ch[3-Phospho-D-glyceroyl phosphate]
x Proton ch[proton]
x GAP cyt[glyceraldehyde 3-phosphate; Glyceraldehyde 3-phosphate]
x NADP ch[NADP(+); NADP+]
G6P ch[D-glucose 6-phosphate; alpha-D-Glucose 6-phosphate]
F6P ch[beta-D-fructofuranose 6-phosphate(2-)]
X5P ch[D-xylulose 5-phosphate; D-Xylulose 5-phosphate]
x CO2[carbon dioxide; CO2]
G1P ch[alpha-D-glucose 1-phosphate(2-); D-Glucose 1-phosphate]
SBP ch[sedoheptulose 1,7-bisphosphate; Sedoheptulose 1,7-bisphosphate]

Poolman2009_GS_Metabolism_Arabidopsis: MODEL3618435756v0.0.1

This is the full scale model of the Arabidopsis metabolic network described in the article: A Genome-scale Metabolic M…

Details

We describe the construction and analysis of a genome-scale metabolic model of Arabidopsis (Arabidopsis thaliana) primarily derived from the annotations in the Aracyc database. We used techniques based on linear programming to demonstrate the following: (1) that the model is capable of producing biomass components (amino acids, nucleotides, lipid, starch, and cellulose) in the proportions observed experimentally in a heterotrophic suspension culture; (2) that approximately only 15% of the available reactions are needed for this purpose and that the size of this network is comparable to estimates of minimal network size for other organisms; (3) that reactions may be grouped according to the changes in flux resulting from a hypothetical stimulus (in this case demand for ATP) and that this allows the identification of potential metabolic modules; and (4) that total ATP demand for growth and maintenance can be inferred and that this is consistent with previous estimates in prokaryotes and yeast. link: http://identifiers.org/pubmed/19755544

Poolman2009_Metab_Arabidopsis_reduced: MODEL3618487388v0.0.1

This is the reduced model of the Arabidopsis metabolic network described in the article: A Genome-scale Metabolic Mode…

Details

We describe the construction and analysis of a genome-scale metabolic model of Arabidopsis (Arabidopsis thaliana) primarily derived from the annotations in the Aracyc database. We used techniques based on linear programming to demonstrate the following: (1) that the model is capable of producing biomass components (amino acids, nucleotides, lipid, starch, and cellulose) in the proportions observed experimentally in a heterotrophic suspension culture; (2) that approximately only 15% of the available reactions are needed for this purpose and that the size of this network is comparable to estimates of minimal network size for other organisms; (3) that reactions may be grouped according to the changes in flux resulting from a hypothetical stimulus (in this case demand for ATP) and that this allows the identification of potential metabolic modules; and (4) that total ATP demand for growth and maintenance can be inferred and that this is consistent with previous estimates in prokaryotes and yeast. link: http://identifiers.org/pubmed/19755544

Potter2006_AndrogenicRegulation: MODEL8684444027v0.0.1

This a model from the article: Mathematical model for the androgenic regulation of the prostate in intact and castrate…

Details

The testicular-hypothalamic-pituitary axis regulates male reproductive system functions. Understanding these regulatory mechanisms is important for assessing the reproductive effects of environmental and pharmaceutical androgenic and antiandrogenic compounds. A mathematical model for the dynamics of androgenic synthesis, transport, metabolism, and regulation of the adult rodent ventral prostate was developed on the basis of a model by Barton and Anderson (1997). The model describes the systemic and local kinetics of testosterone (T), 5alpha-dihydrotestosterone (DHT), and luteinizing hormone (LH), with metabolism of T to DHT by 5alpha-reductase in liver and prostate. Also included are feedback loops for the positive regulation of T synthesis by LH and negative regulation of LH by T and DHT. The model simulates maintenance of the prostate as a function of hormone concentrations and androgen receptor (AR)-mediated signal transduction. The regulatory processes involved in prostate size and function include cell proliferation, apoptosis, fluid production, and 5alpha-reductase activity. Each process is controlled through the occupancy of a representative gene by androgen-AR dimers. The model simulates prostate dynamics for intact, castrated, and intravenous T-injected rats. After calibration, the model accurately captures the castration-induced regression of the prostate compared with experimental data that show that the prostate regresses to approximately 17 and 5% of its intact weight at 14 and 30 days postcastration, respectively. The model also accurately predicts serum T and AR levels following castration compared with data. This model provides a framework for quantifying the kinetics and effects of environmental and pharmaceutical endocrine active compounds on the prostate. link: http://identifiers.org/pubmed/16757547

Precup2012 - Mathematical modeling of cell dynamics after allogeneic bone marrow transplantation: BIOMD0000000800v0.0.1

This is a basic mathematical model describing the dynamics of three cell lines (normal host cells, leukemic host cells a…

Details

In this paper a basic mathematical model is introduced to describe the dynamics of three cell lines after allogeneic stem cell transplantation: normal host cells, leukemic host cells and donor cells. Their evolution is one of competitive type and depends upon kinetic and cellcell interaction parameters. Numerical simulations prove that the evolution can ultimately lead either to the normal hematopoietic state achieved by the expansion of the donor cells and the elimination of the host cells, or to the leukemic hematopoietic state characterized by the proliferation of the cancer line and the suppression of the other cell lines. One state or the other is reached depending on cellcell interactions (anti-host, anti-leukemia and anti-graft effects) and initial cell concentrations at transplantation. The model also provides a theoretical basis for the control of post-transplant evolution aimed at the achievement of normal hematopoiesis. link: http://identifiers.org/doi/10.1142/S1793524511001684

Parameters:

NameDescription
C = 0.01Reaction: y =>, Rate Law: compartment*C*y
epsilon = 1.0; A = 0.45; B = 2.2E-8; G = 2.0Reaction: => y; x, z, Rate Law: compartment*A/(1+B*(x+y+z))*(x+y+epsilon)/(x+y+epsilon+G*z)*y
epsilon = 1.0; b = 2.2E-8; h = 2.0; a = 0.23Reaction: => z; x, y, Rate Law: compartment*a/(1+b*(x+y+z))*(1-h*(x+y)/(z+epsilon+h*(x+y)))*z
epsilon = 1.0; b = 2.2E-8; a = 0.23; g = 2.0Reaction: => x; y, z, Rate Law: compartment*a/(1+b*(x+y+z))*(x+y+epsilon)/(x+y+epsilon+g*z)*x
c = 0.01Reaction: z =>, Rate Law: compartment*c*z

States:

NameDescription
x[hematopoietic stem cell; bone marrow]
z[hematopoietic stem cell; bone marrow]
y[leukemic stem cell; bone marrow]

Priebe1998_VentricularArrhythmias: MODEL8683876463v0.0.1

This a model from the article: Simulation study of cellular electric properties in heart failure Priebe L, Beuckelma…

Details

Patients with severe heart failure are at high risk of sudden cardiac death. In the majority of these patients, sudden cardiac death is thought to be due to ventricular tachyarrhythmias. Alterations of the electric properties of single myocytes in heart failure may favor the occurrence of ventricular arrhythmias in these patients by inducing early or delayed afterdepolarizations. Mathematical models of the cellular action potential and its underlying ionic currents could help to elucidate possible arrhythmogenic mechanisms on a cellular level. In the present study, selected ionic currents based on human data are incorporated into a model of the ventricular action potential for the purpose of studying the cellular electrophysiological consequences of heart failure. Ionic currents that are not yet sufficiently characterized in human ventricular myocytes are adopted from the action potential model developed by Luo and Rudy (LR model). The main results obtained from this model are as follows: The action potential in ventricular myocytes from failing hearts is longer than in nonfailing control hearts. The major underlying mechanisms for this prolongation are the enhanced activity of the Na+-Ca2+ exchanger, the slowed diastolic decay of the [Ca2+]i transient, and the reduction of the inwardly rectifying K+ current and the Na+-K+ pump current in myocytes of failing hearts. Furthermore, the fast and slow components of the delayed rectifier K+ current (I(Kr) and I(Ks), respectively) are of utmost importance in determining repolarization of the human ventricular action potential. In contrast, the influence of the transient outward K+ current on APD is only small in both cell groups. Inhibition of I(Kr) promotes the development of early afterdepolarizations in failing, but not nonfailing, myocytes. Furthermore, spontaneous Ca2+ release from the sarcoplasmic reticulum triggers a premature action potential only in failing myocytes. This model of the ventricular action potential and its alterations in heart failure is intended to serve as a tool for investigating the effects of therapeutic interventions on the electric excitability of the human ventricular myocardium. link: http://identifiers.org/pubmed/9633920

Pritchard2002_glycolysis: BIOMD0000000172v0.0.1

from: **Schemes of fluc control in a model of Saccharomyces cerevisiae glycolysis ** **Pritchard, L and Kell, DB**Eu…

Details

We used parameter scanning to emulate changes to the limiting rate for steps in a fitted model of glucose-derepressed yeast glycolysis. Three flux-control regimes were observed, two of which were under the dominant control of hexose transport, in accordance with various experimental studies and other model predictions. A third control regime in which phosphofructokinase exerted dominant glycolytic flux control was also found, but it appeared to be physiologically unreachable by this model, and all realistically obtainable flux control regimes featured hexose transport as a step involving high flux control. link: http://identifiers.org/pubmed/12180966

Parameters:

NameDescription
k_19=21.4Reaction: NAD + AcAld => NADH + Succinate, Rate Law: cell*k_19*AcAld
k1_15=45.0; k2_15=100.0Reaction: ADP => ATP + AMP, Rate Law: cell*(k1_15*ADP*ADP-k2_15*ATP*AMP)
Kp2g_9=0.08; Kp3g_9=1.2; Keq_9=0.19; Vmax_9=2585.0Reaction: P3G => P2G, Rate Law: cell*Vmax_9*(P3G/Kp3g_9-P2G/(Kp3g_9*Keq_9))/(1+P3G/Kp3g_9+P2G/Kp2g_9)
KGLYCOGEN_17=6.0Reaction: ATP + G6P => ADP + Glycogen, Rate Law: cell*KGLYCOGEN_17
Ktrehalose_18=2.4Reaction: ATP + G6P => ADP + Trehalose, Rate Law: cell*Ktrehalose_18
Keq_3=0.29; Kf6p_3=0.3; Vmax_3=1056.0; Kg6p_3=1.4Reaction: G6P => F6P, Rate Law: cell*Vmax_3*(G6P/Kg6p_3-F6P/(Kg6p_3*Keq_3))/(1+G6P/Kg6p_3+F6P/Kf6p_3)
L0_4=0.66; Kf16_4=0.111; Kamp_4=0.0995; Camp_4=0.0845; Vmax_4=110.0; Cf16_4=0.397; Katp_4=0.71; Kiatp_4=0.65; Kf6p_4=0.1; Ciatp_4=100.0; Catp_4=3.0; Kf26_4=6.82E-4; Cf26_4=0.0174; gR_4=5.12Reaction: ATP + F6P => ADP + F16bP; AMP, F26bP, Rate Law: cell*Vmax_4*gR_4*F6P/Kf6p_4*ATP/Katp_4*(1+F6P/Kf6p_4+ATP/Katp_4+gR_4*F6P/Kf6p_4*ATP/Katp_4)/((1+F6P/Kf6p_4+ATP/Katp_4+gR_4*F6P/Kf6p_4*ATP/Katp_4)^2+L0_4*((1+Ciatp_4*ATP/Kiatp_4)/(1+ATP/Kiatp_4))^2*((1+Camp_4*AMP/Kamp_4)/(1+AMP/Kamp_4))^2*((1+Cf26_4*F26bP/Kf26_4+Cf16_4*F16bP/Kf16_4)/(1+F26bP/Kf26_4+F16bP/Kf16_4))^2*(1+Catp_4*ATP/Katp_4)^2)
Kgap_5=2.4; Kf16bp_5=0.3; Kdhap_5=2.0; Kigap_5=10.0; Vmax_5=94.69; Keq_5=0.069Reaction: F16bP => DHAP + GAP, Rate Law: cell*Vmax_5*(F16bP/Kf16bp_5-DHAP*GAP/(Kf16bp_5*Keq_5))/(1+F16bP/Kf16bp_5+DHAP/Kdhap_5+GAP/Kgap_5+F16bP*GAP/(Kf16bp_5*Kigap_5)+DHAP*GAP/(Kdhap_5*Kgap_5))
Katp_8=0.3; Kp3g_8=0.53; Keq_8=3200.0; Kadp_8=0.2; Vmax_8=1288.0; Kbpg_8=0.003Reaction: ADP + BPG => ATP + P3G, Rate Law: cell*Vmax_8*(Keq_8*BPG*ADP-P3G*ATP)/(Kp3g_8*Katp_8)/((1+BPG/Kbpg_8+P3G/Kp3g_8)*(1+ADP/Kadp_8+ATP/Katp_8))
Keq_11=6500.0; Kpyr_11=21.0; Kadp_11=0.53; Vmax_11=1000.0; Kpep_11=0.14; Katp_11=1.5Reaction: ADP + PEP => ATP + PYR, Rate Law: cell*Vmax_11*(PEP*ADP/(Kpep_11*Kadp_11)-PYR*ATP/(Kpep_11*Kadp_11*Keq_11))/((1+PEP/Kpep_11+PYR/Kpyr_11)*(1+ADP/Kadp_11+ATP/Katp_11))
Kiacald_13=1.1; Kinad_13=0.92; Keq_13=6.9E-5; Kinadh_13=0.031; Kacald_13=1.11; Kietoh_13=90.0; Knadh_13=0.11; Ketoh_13=17.0; Vmax_13=209.5; Knad_13=0.17Reaction: NAD + EtOH => NADH + AcAld, Rate Law: cell*Vmax_13*(EtOH*NAD/(Ketoh_13*Kinad_13)-AcAld*NADH/(Ketoh_13*Kinad_13*Keq_13))/(1+NAD/Kinad_13+EtOH*Knad_13/(Kinad_13*Ketoh_13)+AcAld*Knadh_13/(Kinadh_13*Kacald_13)+NADH/Kinadh_13+EtOH*NAD/(Kinad_13*Ketoh_13)+NAD*AcAld*Knadh_13/(Kinad_13*Kinadh_13*Kacald_13)+EtOH*NADH*Knad_13/(Kinad_13*Kinadh_13*Ketoh_13)+AcAld*NADH/(Kacald_13*Kinadh_13)+EtOH*NAD*AcAld/(Kinad_13*Kiacald_13*Ketoh_13)+EtOH*AcAld*NADH/(Kietoh_13*Kinadh_13*Kacald_13))
Vmax_10=201.6; Kpep_10=0.5; Kp2g_10=0.04; Keq_10=6.7Reaction: P2G => PEP, Rate Law: cell*Vmax_10*(P2G/Kp2g_10-PEP/(Kp2g_10*Keq_10))/(1+P2G/Kp2g_10+PEP/Kpep_10)
Katpase_14=39.5Reaction: ATP => ADP, Rate Law: cell*Katpase_14*ATP
Kglc_1=1.1918; Ki_1=0.91; Vmax_1=97.24Reaction: GLCo => GLCi, Rate Law: Vmax_1*(GLCo-GLCi)/Kglc_1/(1+(GLCo+GLCi)/Kglc_1+Ki_1*GLCo*GLCi/Kglc_1^2)
k2_6=1.0E7; k1_6=450000.0Reaction: DHAP => GAP, Rate Law: cell*(k1_6*DHAP-k2_6*GAP)
Kadp_2=0.23; Katp_2=0.15; Kg6p_2=30.0; Kglc_2=0.08; Keq_2=2000.0; Vmax_2=236.7Reaction: GLCi + ATP => G6P + ADP, Rate Law: cell*Vmax_2*(GLCi*ATP/(Kglc_2*Katp_2)-G6P*ADP/(Kglc_2*Katp_2*Keq_2))/((1+GLCi/Kglc_2+G6P/Kg6p_2)*(1+ATP/Katp_2+ADP/Kadp_2))
C_7=1.0; Vmaxf_7=1152.0; Knadh_7=0.06; Vmaxr_7=6719.0; Knad_7=0.09; Kgap_7=0.21; Kbpg_7=0.0098Reaction: GAP + NAD => BPG + NADH, Rate Law: cell*C_7*(Vmaxf_7*GAP*NAD/(Kgap_7*Knad_7)-Vmaxr_7*BPG*NADH/(Kbpg_7*Knadh_7))/((1+GAP/Kgap_7+BPG/Kbpg_7)*(1+NAD/Knad_7+NADH/Knadh_7))
Keq_16=4300.0; Kdhap_16=0.4; Kglycerol_16=1.0; Knadh_16=0.023; Vmax_16=47.11; Knad_16=0.93Reaction: DHAP + NADH => NAD + Glycerol, Rate Law: cell*Vmax_16*(DHAP/Kdhap_16*NADH/Knadh_16-Glycerol/Kdhap_16*NAD/Knadh_16*1/Keq_16)/((1+DHAP/Kdhap_16+Glycerol/Kglycerol_16)*(1+NADH/Knadh_16+NAD/Knad_16))
Vmax_12=857.8; nH_12=1.9; Kpyr_12=4.33Reaction: PYR => AcAld + CO2, Rate Law: cell*Vmax_12*(PYR/Kpyr_12)^nH_12/(1+(PYR/Kpyr_12)^nH_12)

States:

NameDescription
ATP[ATP; ATP]
Trehalose[alpha,alpha-trehalose; alpha,alpha-Trehalose]
F16bP[beta-D-fructofuranose 1,6-bisphosphate; beta-D-Fructose 1,6-bisphosphate]
AMP[AMP; AMP]
DHAP[dihydroxyacetone phosphate; Glycerone phosphate]
GLCi[D-glucopyranose; D-Glucose]
P2G[2-phospho-D-glyceric acid; 2-Phospho-D-glycerate]
P3G[3-phospho-D-glyceric acid; 3-Phospho-D-glycerate]
Succinate[succinate(2-); Succinate]
GLCo[D-glucopyranose; D-Glucose]
AcAld[acetaldehyde; Acetaldehyde]
PYR[pyruvate; Pyruvate]
NADH[NADH; NADH]
EtOH[ethanol; Ethanol]
BPG[3-phospho-D-glyceroyl dihydrogen phosphate; 3-Phospho-D-glyceroyl phosphate]
F6P[beta-D-fructofuranose 6-phosphate; beta-D-Fructose 6-phosphate]
CO2[carbon dioxide; CO2]
Glycerol[glycerol; Glycerol]
GAP[D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]
G6P[alpha-D-glucose 6-phosphate; alpha-D-Glucose 6-phosphate]
Glycogen[glycogen; Glycogen]
NAD[NAD(+); NAD+]
ADP[ADP; ADP]
PEP[phosphoenolpyruvate; Phosphoenolpyruvate]

Pritchard2014 - plant-microbe interaction: BIOMD0000000563v0.0.1

Pritchard2014 - plant-microbe interaction[](http://www.researchgate.net/publication/269416257_Phosphoproteomic_analyses_…

Details

link: http://identifiers.org/pubmed/25382065

Parameters:

NameDescription
V=0.1; Ki=0.1; Km=0.1Reaction: E => E_int; Callose, E, Callose, Rate Law: V*E/(Km+E+Km*Callose/Ki)
k1=0.1Reaction: Path => PAMP + Path; Path, Rate Law: k1*Path
k1=0.1; k2=0.1Reaction: R + E_int => R_0; R, E_int, R_0, Rate Law: Cell*(k1*R*E_int-k2*R_0)

States:

NameDescription
PAMPPAMP
PRRPRR*
Path bulkPath_bulk
PathPath
R 0R*
CalloseCallose
RR
EE
E intE_int
PRR 0PRR

Proctor2005 - Actions of chaperones and their role in ageing: BIOMD0000000091v0.0.1

Proctor2005 - Actions of chaperones and their role in ageingThis model is described in the article: [Modelling the acti…

Details

Many molecular chaperones are also known as heat shock proteins because they are synthesised in increased amounts after brief exposure of cells to elevated temperatures. They have many cellular functions and are involved in the folding of nascent proteins, the re-folding of denatured proteins, the prevention of protein aggregation, and assisting the targeting of proteins for degradation by the proteasome and lysosomes. They also have a role in apoptosis and are involved in modulating signals for immune and inflammatory responses. Stress-induced transcription of heat shock proteins requires the activation of heat shock factor (HSF). Under normal conditions, HSF is bound to heat shock proteins resulting in feedback repression. During stress, cellular proteins undergo denaturation and sequester heat shock proteins bound to HSF, which is then able to become transcriptionally active. The induction of heat shock proteins is impaired with age and there is also a decline in chaperone function. Aberrant/damaged proteins accumulate with age and are implicated in several important age-related conditions (e.g. Alzheimer's disease, Parkinson's disease, and cataract). Therefore, the balance between damaged proteins and available free chaperones may be greatly disturbed during ageing. We have developed a mathematical model to describe the heat shock system. The aim of the model is two-fold: to explore the heat shock system and its implications in ageing; and to demonstrate how to build a model of a biological system using our simulation system (biology of ageing e-science integration and simulation (BASIS)). link: http://identifiers.org/pubmed/15610770

Parameters:

NameDescription
k14 = 0.05Reaction: TriH + HSE => HSETriH, Rate Law: k14*HSE*TriH
k3 = 50.0Reaction: MisP + Hsp90 => MCom, Rate Law: k3*MisP*Hsp90
k18 = 12.0Reaction: ADP => ATP, Rate Law: k18*ADP
k15 = 0.08Reaction: HSETriH => HSE + TriH, Rate Law: k15*HSETriH
k6 = 6.0E-7Reaction: MisP + ATP => ADP, Rate Law: k6*MisP*ATP
k4 = 1.0E-5Reaction: MCom => MisP + Hsp90, Rate Law: k4*MCom
k5 = 4.0E-6Reaction: MCom + ATP => Hsp90 + NatP + ADP, Rate Law: k5*MCom*ATP
k2 = 2.0E-5Reaction: NatP + ROS => MisP + ROS, Rate Law: k2*NatP*ROS
k13 = 0.5Reaction: DiH => HSF1, Rate Law: k13*DiH
k17 = 8.02E-9Reaction: Hsp90 + ATP => ADP, Rate Law: k17*Hsp90*ATP
k1 = 10.0Reaction: source => NatP, Rate Law: k1
k8 = 500.0Reaction: Hsp90 + HSF1 => HCom, Rate Law: k8*Hsp90*HSF1
k19 = 0.02Reaction: ATP => ADP, Rate Law: k19*ATP
k7 = 1.0E-7Reaction: MisP + AggP => AggP, Rate Law: k7*MisP*AggP
k20 = 0.1Reaction: source => ROS, Rate Law: k20
k10 = 0.01Reaction: HSF1 => DiH, Rate Law: (HSF1-1)*k10*HSF1/2
k11 = 100.0Reaction: HSF1 + DiH => TriH, Rate Law: k11*HSF1*DiH
k12 = 0.5Reaction: TriH => HSF1 + DiH, Rate Law: k12*TriH
k9 = 1.0Reaction: HCom => Hsp90 + HSF1, Rate Law: k9*HCom
k21 = 0.001Reaction: ROS =>, Rate Law: k21*ROS
k16 = 1000.0Reaction: HSETriH => HSETriH + Hsp90, Rate Law: k16*HSETriH

States:

NameDescription
DiH[protein complex; IPR000232]
ROS[reactive oxygen species]
ATP[ATP; ATP]
XX
HSETriHHSETriH
Hsp90[IPR001404]
MisPMisP
HSF1[IPR000232]
MCom[protein complex]
HCom[protein complex]
sourcesource
NatPNatP
HSEHSE
ADP[ADP; ADP]
AggPAggP
TriH[protein complex; IPR000232]

Proctor2006_telomere: BIOMD0000000087v0.0.1

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

Details

One of the DNA damage-response mechanisms in budding yeast is temporary cell-cycle arrest while DNA repair takes place. The DNA damage response requires the coordinated interaction between DNA repair and checkpoint pathways. Telomeres of budding yeast are capped by the Cdc13 complex. In the temperature-sensitive cdc13-1 strain, telomeres are unprotected over a specific temperature range leading to activation of the DNA damage response and subsequently cell-cycle arrest. Inactivation of cdc13-1 results in the generation of long regions of single-stranded DNA (ssDNA) and is affected by the activity of various checkpoint proteins and nucleases. This paper describes a mathematical model of how uncapped telomeres in budding yeast initiate the checkpoint pathway leading to cell-cycle arrest. The model was encoded in the Systems Biology Markup Language (SBML) and simulated using the stochastic simulation system Biology of Ageing e-Science Integration and Simulation (BASIS). Each simulation follows the time course of one mother cell keeping track of the number of cell divisions, the level of activity of each of the checkpoint proteins, the activity of nucleases and the amount of ssDNA generated. The model can be used to carry out a variety of in silico experiments in which different genes are knocked out and the results of simulation are compared to experimental data. Possible extensions to the model are also discussed. link: http://identifiers.org/pubmed/17015293

Parameters:

NameDescription
k6a=5.0E-5; kalive = 1.0Reaction: Exo1I => Exo1A, Rate Law: k6a*Exo1I*kalive
kalive = 1.0; k18a=0.001Reaction: S + ssDNA => S, Rate Law: k18a*S*ssDNA*kalive
k8a=0.001; kalive = 1.0Reaction: ssDNA + RPA => RPAssDNA1, Rate Law: k8a*RPA*ssDNA*kalive
k8c=100.0; kalive = 1.0Reaction: ssDNA + RPAssDNA2 => RPAssDNA, Rate Law: k8c*RPAssDNA2*ssDNA*kalive
kc4=0.01; kalive = 1.0Reaction: M + MCdkA + MG1on => budscar + G1 + MCdkI + MG1off, Rate Law: kc4*M*MCdkA*MG1on*kalive
kalive = 1.0; k18b=1.0E-5Reaction: G2 + G2Moff + ssDNA => G2 + G2Moff, Rate Law: G2*G2Moff*k18b*ssDNA*kalive
k19=0.001; kalive = 1.0Reaction: Cdc13 + Rad17Utelo + recovery => Ctelo + Rad17 + recovery, Rate Law: Cdc13*k19*Rad17Utelo*recovery*kalive
k17a=0.05; kalive = 1.0Reaction: Mec1RPAssDNA + S => Mec1 + RPA + S + ssDNA, Rate Law: k17a*Mec1RPAssDNA*S*kalive
kalive = 1.0; k16=0.1Reaction: Dun1A + G2Mon => Dun1A + G2Moff, Rate Law: Dun1A*G2Mon*k16*kalive
k9=100.0; kalive = 1.0Reaction: Rad9Kin + Rad9I => Rad9Kin + Rad9A, Rate Law: k9*Rad9Kin*Rad9I*kalive
kalive = 1.0; k8d=0.004Reaction: RPAssDNA + Mec1 => Mec1RPAssDNA, Rate Law: k8d*RPAssDNA*Mec1*kalive
kalive = 1.0; k1=5.0E-4Reaction: Cdc13 + Utelo => Ctelo, Rate Law: k1*Cdc13*Utelo*kalive
k5=3.0E-4; kalive = 1.0Reaction: ExoXA + Rad17Utelo => ExoXA + Rad17Utelo + ssDNA, Rate Law: k5*ExoXA*Rad17Utelo*kalive
kalive = 1.0; k7a=3.0E-5Reaction: Utelo + Exo1A => Utelo + Exo1A + ssDNA, Rate Law: k7a*Utelo*Exo1A*kalive
k15=0.2; kalive = 1.0Reaction: Chk1A + G2Mon => Chk1A + G2Moff, Rate Law: Chk1A*G2Mon*k15*kalive
kalive = 1.0; k6b=5.0E-4Reaction: Exo1I + Rad24 => Exo1A + Rad24, Rate Law: k6b*Exo1I*Rad24*kalive
k14=3.3E-6; kalive = 1.0Reaction: Dun1I + Rad53A => Dun1A + Rad53A, Rate Law: Dun1I*k14*Rad53A*kalive
kalive = 1.0; k4=0.01Reaction: ExoXI + Rad17Utelo => ExoXA + Rad17Utelo, Rate Law: k4*ExoXI*Rad17Utelo*kalive
kalive = 1.0; k10a=0.05Reaction: ExoXA + Rad9A => ExoXI + Rad9A, Rate Law: ExoXA*k10a*Rad9A*kalive
kc3=0.0012; kalive = 1.0Reaction: Scyclin => sink, Rate Law: kc3*Scyclin*kalive
k7b=3.0E-5; kalive = 1.0Reaction: Rad17Utelo + Exo1A => Rad17Utelo + Exo1A + ssDNA, Rate Law: k7b*Rad17Utelo*Exo1A*kalive
k8b=100.0; kalive = 1.0Reaction: ssDNA + RPAssDNA1 => RPAssDNA2, Rate Law: k8b*RPAssDNA1*ssDNA*kalive
kc1=0.16; kalive = 1.0Reaction: S => Scyclin + S, Rate Law: kc1*S*kalive
kalive = 1.0; k3=1.5E-8Reaction: Utelo + Rad17 + Rad24 + ATP => Rad17Utelo + Rad24 + ADP, Rate Law: k3*Utelo*Rad17*Rad24*ATP*kalive/(5000+ATP)
k10b=0.05; kalive = 1.0Reaction: ExoXA + Rad9I => ExoXI + Rad9I, Rate Law: ExoXA*k10b*Rad9I*kalive
k17b=0.05; kalive = 1.0Reaction: G2 + G2Moff + Mec1RPAssDNA => G2 + G2Moff + Mec1 + RPA + ssDNA, Rate Law: G2*G2Moff*k17b*Mec1RPAssDNA*kalive
kalive = 1.0; k2=3.85E-4Reaction: Ctelo => Cdc13 + Utelo, Rate Law: k2*Ctelo*kalive
k13=1.0; kalive = 1.0Reaction: Exo1A + Rad53A => Exo1I + Rad53A, Rate Law: Exo1A*k13*Rad53A*kalive
kc2=0.01; kalive = 1.0Reaction: G1Soff + G1 + G1CdkA => G1Son + G1 + G1CdkA, Rate Law: G1*G1CdkA*G1Soff*kc2*kalive

States:

NameDescription
G2Mon[G2/M transition of mitotic cell cycle]
SG2off[obsolete regulation of transcription involved in S phase of mitotic cell cycle]
G2CdkA[nuclear cyclin-dependent protein kinase holoenzyme complex]
Rad9I[DNA repair protein RAD9]
RPAssDNA[C00271; PIRSF002091; single-stranded DNA]
Rad17[DNA damage checkpoint control protein RAD17]
Dun1I[DNA damage response protein kinase DUN1]
MCdkI[nuclear cyclin-dependent protein kinase holoenzyme complex]
Cdc13[Cell division control protein 13]
Rad17Utelo[DNA damage checkpoint control protein RAD17; chromosome, telomeric region]
RPAssDNA2[C00271; PIRSF002091; single-stranded DNA]
Exo1I[Exodeoxyribonuclease 1]
M[M phase]
G2CdkI[nuclear cyclin-dependent protein kinase holoenzyme complex]
G2[G2 phase]
G2cyclin[G2/mitotic-specific cyclin-1]
G1CdkA[nuclear cyclin-dependent protein kinase holoenzyme complex]
RPA[PIRSF002091]
ExoXA[Exodeoxyribonuclease 10]
RPAssDNA1[C00271; PIRSF002091; single-stranded DNA]
G1Son[G1/S transition of mitotic cell cycle]
ExoXI[Exodeoxyribonuclease 10]
G1[G1 phase]
G1CdkI[nuclear cyclin-dependent protein kinase holoenzyme complex]
MG1offMG1off
G1Soff[mitotic cell cycle checkpoint]
Ctelo[telomere cap complex; chromosome, telomeric region]
Mec1RPAssDNA[Serine/threonine-protein kinase MEC1; C00271; PIRSF002091; single-stranded DNA]
MCdkA[nuclear cyclin-dependent protein kinase holoenzyme complex]
SCdkI[nuclear cyclin-dependent protein kinase holoenzyme complex]
Utelo[chromosome, telomeric region]
Mcyclin[Meiosis-specific cyclin rem1]
Mec1[Serine/threonine-protein kinase MEC1]
SCdkA[nuclear cyclin-dependent protein kinase holoenzyme complex]
S[mitotic S phase]
ssDNA[CHEBI:09160; C00271]
G2Moff[G2 DNA damage checkpoint]
SG2on[obsolete regulation of transcription involved in S phase of mitotic cell cycle]
ADP[ADP]
Exo1A[Exodeoxyribonuclease 1]
Scyclin[S-phase entry cyclin-5]
Rad9A[DNA repair protein RAD9]

Proctor2007 - Age related decline of proteolysis, ubiquitin-proteome system: BIOMD0000000105v0.0.1

Proctor2007 - Age related decline of proteolysis, ubiquitin-proteome systemThis is a stochastic model of the ubiquitin-…

Details

The ubiquitin-proteasome system is responsible for homeostatic degradation of intact protein substrates as well as the elimination of damaged or misfolded proteins that might otherwise aggregate. During ageing there is a decline in proteasome activity and an increase in aggregated proteins. Many neurodegenerative diseases are characterised by the presence of distinctive ubiquitin-positive inclusion bodies in affected regions of the brain. These inclusions consist of insoluble, unfolded, ubiquitinated polypeptides that fail to be targeted and degraded by the proteasome. We are using a systems biology approach to try and determine the primary event in the decline in proteolytic capacity with age and whether there is in fact a vicious cycle of inhibition, with accumulating aggregates further inhibiting proteolysis, prompting accumulation of aggregates and so on. A stochastic model of the ubiquitin-proteasome system has been developed using the Systems Biology Mark-up Language (SBML). Simulations are carried out on the BASIS (Biology of Ageing e-Science Integration and Simulation) system and the model output is compared to experimental data wherein levels of ubiquitin and ubiquitinated substrates are monitored in cultured cells under various conditions. The model can be used to predict the effects of different experimental procedures such as inhibition of the proteasome or shutting down the enzyme cascade responsible for ubiquitin conjugation.The model output shows good agreement with experimental data under a number of different conditions. However, our model predicts that monomeric ubiquitin pools are always depleted under conditions of proteasome inhibition, whereas experimental data show that monomeric pools were depleted in IMR-90 cells but not in ts20 cells, suggesting that cell lines vary in their ability to replenish ubiquitin pools and there is the need to incorporate ubiquitin turnover into the model. Sensitivity analysis of the model revealed which parameters have an important effect on protein turnover and aggregation kinetics.We have developed a model of the ubiquitin-proteasome system using an iterative approach of model building and validation against experimental data. Using SBML to encode the model ensures that it can be easily modified and extended as more data become available. Important aspects to be included in subsequent models are details of ubiquitin turnover, models of autophagy, the inclusion of a pool of short-lived proteins and further details of the aggregation process. link: http://identifiers.org/pubmed/17408507

Parameters:

NameDescription
k3 = 4.0E-6Reaction: MisP => NatP + refNatP, Rate Law: k3*MisP
k61 = 1.7E-5Reaction: MisP + E3 => E3_MisP, Rate Law: k61*MisP*E3
k1 = 0.01Reaction: Source => NatP, Rate Law: k1*Source
k72 = 1.0E-8Reaction: MisP_Ub3 + MisP_Ub4 => AggP, Rate Law: k72*MisP_Ub3*MisP_Ub4
k61r = 2.0E-4Reaction: E3_MisP => MisP + E3, Rate Law: k61r*E3_MisP
k71 = 1.0E-8Reaction: MisP => AggP, Rate Law: k71*MisP*(MisP-1)*0.5
k63 = 0.001Reaction: E2 + E1_Ub => E2_Ub + E1, Rate Law: k63*E2*E1_Ub
k69 = 0.0Reaction: MisP_Ub4_Proteasome + ATP => Ub + Proteasome + ADP + degUb4, Rate Law: k69*MisP_Ub4_Proteasome*ATP/(5000+ATP)
k65 = 0.01Reaction: MisP_Ub + E2_Ub => MisP_Ub2 + E2, Rate Law: k65*MisP_Ub*E2_Ub
k2 = 2.0E-6Reaction: NatP + ROS => MisP + ROS + totMisP, Rate Law: k2*NatP*ROS
k66 = 1.0E-5Reaction: MisP_Ub5 + DUB => MisP_Ub4 + DUB + Ub, Rate Law: k66*MisP_Ub5*DUB
k64 = 0.001Reaction: E2_Ub + E3_MisP => MisP_Ub + E2 + E3, Rate Law: k64*E2_Ub*E3_MisP
k67 = 1.0E-5Reaction: MisP_Ub6 + Proteasome => MisP_Ub6_Proteasome, Rate Law: k67*MisP_Ub6*Proteasome
k62 = 2.0E-4Reaction: E1 + Ub + ATP => E1_Ub + AMP, Rate Law: k62*E1*Ub*ATP/(5000+ATP)
k68 = 1.0E-5Reaction: MisP_Ub4_Proteasome + DUB => MisP_Ub3 + Proteasome + Ub + DUB, Rate Law: k68*MisP_Ub4_Proteasome*DUB

States:

NameDescription
MisP Ub4MisP_Ub4
MisP Ub5MisP_Ub5
MisP Ub7MisP_Ub7
E3 MisPE3_MisP
MisPMisP
E1 Ub[IPR000011; IPR000626]
MisP UbMisP_Ub
MisP Ub8MisP_Ub8
MisP Ub6MisP_Ub6
MisP Ub5 ProteasomeMisP_Ub5_Proteasome
MisP Ub2MisP_Ub2
MisP Ub4 ProteasomeMisP_Ub4_Proteasome
MisP Ub3MisP_Ub3
E2 Ub[IPR000626; IPR000608]
NatPNatP

Proctor2008 - p53/Mdm2 circuit - p53 stabilisation by ATM: BIOMD0000000188v0.0.1

Proctor2008 - p53/Mdm2 circuit - p53 stabilisation by ATMThis model is described in the article: [Explaining oscillatio…

Details

In individual living cells p53 has been found to be expressed in a series of discrete pulses after DNA damage. Its negative regulator Mdm2 also demonstrates oscillatory behaviour. Attempts have been made recently to explain this behaviour by mathematical models but these have not addressed explicit molecular mechanisms. We describe two stochastic mechanistic models of the p53/Mdm2 circuit and show that sustained oscillations result directly from the key biological features, without assuming complicated mathematical functions or requiring more than one feedback loop. Each model examines a different mechanism for providing a negative feedback loop which results in p53 activation after DNA damage. The first model (ARF model) looks at the mechanism of p14ARF which sequesters Mdm2 and leads to stabilisation of p53. The second model (ATM model) examines the mechanism of ATM activation which leads to phosphorylation of both p53 and Mdm2 and increased degradation of Mdm2, which again results in p53 stabilisation. The models can readily be modified as further information becomes available, and linked to other models of cellular ageing.The ARF model is robust to changes in its parameters and predicts undamped oscillations after DNA damage so long as the signal persists. It also predicts that if there is a gradual accumulation of DNA damage, such as may occur in ageing, oscillations break out once a threshold level of damage is acquired. The ATM model requires an additional step for p53 synthesis for sustained oscillations to develop. The ATM model shows much more variability in the oscillatory behaviour and this variability is observed over a wide range of parameter values. This may account for the large variability seen in the experimental data which so far has examined ARF negative cells.The models predict more regular oscillations if ARF is present and suggest the need for further experiments in ARF positive cells to test these predictions. Our work illustrates the importance of systems biology approaches to understanding the complex role of p53 in both ageing and cancer. link: http://identifiers.org/pubmed/18706112

Parameters:

NameDescription
ksynp53 = 0.006 psecReaction: p53_mRNA => p53 + p53_mRNA + p53syn, Rate Law: ksynp53*p53_mRNA
ksynMdm2 = 4.95E-4 psecReaction: Mdm2_mRNA => Mdm2_mRNA + Mdm2 + mdm2syn, Rate Law: ksynMdm2*Mdm2_mRNA
IR = 0.0 dGy; kdam = 0.08 molepsecpdGyReaction: => damDNA, Rate Law: kdam*IR
krepair = 2.0E-5 psecReaction: damDNA => Sink, Rate Law: krepair*damDNA
kdegMdm2mRNA = 1.0E-4 psecReaction: Mdm2_mRNA => Sink + Mdm2mRNAdeg, Rate Law: kdegMdm2mRNA*Mdm2_mRNA
kproteff = 1.0 dimensionless; kdegp53 = 8.25E-4 psecReaction: Mdm2_p53 => Mdm2 + p53deg, Rate Law: kdegp53*Mdm2_p53*kproteff
kdegATMMdm2 = 4.0E-4 psecReaction: Mdm2_P => Sink + mdm2deg, Rate Law: kdegATMMdm2*Mdm2_P
kdephosp53 = 0.5 psecReaction: p53_P => p53, Rate Law: kdephosp53*p53_P
kbinMdm2p53 = 0.001155 pmolpsecReaction: p53 + Mdm2 => Mdm2_p53, Rate Law: kbinMdm2p53*p53*Mdm2
krelMdm2p53 = 1.155E-5 psecReaction: Mdm2_p53 => p53 + Mdm2, Rate Law: krelMdm2p53*Mdm2_p53
kdephosMdm2 = 0.5 psecReaction: Mdm2_P => Mdm2, Rate Law: kdephosMdm2*Mdm2_P
kphosMdm2 = 2.0 pmolpsecReaction: Mdm2 + ATMA => Mdm2_P + ATMA, Rate Law: kphosMdm2*Mdm2*ATMA
kproteff = 1.0 dimensionless; kdegMdm2 = 4.33E-4 psecReaction: Mdm2 => Sink + mdm2deg, Rate Law: kdegMdm2*Mdm2*kproteff
kinactATM = 5.0E-4 psecReaction: ATMA => ATMI, Rate Law: kinactATM*ATMA
kphosp53 = 5.0E-4 pmolpsecReaction: p53 + ATMA => p53_P + ATMA, Rate Law: kphosp53*p53*ATMA
kactATM = 1.0E-4 pmolpsecReaction: damDNA + ATMI => damDNA + ATMA, Rate Law: kactATM*damDNA*ATMI
ksynp53mRNA = 0.001 psecReaction: Source => p53_mRNA, Rate Law: ksynp53mRNA*Source
kdegp53mRNA = 1.0E-4 psecReaction: p53_mRNA => Sink, Rate Law: kdegp53mRNA*p53_mRNA
ksynMdm2mRNA = 1.0E-4 psecReaction: p53_P => p53_P + Mdm2_mRNA + Mdm2mRNAsyn, Rate Law: ksynMdm2mRNA*p53_P

States:

NameDescription
Mdm2 P[MDM2; E3 ubiquitin-protein ligase Mdm2]
mdm2deg[proteasome-mediated ubiquitin-dependent protein catabolic process]
damDNA[deoxyribonucleic acid; cellular response to DNA damage stimulus]
p53 mRNA[messenger RNA; RNA]
Mdm2mRNAdeg[mRNA catabolic process]
mdm2syn[translation]
ATMA[Serine-protein kinase ATM]
p53[Cellular tumor antigen p53; TP53]
p53deg[proteasome-mediated ubiquitin-dependent protein catabolic process]
totp53totp53
Mdm2 p53[Cellular tumor antigen p53; E3 ubiquitin-protein ligase Mdm2]
SourceSource
p53 P[Cellular tumor antigen p53; TP53]
ATMI[Serine-protein kinase ATM]
p53syn[translation]
totMdm2totMdm2
SinkSink
Mdm2[E3 ubiquitin-protein ligase Mdm2; MDM2]
Mdm2 mRNA[messenger RNA; RNA]
Mdm2mRNAsyn[transcription factor activity, sequence-specific DNA binding]

Proctor2008 - p53/Mdm2 circuit - p53 stablisation by p14ARF: BIOMD0000000189v0.0.1

Proctor2008 - p53/Mdm2 circuit - p53 stabilisation by p14ARFThis model is described in the article: [Explaining oscilla…

Details

In individual living cells p53 has been found to be expressed in a series of discrete pulses after DNA damage. Its negative regulator Mdm2 also demonstrates oscillatory behaviour. Attempts have been made recently to explain this behaviour by mathematical models but these have not addressed explicit molecular mechanisms. We describe two stochastic mechanistic models of the p53/Mdm2 circuit and show that sustained oscillations result directly from the key biological features, without assuming complicated mathematical functions or requiring more than one feedback loop. Each model examines a different mechanism for providing a negative feedback loop which results in p53 activation after DNA damage. The first model (ARF model) looks at the mechanism of p14ARF which sequesters Mdm2 and leads to stabilisation of p53. The second model (ATM model) examines the mechanism of ATM activation which leads to phosphorylation of both p53 and Mdm2 and increased degradation of Mdm2, which again results in p53 stabilisation. The models can readily be modified as further information becomes available, and linked to other models of cellular ageing.The ARF model is robust to changes in its parameters and predicts undamped oscillations after DNA damage so long as the signal persists. It also predicts that if there is a gradual accumulation of DNA damage, such as may occur in ageing, oscillations break out once a threshold level of damage is acquired. The ATM model requires an additional step for p53 synthesis for sustained oscillations to develop. The ATM model shows much more variability in the oscillatory behaviour and this variability is observed over a wide range of parameter values. This may account for the large variability seen in the experimental data which so far has examined ARF negative cells.The models predict more regular oscillations if ARF is present and suggest the need for further experiments in ARF positive cells to test these predictions. Our work illustrates the importance of systems biology approaches to understanding the complex role of p53 in both ageing and cancer. link: http://identifiers.org/pubmed/18706112

Parameters:

NameDescription
kbinMdm2p53 = 0.001155 pmolepsecReaction: p53 + Mdm2 => Mdm2_p53, Rate Law: kbinMdm2p53*p53*Mdm2
ksynMdm2 = 4.95E-4 psecReaction: Mdm2_mRNA => Mdm2_mRNA + Mdm2 + mdm2syn, Rate Law: ksynMdm2*Mdm2_mRNA
IR = 0.0 dGy; kdam = 0.08 molepsecpdGyReaction: => damDNA + totdamDNA, Rate Law: kdam*IR
kdegMdm2mRNA = 1.0E-4 psecReaction: Mdm2_mRNA => Sink + Mdm2mRNAdeg, Rate Law: kdegMdm2mRNA*Mdm2_mRNA
krepair = 2.0E-5 psecReaction: damDNA => Sink, Rate Law: krepair*damDNA
kproteff = 1.0 dimensionless; kdegp53 = 8.25E-4 psecReaction: Mdm2_p53 => Mdm2 + p53deg, Rate Law: kdegp53*Mdm2_p53*kproteff
ksynp53 = 0.078 psecReaction: Source => p53 + p53syn, Rate Law: ksynp53*Source
kproteff = 1.0 dimensionless; kdegARF = 1.0E-4 psecReaction: ARF => Sink, Rate Law: kdegARF*ARF*kproteff
kactARF = 3.3E-5 psecReaction: damDNA => damDNA + ARF, Rate Law: kactARF*damDNA
krelMdm2p53 = 1.155E-5 psecReaction: Mdm2_p53 => p53 + Mdm2, Rate Law: krelMdm2p53*Mdm2_p53
kproteff = 1.0 dimensionless; kdegARFMdm2 = 0.001 psecReaction: ARF_Mdm2 => ARF + mdm2deg, Rate Law: kdegARFMdm2*ARF_Mdm2*kproteff
kbinARFMdm2 = 0.01 pmolepsecReaction: ARF + Mdm2 => ARF_Mdm2, Rate Law: kbinARFMdm2*ARF*Mdm2
kproteff = 1.0 dimensionless; kdegMdm2 = 4.33E-4 psecReaction: Mdm2 => Sink + mdm2deg, Rate Law: kdegMdm2*Mdm2*kproteff
ksynMdm2mRNA = 1.0E-4 psecReaction: p53 => p53 + Mdm2_mRNA + Mdm2mRNAsyn, Rate Law: ksynMdm2mRNA*p53

States:

NameDescription
Mdm2mRNAsyn[transcription factor activity, sequence-specific DNA binding]
ARF Mdm2[E3 ubiquitin-protein ligase Mdm2; Tumor suppressor ARF]
damDNA[deoxyribonucleic acid; cellular response to DNA damage stimulus]
Mdm2mRNAdeg[mRNA catabolic process]
mdm2syn[translation]
ARF[CDKN2A; Tumor suppressor ARF]
totp53totp53
p53[Cellular tumor antigen p53; TP53]
totdamDNAtotdamDNA
p53deg[proteasome-mediated ubiquitin-dependent protein catabolic process]
SourceSource
Mdm2 p53[E3 ubiquitin-protein ligase Mdm2; Cellular tumor antigen p53]
p53syn[translation]
totMdm2totMdm2
SinkSink
Mdm2[MDM2; E3 ubiquitin-protein ligase Mdm2]
Mdm2 mRNA[messenger RNA; RNA]
mdm2deg[proteasome-mediated ubiquitin-dependent protein catabolic process]

Proctor2010 - a link between GSK3 and p53 in Alzheimer's Disease: BIOMD0000000286v0.0.1

This is the model described the article: GSK3 and p53 - is there a link in Alzheimer's disease? Carole J Proctor and…

Details

BACKGROUND: Recent evidence suggests that glycogen synthase kinase-3beta (GSK3beta) is implicated in both sporadic and familial forms of Alzheimer's disease. The transcription factor, p53 also plays a role and has been linked to an increase in tau hyperphosphorylation although the effect is indirect. There is also evidence that GSK3beta and p53 interact and that the activity of both proteins is increased as a result of this interaction. Under normal cellular conditions, p53 is kept at low levels by Mdm2 but when cells are stressed, p53 is stabilised and may then interact with GSK3beta. We propose that this interaction has an important contribution to cellular outcomes and to test this hypothesis we developed a stochastic simulation model. RESULTS: The model predicts that high levels of DNA damage leads to increased activity of p53 and GSK3beta and low levels of aggregation but if DNA damage is repaired, the aggregates are eventually cleared. The model also shows that over long periods of time, aggregates may start to form due to stochastic events leading to increased levels of ROS and damaged DNA. This is followed by increased activity of p53 and GSK3beta and a vicious cycle ensues. CONCLUSIONS: Since p53 and GSK3beta are both involved in the apoptotic pathway, and GSK3beta overactivity leads to increased levels of plaques and tangles, our model might explain the link between protein aggregation and neuronal loss in neurodegeneration. link: http://identifiers.org/pubmed/20181016

Parameters:

NameDescription
kactDUBp53 = 1.0E-7Reaction: Mdm2_p53_Ub4 + p53DUB => Mdm2_p53_Ub3 + p53DUB + Ub, Rate Law: kactDUBp53*Mdm2_p53_Ub4*p53DUB
krelMTTau = 1.0E-4Reaction: MT_Tau => Tau, Rate Law: krelMTTau*MT_Tau
krepair = 2.0E-5Reaction: damDNA => Sink, Rate Law: krepair*damDNA
kphosMdm2GSK3bp53 = 0.5Reaction: Mdm2_p53_Ub4 + GSK3b_p53 => Mdm2_P1_p53_Ub4 + GSK3b_p53, Rate Law: kphosMdm2GSK3bp53*Mdm2_p53_Ub4*GSK3b_p53
kaggTauP1 = 1.0E-8Reaction: Tau_P1 => AggTau, Rate Law: kaggTauP1*Tau_P1*(Tau_P1-1)*0.5
kaggTauP2 = 1.0E-7Reaction: Tau_P2 => AggTau, Rate Law: kaggTauP2*Tau_P2*(Tau_P2-1)*0.5
kdephosMdm2 = 0.5Reaction: Mdm2_P => Mdm2, Rate Law: kdephosMdm2*Mdm2_P
kdephosp53 = 0.5Reaction: p53_P => p53, Rate Law: kdephosp53*p53_P
ksynp53mRNAAbeta = 1.0E-5Reaction: Abeta => p53_mRNA + Abeta, Rate Law: ksynp53mRNAAbeta*Abeta
kbinTauProt = 1.925E-7Reaction: Tau + Proteasome => Proteasome_Tau, Rate Law: kbinTauProt*Tau*Proteasome
krelGSK3bp53 = 0.002Reaction: GSK3b_p53 => GSK3b + p53, Rate Law: krelGSK3bp53*GSK3b_p53
kdegTau20SProt = 0.01Reaction: Proteasome_Tau => Proteasome, Rate Law: kdegTau20SProt*Proteasome_Tau
krelMdm2p53 = 1.155E-5Reaction: Mdm2_p53 => p53 + Mdm2, Rate Law: krelMdm2p53*Mdm2_p53
kaggTau = 1.0E-8Reaction: Tau + AggTau => AggTau, Rate Law: kaggTau*Tau*AggTau
kphosp53 = 2.0E-4Reaction: p53 + ATMA => p53_P + ATMA, Rate Law: kphosp53*p53*ATMA
kinactATM = 5.0E-4Reaction: ATMA => ATMI, Rate Law: kinactATM*ATMA
kgenROSAbeta = 1.0E-5Reaction: AggAbeta => AggAbeta + ROS, Rate Law: kgenROSAbeta*AggAbeta
kinhibprot = 1.0E-5Reaction: AggTau + Proteasome => AggTau_Proteasome, Rate Law: kinhibprot*AggTau*Proteasome
kprodAbeta = 5.0E-5Reaction: GSK3b_p53 => Abeta + GSK3b_p53, Rate Law: kprodAbeta*GSK3b_p53
kbinGSK3bp53 = 2.0E-6Reaction: GSK3b + p53_P => GSK3b_p53_P, Rate Law: kbinGSK3bp53*GSK3b*p53_P
kMdm2PolyUb = 0.00456Reaction: Mdm2_Ub2 + E2_Ub => Mdm2_Ub3 + E2, Rate Law: kMdm2PolyUb*Mdm2_Ub2*E2_Ub
ksynMdm2mRNAGSK3bp53 = 7.0E-4Reaction: GSK3b_p53_P => GSK3b_p53_P + Mdm2_mRNA, Rate Law: ksynMdm2mRNAGSK3bp53*GSK3b_p53_P
kbinProt = 2.0E-6Reaction: Mdm2_P1_p53_Ub4 + Proteasome => p53_Ub4_Proteasome + Mdm2, Rate Law: kbinProt*Mdm2_P1_p53_Ub4*Proteasome
kphosMdm2GSK3b = 0.005Reaction: Mdm2_p53_Ub4 + GSK3b => Mdm2_P1_p53_Ub4 + GSK3b, Rate Law: kphosMdm2GSK3b*Mdm2_p53_Ub4*GSK3b
kbinE1Ub = 2.0E-4Reaction: E1 + Ub + ATP => E1_Ub + AMP, Rate Law: kbinE1Ub*E1*Ub*ATP/(5000+ATP)
kdegAbeta = 1.0E-4Reaction: Abeta => Sink, Rate Law: kdegAbeta*Abeta
ksynMdm2 = 4.95E-4Reaction: Mdm2_mRNA => Mdm2_mRNA + Mdm2, Rate Law: ksynMdm2*Mdm2_mRNA
kp53Ub = 5.0E-5Reaction: E2_Ub + Mdm2_p53 => Mdm2_p53_Ub + E2, Rate Law: kp53Ub*E2_Ub*Mdm2_p53
kphospTauGSK3bp53 = 0.1Reaction: GSK3b_p53_P + Tau => GSK3b_p53_P + Tau_P1, Rate Law: kphospTauGSK3bp53*GSK3b_p53_P*Tau
kp53PolyUb = 0.01Reaction: Mdm2_p53_Ub + E2_Ub => Mdm2_p53_Ub2 + E2, Rate Law: kp53PolyUb*Mdm2_p53_Ub*E2_Ub
kproteff = 1.0; kdegp53 = 0.005Reaction: p53_Ub4_Proteasome + ATP => Ub + Proteasome + ADP, Rate Law: kdegp53*kproteff*p53_Ub4_Proteasome*ATP/(5000+ATP)
kdephospTau = 0.01Reaction: Tau_P1 + PP1 => Tau + PP1, Rate Law: kdephospTau*Tau_P1*PP1
ksynTau = 8.0E-5Reaction: Source => Tau, Rate Law: ksynTau*Source
kdam = 0.08Reaction: => damDNA; IR, Rate Law: kdam*IR
ksynMdm2mRNA = 5.0E-4Reaction: p53_P => p53_P + Mdm2_mRNA, Rate Law: ksynMdm2mRNA*p53_P
kMdm2Ub = 4.56E-6Reaction: Mdm2 + E2_Ub => Mdm2_Ub + E2, Rate Law: kMdm2Ub*Mdm2*E2_Ub
kactATM = 1.0E-4Reaction: damDNA + ATMI => damDNA + ATMA, Rate Law: kactATM*damDNA*ATMI
kphospTauGSK3b = 2.0E-4Reaction: GSK3b + Tau => GSK3b + Tau_P1, Rate Law: kphospTauGSK3b*GSK3b*Tau
kdegMdm2 = 0.01; kproteff = 1.0Reaction: Mdm2_Ub4_Proteasome => Proteasome + Ub, Rate Law: kdegMdm2*Mdm2_Ub4_Proteasome*kproteff
kactDUBMdm2 = 1.0E-7Reaction: Mdm2_Ub2 + Mdm2DUB => Mdm2_Ub + Mdm2DUB + Ub, Rate Law: kactDUBMdm2*Mdm2_Ub2*Mdm2DUB
kpf = 0.001Reaction: AggAbeta + AbetaPlaque => AbetaPlaque, Rate Law: kpf*AggAbeta*AbetaPlaque
kaggAbeta = 1.0E-8Reaction: Abeta => AggAbeta, Rate Law: kaggAbeta*Abeta*(Abeta-1)*0.5
kphosMdm2 = 2.0Reaction: Mdm2 + ATMA => Mdm2_P + ATMA, Rate Law: kphosMdm2*Mdm2*ATMA
ksynp53mRNA = 0.001Reaction: Source => p53_mRNA, Rate Law: ksynp53mRNA*Source
kMdm2PUb = 6.84E-6Reaction: Mdm2_P + E2_Ub => Mdm2_P_Ub + E2, Rate Law: kMdm2PUb*Mdm2_P*E2_Ub
kdegp53mRNA = 1.0E-4Reaction: p53_mRNA => Sink, Rate Law: kdegp53mRNA*p53_mRNA
ktangfor = 0.001Reaction: AggTau => NFT, Rate Law: ktangfor*AggTau*(AggTau-1)*0.5
kbinMTTau = 0.1Reaction: Tau => MT_Tau, Rate Law: kbinMTTau*Tau
ksynp53 = 0.007Reaction: p53_mRNA => p53 + p53_mRNA, Rate Law: ksynp53*p53_mRNA
kdegMdm2mRNA = 5.0E-4Reaction: Mdm2_mRNA => Sink, Rate Law: kdegMdm2mRNA*Mdm2_mRNA

States:

NameDescription
Mdm2 P[E3 ubiquitin-protein ligase Mdm2]
AggAbeta[Amyloid beta A4 protein]
AggTau Proteasome[IPR002955; proteasome complex]
ATP[ATP]
AbetaPlaque[Amyloid beta A4 protein]
MT Tau[IPR015562]
Ub[Ubiquitin-60S ribosomal protein L40]
Proteasome Tau[IPR002955; proteasome complex]
Mdm2 P Ub3[E3 ubiquitin-protein ligase Mdm2; Ubiquitin-60S ribosomal protein L40]
AMP[AMP; AMP]
Mdm2 Ub2[E3 ubiquitin-protein ligase Mdm2; Ubiquitin-60S ribosomal protein L40]
p53[Cellular tumor antigen p53]
SourceSource
p53 P[Cellular tumor antigen p53]
AggAbeta Proteasome[Amyloid beta A4 protein; proteasome complex]
IRIR
E2 Ub[Ubiquitin-60S ribosomal protein L40; IPR000608]
GSK3b p53 P[Glycogen synthase kinase-3 beta; Cellular tumor antigen p53]
Abeta[Amyloid beta A4 protein]
Mdm2[E3 ubiquitin-protein ligase Mdm2]
Mdm2DUB[IPR001394]
Tau P1[IPR002955]
ROS[reactive oxygen species]
GSK3b p53[Glycogen synthase kinase-3 beta; Cellular tumor antigen p53]
AggTau[IPR002955]
Tau[IPR002955]
PP1[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit]
damDNA[deoxyribonucleic acid]
Mdm2 P Ub2[E3 ubiquitin-protein ligase Mdm2; Ubiquitin-60S ribosomal protein L40]
Mdm2 p53 Ub[E3 ubiquitin-protein ligase Mdm2; Cellular tumor antigen p53; Ubiquitin-60S ribosomal protein L40]
Mdm2 p53 Ub3[E3 ubiquitin-protein ligase Mdm2; Cellular tumor antigen p53; Ubiquitin-60S ribosomal protein L40]
Mdm2 p53[E3 ubiquitin-protein ligase Mdm2; Cellular tumor antigen p53]
NFT[IPR002955]
Mdm2 Ub[E3 ubiquitin-protein ligase Mdm2; Ubiquitin-60S ribosomal protein L40]
ATMI[Serine-protein kinase ATM]
Mdm2 p53 Ub4[E3 ubiquitin-protein ligase Mdm2; Cellular tumor antigen p53; Ubiquitin-60S ribosomal protein L40]
ADP[ADP]
Tau P2[IPR002955]
SinkSink
Mdm2 p53 Ub2[E3 ubiquitin-protein ligase Mdm2; Cellular tumor antigen p53; Ubiquitin-60S ribosomal protein L40]

Proctor2010 - UCHL1 Protein Aggregation: BIOMD0000000293v0.0.1

This a model from the article: Modelling the Role of UCH-L1 on Protein Aggregation in Age-Related Neurodegeneration.…

Details

Overexpression of the de-ubiquitinating enzyme UCH-L1 leads to inclusion formation in response to proteasome impairment. These inclusions contain components of the ubiquitin-proteasome system and α-synuclein confirming that the ubiquitin-proteasome system plays an important role in protein aggregation. The processes involved are very complex and so we have chosen to take a systems biology approach to examine the system whereby we combine mathematical modelling with experiments in an iterative process. The experiments show that cells are very heterogeneous with respect to inclusion formation and so we use stochastic simulation. The model shows that the variability is partly due to stochastic effects but also depends on protein expression levels of UCH-L1 within cells. The model also indicates that the aggregation process can start even before any proteasome inhibition is present, but that proteasome inhibition greatly accelerates aggregation progression. This leads to less efficient protein degradation and hence more aggregation suggesting that there is a vicious cycle. However, proteasome inhibition may not necessarily be the initiating event. Our combined modelling and experimental approach show that stochastic effects play an important role in the aggregation process and could explain the variability in the age of disease onset. Furthermore, our model provides a valuable tool, as it can be easily modified and extended to incorporate new experimental data, test hypotheses and make testable predictions. link: http://identifiers.org/pubmed/20949132

Parameters:

NameDescription
kpolyUb = 0.01Reaction: E3_MisP_Ub6 + E2_Ub => E3_MisP_Ub7 + E2, Rate Law: kpolyUb*E3_MisP_Ub6*E2_Ub
kbinAggProt = 5.0E-9Reaction: AggA1 + Proteasome => AggP_Proteasome, Rate Law: kbinAggProt*AggA1*Proteasome
kdisaggasyn3 = 6.0E-9Reaction: AggA3 => AggA2 + asyn, Rate Law: kdisaggasyn3*AggA3
kgenROSAggP = 2.0E-5Reaction: AggP5 => AggP5 + ROS, Rate Law: kgenROSAggP*AggP5
kdisaggasyn5 = 2.0E-9Reaction: AggA5 => AggA4 + asyn, Rate Law: kdisaggasyn5*AggA5
kbinProt = 5.0E-6Reaction: Parkin_asyn_dam_Ub7 + Proteasome => asyn_dam_Ub7_Proteasome + Parkin, Rate Law: kbinProt*Parkin_asyn_dam_Ub7*Proteasome
kdisagg2 = 8.0E-9Reaction: AggP2 => AggP1 + MisP, Rate Law: kdisagg2*AggP2
kactDUB = 1.0E-4Reaction: Parkin_asyn_dam_Ub2_DUB => Parkin_asyn_dam_Ub_DUB + Ub, Rate Law: kactDUB*Parkin_asyn_dam_Ub2_DUB
kdisaggasyn2 = 8.0E-9Reaction: AggA2 => AggA1 + asyn, Rate Law: kdisaggasyn2*AggA2
kbinasynDUB = 2.0E-7Reaction: Parkin_asyn_dam_Ub7 + DUB => Parkin_asyn_dam_Ub7_DUB, Rate Law: kbinasynDUB*Parkin_asyn_dam_Ub7*DUB
krelMisPE3 = 2.0E-4Reaction: E3_MisP => MisP + E3, Rate Law: krelMisPE3*E3_MisP
kbinSUBUCHL1 = 4.0E-8Reaction: E3SUB_SUB_misfolded_Ub2 + UCHL1 => E3SUB_SUB_misfolded_Ub2_UCHL1, Rate Law: kbinSUBUCHL1*E3SUB_SUB_misfolded_Ub2*UCHL1
kaggasyn2 = 5.0E-10Reaction: asyn + AggA2 => AggA3, Rate Law: kaggasyn2*asyn*AggA2
kactDUBProt = 1.0E-6Reaction: SUB_misfolded_Ub4_Proteasome + DUB => SUB_misfolded + Proteasome + Ub + DUB, Rate Law: kactDUBProt*SUB_misfolded_Ub4_Proteasome*DUB
kactProt = 0.01; kproteff = 1.0Reaction: SUB_misfolded_Ub7_Proteasome + ATP => Ub + Proteasome + ADP, Rate Law: kactProt*SUB_misfolded_Ub7_Proteasome*kproteff*ATP/(5000+ATP)
kbinE2Ub = 0.001Reaction: E2 + E1_Ub => E2_Ub + E1, Rate Law: kbinE2Ub*E2*E1_Ub
kigrowth2 = 5.0E-9Reaction: E3_MisP_Ub6 + SeqAggP => SeqAggP + aggMisP + aggUb + aggE3, Rate Law: kigrowth2*E3_MisP_Ub6*SeqAggP
kremROS = 0.001Reaction: ROS => Sink, Rate Law: kremROS*ROS
kactUchl1 = 1.0E-4Reaction: E3SUB_SUB_misfolded_Ub3_UCHL1 => E3SUB_SUB_misfolded_Ub2_UCHL1 + Ub, Rate Law: kactUchl1*E3SUB_SUB_misfolded_Ub3_UCHL1
kubss = 0.1Reaction: MisP => MisP + Ub + upregUb, Rate Law: kubss*MisP^6/(1500^6+MisP^6)
kbinMisPE3 = 1.0E-4Reaction: MisP + E3 => E3_MisP, Rate Law: kbinMisPE3*MisP*E3

States:

NameDescription
aggMisPaggMisP
Ub[Ubiquitin-60S ribosomal protein L40]
SeqAggPSeqAggP
aggUb[Ubiquitin-60S ribosomal protein L40]
MisP[protein]
AggP5AggP5
AggA3[Alpha-synuclein]
E3 MisP Ub7[protein; Ubiquitin-60S ribosomal protein L40; IPR000569]
Parkin asyn dam Ub6[E3 ubiquitin-protein ligase parkin; Alpha-synuclein; Ubiquitin-60S ribosomal protein L40]
aggE3[IPR000569]
E3SUB SUB misfolded Ub3 UCHL1[Ubiquitin carboxyl-terminal hydrolase isozyme L1; Ubiquitin-60S ribosomal protein L40; IPR000569]
AggA4[Alpha-synuclein]
aggDUB[IPR001394]
E2 Ub[Ubiquitin-60S ribosomal protein L40; IPR000608]
E3 MisP Ub8[protein; Ubiquitin-60S ribosomal protein L40; IPR000569]
ROS[reactive oxygen species]
Parkin asyn dam Ub7[E3 ubiquitin-protein ligase parkin; Alpha-synuclein; Ubiquitin-60S ribosomal protein L40]
E3SUB SUB misfolded Ub2 UCHL1[Ubiquitin carboxyl-terminal hydrolase isozyme L1; Ubiquitin-60S ribosomal protein L40; IPR000569]
Parkin asyn dam Ub DUB[E3 ubiquitin-protein ligase parkin; Alpha-synuclein; Ubiquitin-60S ribosomal protein L40]
E3[IPR000569]
E2[IPR000608]
AggA1[Alpha-synuclein]
AggA5[Alpha-synuclein]
E3 MisP Ub6[protein; Ubiquitin-60S ribosomal protein L40; IPR000569]
Parkin asyn dam Ub8[E3 ubiquitin-protein ligase parkin; Alpha-synuclein; Ubiquitin-60S ribosomal protein L40]
Parkin asyn dam Ub2 DUB[E3 ubiquitin-protein ligase parkin; Alpha-synuclein; Ubiquitin-60S ribosomal protein L40]
AggA2[Alpha-synuclein]

Proctor2011_ProteinHomeostasis_NormalCondition: BIOMD0000000344v0.0.1

This model is from the article: Modelling the Role of the Hsp70/Hsp90 System in the Maintenance of Protein Homeostas…

Details

Neurodegeneration is an age-related disorder which is characterised by the accumulation of aggregated protein and neuronal cell death. There are many different neurodegenerative diseases which are classified according to the specific proteins involved and the regions of the brain which are affected. Despite individual differences, there are common mechanisms at the sub-cellular level leading to loss of protein homeostasis. The two central systems in protein homeostasis are the chaperone system, which promotes correct protein folding, and the cellular proteolytic system, which degrades misfolded or damaged proteins. Since these systems and their interactions are very complex, we use mathematical modelling to aid understanding of the processes involved. The model developed in this study focuses on the role of Hsp70 (IPR00103) and Hsp90 (IPR001404) chaperones in preventing both protein aggregation and cell death. Simulations were performed under three different conditions: no stress; transient stress due to an increase in reactive oxygen species; and high stress due to sustained increases in reactive oxygen species. The model predicts that protein homeostasis can be maintained during short periods of stress. However, under long periods of stress, the chaperone system becomes overwhelmed and the probability of cell death pathways being activated increases. Simulations were also run in which cell death mediated by the JNK (P45983) and p38 (Q16539) pathways was inhibited. The model predicts that inhibiting either or both of these pathways may delay cell death but does not stop the aggregation process and that eventually cells die due to aggregated protein inhibiting proteasomal function. This problem can be overcome if the sequestration of aggregated protein into inclusion bodies is enhanced. This model predicts responses to reactive oxygen species-mediated stress that are consistent with currently available experimental data. The model can be used to assess specific interventions to reduce cell death due to impaired protein homeostasis. link: http://identifiers.org/pubmed/21779370

Parameters:

NameDescription
kdegHsp90 = 0.01; kalive = 1.0Reaction: Hsp90_Proteasome + ATP => Proteasome + ADP, Rate Law: kdegHsp90*Hsp90_Proteasome*kalive*ATP/(5000+ATP)
kalive = 1.0; kdephosp38Mkp1 = 0.05Reaction: p38_P + Mkp1_P => p38 + Mkp1_P, Rate Law: kdephosp38Mkp1*p38_P*Mkp1_P*kalive
kdegMkp1 = 0.01; kalive = 1.0Reaction: Mkp1_Proteasome + ATP => Proteasome + ADP, Rate Law: kdegMkp1*Mkp1_Proteasome*kalive*ATP/(5000+ATP)
kalive = 1.0; kbinHspMisp = 8.0E-6Reaction: MisP + Hsp70 => Hsp70_MisP, Rate Law: kbinHspMisp*MisP*Hsp70*kalive
kdephosJnkMkp1 = 0.05; kalive = 1.0Reaction: Jnk_P + Mkp1_P => Jnk + Mkp1_P, Rate Law: kdephosJnkMkp1*Jnk_P*Mkp1_P*kalive
kalive = 1.0; kbinAggPProt = 1.0E-5Reaction: AggP + Proteasome => AggP_Proteasome, Rate Law: kbinAggPProt*AggP*Proteasome*kalive
kgenROS = 0.01; kalive = 1.0Reaction: Source => ROS, Rate Law: kgenROS*Source*kalive
kbinMisPProt = 1.0E-7; kalive = 1.0Reaction: Hsp70_MisP + Proteasome => MisP_Proteasome + Hsp70, Rate Law: kbinMisPProt*Hsp70_MisP*Proteasome*kalive
kalive = 1.0; kbinHsp90client = 2.0E-4Reaction: Hsp90 + Hsp90Client => Hsp90_Hsp90Client, Rate Law: kbinHsp90client*Hsp90*Hsp90Client*kalive
kalive = 1.0; kdegAkt = 0.01Reaction: Akt_Proteasome + ATP => Proteasome + ADP, Rate Law: kdegAkt*Akt_Proteasome*kalive*ATP/(5000+ATP)
kPIdeath = 2.0E-8; kalive = 1.0Reaction: AggP_Proteasome => AggP_Proteasome + PIDeath + CellDeath, Rate Law: kPIdeath*AggP_Proteasome*kalive
kalive = 1.0; krelHsp70Ppx = 5.0Reaction: Hsp70_Ppx => Hsp70 + Ppx, Rate Law: krelHsp70Ppx*Hsp70_Ppx*kalive
kalive = 1.0; krelAktProt = 1.0E-8Reaction: Akt_Proteasome => Akt + Proteasome, Rate Law: krelAktProt*Akt_Proteasome*kalive
kagg = 1.0E-8; kalive = 1.0Reaction: MisP => AggP, Rate Law: kagg*MisP*(MisP-1)*0.5*kalive
kphosMkp1 = 0.02; kalive = 1.0Reaction: Mkp1 + Hsp70 => Mkp1_P + Hsp70, Rate Law: kphosMkp1*Mkp1*Hsp70*kalive
kbinHsp90Prot = 1.0E-8; kalive = 1.0Reaction: Hsp90 + Proteasome => Hsp90_Proteasome, Rate Law: kbinHsp90Prot*Hsp90*Proteasome*kalive
kalive = 1.0; kdegMisP = 0.01Reaction: MisP_Proteasome + ATP => Proteasome + ADP, Rate Law: kdegMisP*MisP_Proteasome*kalive*ATP/(5000+ATP)
kgenROSp38 = 1.0E-4; kalive = 1.0; kp38act = 1.0Reaction: p38_P => p38_P + ROS, Rate Law: kgenROSp38*p38_P*kalive*kp38act
kmisfold = 2.0E-6; kalive = 1.0Reaction: NatP + ROS => MisP + ROS, Rate Law: kmisfold*NatP*ROS*kalive
kbinHsp70client = 2.0E-4; kalive = 1.0Reaction: Hsp70 + Hsp70Client => Hsp70_Hsp70Client, Rate Law: kbinHsp70client*Hsp70*Hsp70Client*kalive
kbinHsp70Ppx = 0.2; kalive = 1.0Reaction: Hsp70 + Ppx => Hsp70_Ppx, Rate Law: kbinHsp70Ppx*Hsp70*Ppx*kalive
ksynMkp1 = 1.0E-5; kalive = 1.0Reaction: Source => Mkp1, Rate Law: ksynMkp1*Source*kalive
kbinHsp70Prot = 1.2E-8; kalive = 1.0Reaction: Hsp70 + Proteasome => Hsp70_Proteasome, Rate Law: kbinHsp70Prot*Hsp70*Proteasome*kalive
krefold = 5.5E-4; kalive = 1.0Reaction: Hsp90_MisP + ATP => Hsp90 + NatP + ADP, Rate Law: krefold*Hsp90_MisP*kalive*ATP/(5000+ATP)
kbasalsynHsp70 = 0.008; kalive = 1.0Reaction: Source => Hsp70, Rate Law: kbasalsynHsp70*kalive
kalive = 1.0; kphosp38 = 0.02Reaction: ROS + p38 => ROS + p38_P, Rate Law: kphosp38*ROS*p38*kalive
kphosHsf1 = 0.03; kalive = 1.0Reaction: Hsf1_Hsf1_Hsf1 + Pkc => Hsf1_Hsf1_Hsf1_P + Pkc, Rate Law: kphosHsf1*Hsf1_Hsf1_Hsf1*Pkc*kalive
kbinMkp1Prot = 9.6E-9; kalive = 1.0Reaction: Mkp1 + Proteasome => Mkp1_Proteasome, Rate Law: kbinMkp1Prot*Mkp1*Proteasome*kalive
kalive = 1.0; kgenROSAggP = 1.0E-6Reaction: AggP => AggP + ROS, Rate Law: kgenROSAggP*AggP*kalive
kphosJnk = 0.02; kalive = 1.0Reaction: ROS + Jnk => ROS + Jnk_P, Rate Law: kphosJnk*Jnk*ROS*kalive
kbinAktProt = 6.0E-8; kalive = 1.0Reaction: Akt_CHIP_Hsp90 + Proteasome => Akt_Proteasome + CHIP + Hsp90, Rate Law: kbinAktProt*Akt_CHIP_Hsp90*Proteasome*kalive
kalive = 1.0; kp38death = 1.5E-7; kp38act = 1.0Reaction: p38_P => p38_P + p38Death + CellDeath, Rate Law: kp38death*p38_P*kalive*kp38act
kalive = 1.0; kdamHsp = 1.0E-8Reaction: Hsp70 + ROS => Hsp70_dam + ROS, Rate Law: kdamHsp*Hsp70*ROS*kalive
kbinHsf1Hsp90 = 0.02; kalive = 1.0Reaction: Hsp90 + Hsf1 => Hsf1_Hsp90, Rate Law: kbinHsf1Hsp90*Hsp90*Hsf1*kalive
krelHsp70client = 5.0; kalive = 1.0Reaction: Hsp70_Hsp70Client => Hsp70 + Hsp70Client, Rate Law: krelHsp70client*Hsp70_Hsp70Client*kalive
kremROS = 0.001; kalive = 1.0Reaction: ROS => Sink, Rate Law: kremROS*ROS*kalive
kalive = 1.0; kJnkdeath = 1.5E-7Reaction: Jnk_P => Jnk_P + JNKDeath + CellDeath, Rate Law: kJnkdeath*Jnk_P*kalive
krelHsf1Hsp90 = 0.5; kalive = 1.0Reaction: Hsf1_Hsp90 => Hsp90 + Hsf1, Rate Law: krelHsf1Hsp90*Hsf1_Hsp90*kalive
kdegHsp70 = 0.01; kalive = 1.0Reaction: Hsp70_Proteasome + ATP => Proteasome + ADP, Rate Law: kdegHsp70*Hsp70_Proteasome*kalive*ATP/(5000+ATP)
kdephosHsf1 = 0.01; kalive = 1.0Reaction: Hsf1_Hsf1_Hsf1_P + Hsp70_Ppx => Hsf1_Hsf1_Hsf1 + Hsp70_Ppx, Rate Law: kdephosHsf1*Hsf1_Hsf1_Hsf1_P*Hsp70_Ppx*kalive
kupregHsp = 0.2; kalive = 1.0Reaction: HSEHsp70_Hsf1_Hsf1_Hsf1_P => HSEHsp70_Hsf1_Hsf1_Hsf1_P + Hsp70, Rate Law: kupregHsp*HSEHsp70_Hsf1_Hsf1_Hsf1_P*kalive
kalive = 1.0; kdephosMkp1 = 0.001Reaction: Mkp1_P + ROS => Mkp1 + ROS, Rate Law: kdephosMkp1*Mkp1_P*ROS*kalive
kseqagg = 7.0E-7; kalive = 1.0Reaction: SeqAggP + MisP => SeqAggP, Rate Law: kseqagg*SeqAggP*MisP*kalive
krelHsp90client = 5.0; kalive = 1.0Reaction: Hsp90_Hsp90Client => Hsp90 + Hsp90Client, Rate Law: krelHsp90client*Hsp90_Hsp90Client*kalive
kalive = 1.0; krelHspMisp = 8.0E-5Reaction: Hsp90_MisP => MisP + Hsp90, Rate Law: krelHspMisp*Hsp90_MisP*kalive

States:

NameDescription
Proteasome[proteasome complex]
Mkp1[Dual specificity protein phosphatase 1]
ATP[ATP]
Jnk[Mitogen-activated protein kinase 8]
Jnk P[Mitogen-activated protein kinase 8; Phosphoprotein]
ROS[reactive oxygen species]
AggP Proteasome[protein; proteasome complex]
p38 P[Mitogen-activated protein kinase 14; Phosphoprotein]
Hsp90[IPR001404]
p38[Mitogen-activated protein kinase 14]
SeqAggP[protein]
MisP[protein]
Mkp1 P[Dual specificity protein phosphatase 1; Phosphoprotein]
Hsp70[IPR001023]
Pkc[IPR012233]
MisP Proteasome[protein; proteasome complex]
Ppx[protein serine/threonine phosphatase complex]
NatP[protein]
Hsp70 Ppx[IPR001023; protein serine/threonine phosphatase complex]
AggP[protein]
Mkp1 Proteasome[Dual specificity protein phosphatase 1; proteasome complex]

Proctor2012 - Role of Amyloid-beta dimers in aggregation formation: BIOMD0000000462v0.0.1

Proctor2012 - Amyloid-beta aggregationThis model supports the current thinking that levels of dimers are important in in…

Details

Alzheimer's disease (AD) is the most frequently diagnosed neurodegenerative disorder affecting humans, with advanced age being the most prominent risk factor for developing AD. Despite intense research efforts aimed at elucidating the precise molecular underpinnings of AD, a definitive answer is still lacking. In recent years, consensus has grown that dimerisation of the polypeptide amyloid-beta (Aß), particularly Aß₄₂, plays a crucial role in the neuropathology that characterise AD-affected post-mortem brains, including the large-scale accumulation of fibrils, also referred to as senile plaques. This has led to the realistic hope that targeting Aß₄₂ immunotherapeutically could drastically reduce plaque burden in the ageing brain, thus delaying AD onset or symptom progression. Stochastic modelling is a useful tool for increasing understanding of the processes underlying complex systems-affecting disorders such as AD, providing a rapid and inexpensive strategy for testing putative new therapies. In light of the tool's utility, we developed computer simulation models to examine Aß₄₂ turnover and its aggregation in detail and to test the effect of immunization against Aß dimers.Our model demonstrates for the first time that even a slight decrease in the clearance rate of Aß₄₂ monomers is sufficient to increase the chance of dimers forming, which could act as instigators of protofibril and fibril formation, resulting in increased plaque levels. As the process is slow and levels of Aβ are normally low, stochastic effects are important. Our model predicts that reducing the rate of dimerisation leads to a significant reduction in plaque levels and delays onset of plaque formation. The model was used to test the effect of an antibody mediated immunological response. Our results showed that plaque levels were reduced compared to conditions where antibodies are not present.Our model supports the current thinking that levels of dimers are important in initiating the aggregation process. Although substantial knowledge exists regarding the process, no therapeutic intervention is on offer that reliably decreases disease burden in AD patients. Computer modelling could serve as one of a number of tools to examine both the validity of reliable biomarkers and aid the discovery of successful intervention strategies. link: http://identifiers.org/pubmed/22748062

Parameters:

NameDescription
kdimer = 1.1783E-7Reaction: Abeta => AbDim; Abeta, Rate Law: kdimer*Abeta*(Abeta-1)*0.5
kpf = 2.785E-6Reaction: AbDim => AbP; AbDim, Rate Law: kpf*AbDim*(AbDim-1)*0.5
kdegNep = 1.8E-10Reaction: Nep => Sink; Nep, Rate Law: kdegNep*Nep
kdedimer = 8.4655E-6Reaction: AbDim => Abeta; AbDim, Rate Law: kdedimer*AbDim
kprod = 1.86E-5Reaction: Source => Abeta; Source, Rate Law: kprod*Source
kdisagg = 5.4357E-5Reaction: AbP => Abeta; AbP, Rate Law: kdisagg*AbP
kdeg = 2.1E-5Reaction: Abeta + Nep => Sink + Nep; Abeta, Nep, Rate Law: kdeg*Abeta*Nep*0.001
kpg = 0.00574; kpghalf = 4.0Reaction: Abeta + AbP => AbP; Abeta, AbP, Rate Law: kpg*Abeta*AbP^2/(kpghalf^2+AbP^2)

States:

NameDescription
AbP[amyloid-beta; amyloid plaque]
SourceAbetaPlaque
Nep[Neprilysin]
Abeta[amyloid-beta]
SinkAbetaPlaque
AbDim[amyloid-beta; protein complex]

Proctor2013 - Cartilage breakdown, interventions to reduce collagen release: BIOMD0000000504v0.0.1

Proctor2013 - Cartilage breakdown, interventions to reduce collagen releaseThe molecular pathways involved in cartilage…

Details

Objective. To use a novel computational approach to examine the molecular pathways involved in cartilage breakdown and to use computer simulation to test possible interventions to reduce collagen release. Methods. We constructed a computational model of the relevant molecular pathways using the Systems Biology Markup Language (SBML), a computer-readable format of a biochemical network. The model was constructed using our experimental data showing that interleukin-1 (IL-1) and oncostatin M (OSM) act synergistically to up-regulate collagenase protein and activity and initiate cartilage collagen breakdown. Simulations were performed in the COPASI software package. Results. The model predicted that simulated inhibition of c-Jun N-terminal kinase (JNK) or p38 mitogen-activated protein kinase, and over-expression of tissue inhibitor of metalloproteinases 3 (TIMP-3) led to a reduction in collagen release. Over-expression of TIMP-1 was much less effective than TIMP-3 and led to a delay, rather than a reduction, in collagen release. Simulated interventions of receptor antagonists and inhibition of Janus kinase 1 (JAK1), the first kinase in the OSM pathway, were ineffective. So, importantly, the model predicts that it is more effective to intervene at targets which are downstream, such as the JNK pathway, rather than close to the cytokine signal. In vitro experiments confirmed the effectiveness of JNK inhibition. Conclusion. Our study shows the value of computer modelling as a tool for examining possible interventions to reduce cartilage collagen breakdown. The model predicts interventions that either prevent transcription or inhibit activity of collagenases are promising strategies and should be investigated further in an experimental setting. © 2013 American College of Rheumatology. link: http://identifiers.org/pubmed/24285357

Parameters:

NameDescription
kdegMKP1 = 1.0E-4Reaction: MKP1 => Sink; MKP1, Rate Law: kdegMKP1*MKP1
kdegADAMTS4 = 5.0E-5Reaction: ADAMTS4 => Sink; ADAMTS4, Rate Law: kdegADAMTS4*ADAMTS4
ksynDUSP16 = 0.005; kAP1activity = 1.0Reaction: cFos_cJun => cFos_cJun + DUSP16; cFos_cJun, Rate Law: ksynDUSP16*cFos_cJun*kAP1activity
ksynMMP1 = 1.5E-4Reaction: MMP1_mRNA => MMP1_mRNA + proMMP1; MMP1_mRNA, Rate Law: ksynMMP1*MMP1_mRNA
ksyncFosmRNAStat3 = 0.05Reaction: STAT3_P_nuc => STAT3_P_nuc + cFos_mRNA; STAT3_P_nuc, Rate Law: ksyncFosmRNAStat3*STAT3_P_nuc
kdephosJNKDUSP16 = 0.001Reaction: JNK_P + DUSP16 => JNK + DUSP16; JNK_P, DUSP16, Rate Law: kdephosJNKDUSP16*JNK_P*DUSP16
ksynTIMP3mRNAStat3 = 4.0E-5; kAP1activity = 1.0Reaction: STAT3_P_nuc => STAT3_P_nuc + TIMP3_mRNA; STAT3_P_nuc, Rate Law: ksynTIMP3mRNAStat3*STAT3_P_nuc*kAP1activity
kdephoscFosDUSP16 = 1.0E-4Reaction: cFos_P + DUSP16 => cFos + DUSP16; cFos_P, DUSP16, Rate Law: kdephoscFosDUSP16*cFos_P*DUSP16
krelTRAF6PP4 = 1.0E-6Reaction: TRAF6_PP4 => TRAF6 + PP4; TRAF6_PP4, Rate Law: krelTRAF6PP4*TRAF6_PP4
ksynPTPRT = 1.0E-4Reaction: STAT3_P_nuc => STAT3_P_nuc + PTPRT; STAT3_P_nuc, Rate Law: ksynPTPRT*STAT3_P_nuc
kcyt2nucSTAT3 = 0.001Reaction: STAT3_P_cyt => STAT3_P_nuc; STAT3_P_cyt, Rate Law: kcyt2nucSTAT3*STAT3_P_cyt
ksynSOCS3 = 0.001Reaction: SOCS3_mRNA => SOCS3_mRNA + SOCS3; SOCS3_mRNA, Rate Law: ksynSOCS3*SOCS3_mRNA
kphosSTAT3 = 0.005Reaction: STAT3_cyt + JAK1_P => STAT3_P_cyt + JAK1_P; STAT3_cyt, JAK1_P, Rate Law: kphosSTAT3*STAT3_cyt*JAK1_P
kbinTRAF6 = 1.0E-5Reaction: IL1_IL1R_IRAK2 + TRAF6 => IL1_IL1R + IRAK2_TRAF6; IL1_IL1R_IRAK2, TRAF6, Rate Law: kbinTRAF6*IL1_IL1R_IRAK2*TRAF6
kdegDUSP16 = 1.3E-4Reaction: DUSP16 => Sink; DUSP16, Rate Law: kdegDUSP16*DUSP16
ksynbasalTIMP3mRNA = 2.8E-4Reaction: Source => TIMP3_mRNA; Source, Rate Law: ksynbasalTIMP3mRNA*Source
kdegcJun = 1.3E-4Reaction: cJun => Sink; cJun, Rate Law: kdegcJun*cJun
krelMMP1 = 0.001Reaction: MMP1_TIMP1 => MMP1 + TIMP1; MMP1_TIMP1, Rate Law: krelMMP1*MMP1_TIMP1
kdephosSTAT3nucPTPRT = 5.0E-4Reaction: STAT3_P_nuc + PTPRT => STAT3_nuc + PTPRT; STAT3_P_nuc, PTPRT, Rate Law: kdephosSTAT3nucPTPRT*STAT3_P_nuc*PTPRT
krelADAMTS4TIMP1 = 0.001Reaction: ADAMTS4_TIMP1 => ADAMTS4 + TIMP1; ADAMTS4_TIMP1, Rate Law: krelADAMTS4TIMP1*ADAMTS4_TIMP1
ksynTIMP1mRNAStat3 = 4.0E-5Reaction: STAT3_P_nuc + TIMP1_DNA => STAT3_P_nuc + TIMP1_DNA + TIMP1_mRNA; STAT3_P_nuc, TIMP1_DNA, Rate Law: ksynTIMP1mRNAStat3*STAT3_P_nuc*TIMP1_DNA
kdephosSTAT3nuc = 1.0E-7Reaction: STAT3_P_nuc => STAT3_nuc; STAT3_P_nuc, Rate Law: kdephosSTAT3nuc*STAT3_P_nuc
ksynSOCS3mRNA = 0.006Reaction: STAT3_P_nuc => STAT3_P_nuc + SOCS3_mRNA; STAT3_P_nuc, Rate Law: ksynSOCS3mRNA*STAT3_P_nuc
ksynDUSP16cJun = 2.0E-4Reaction: cJun_dimer => cJun_dimer + DUSP16; cJun_dimer, Rate Law: ksynDUSP16cJun*cJun_dimer
ksynADAMTS4 = 5.0E-4Reaction: ADAMTS4_mRNA => ADAMTS4_mRNA + ADAMTS4; ADAMTS4_mRNA, Rate Law: ksynADAMTS4*ADAMTS4_mRNA
kphoscJun = 1.0E-4Reaction: cJun + JNK_P => cJun_P + JNK_P; cJun, JNK_P, Rate Law: kphoscJun*cJun*JNK_P
kdegAggrecan = 3.0E-8Reaction: Aggrecan_Collagen2 + ADAMTS4 => ADAMTS4 + Collagen2 + AggFrag; Aggrecan_Collagen2, ADAMTS4, Rate Law: kdegAggrecan*Aggrecan_Collagen2*ADAMTS4
kdephosp38 = 0.001Reaction: p38_P => p38; p38_P, Rate Law: kdephosp38*p38_P
ksynTIMP1 = 2.0E-4Reaction: TIMP1_mRNA => TIMP1_mRNA + TIMP1; TIMP1_mRNA, Rate Law: ksynTIMP1*TIMP1_mRNA
kdegPTPRT = 5.0E-5Reaction: PTPRT => Sink; PTPRT, Rate Law: kdegPTPRT*PTPRT
kdegSOCS3mRNA = 4.0E-4Reaction: SOCS3_mRNA => Sink; SOCS3_mRNA, Rate Law: kdegSOCS3mRNA*SOCS3_mRNA
ksynbasalTIMP1mRNA = 1.4E-4Reaction: TIMP1_DNA => TIMP1_mRNA + TIMP1_DNA; TIMP1_DNA, Rate Law: ksynbasalTIMP1mRNA*TIMP1_DNA
kdegTIMP3 = 2.0E-5Reaction: TIMP3 => Sink; TIMP3, Rate Law: kdegTIMP3*TIMP3
kinhibADAMTS4TIMP1 = 3.0E-6Reaction: ADAMTS4 + TIMP1 => ADAMTS4_TIMP1; ADAMTS4, TIMP1, Rate Law: kinhibADAMTS4TIMP1*ADAMTS4*TIMP1
kdegMMP13mRNA = 6.4E-6Reaction: MMP13_mRNA => Sink; MMP13_mRNA, Rate Law: kdegMMP13mRNA*MMP13_mRNA
ksyncJunmRNAcJun = 0.005Reaction: cJun_dimer => cJun_mRNA + cJun_dimer; cJun_dimer, Rate Law: ksyncJunmRNAcJun*cJun_dimer
kphosJAK1 = 1.0E-5Reaction: JAK1 + OSM_OSMR => JAK1_P + OSM_OSMR; JAK1, OSM_OSMR, Rate Law: kphosJAK1*JAK1*OSM_OSMR
kdegTIMP1mRNA = 1.4E-5Reaction: TIMP1_mRNA => Sink; TIMP1_mRNA, Rate Law: kdegTIMP1mRNA*TIMP1_mRNA
kdegTIMP3mRNA = 1.4E-5Reaction: TIMP3_mRNA => Sink; TIMP3_mRNA, Rate Law: kdegTIMP3mRNA*TIMP3_mRNA
kinhibMMP1TIMP3 = 1.0E-8Reaction: MMP1 + TIMP3 => MMP1_TIMP3; MMP1, TIMP3, Rate Law: kinhibMMP1TIMP3*MMP1*TIMP3
kdephosSTAT3 = 1.0E-5Reaction: STAT3_P_cyt => STAT3_cyt; STAT3_P_cyt, Rate Law: kdephosSTAT3*STAT3_P_cyt
ksyncJunmRNA = 0.0125; kAP1activity = 1.0Reaction: cFos_cJun => cJun_mRNA + cFos_cJun; cFos_cJun, Rate Law: ksyncJunmRNA*cFos_cJun*kAP1activity
kdephosSTAT3PTPRT = 8.0E-4Reaction: STAT3_P_cyt + PTPRT => STAT3_cyt + PTPRT; STAT3_P_cyt, PTPRT, Rate Law: kdephosSTAT3PTPRT*STAT3_P_cyt*PTPRT
kdegMMP13 = 1.0E-6Reaction: MMP13 => Sink; MMP13, Rate Law: kdegMMP13*MMP13
ksynPP4cJun = 2.0E-4Reaction: cJun_dimer => cJun_dimer + PP4; cJun_dimer, Rate Law: ksynPP4cJun*cJun_dimer
ksynMMP1mRNAcJun = 2.0E-4Reaction: cJun_dimer => cJun_dimer + MMP1_mRNA; cJun_dimer, Rate Law: ksynMMP1mRNAcJun*cJun_dimer
kinhibTRAF6 = 0.5Reaction: TRAF6 + PP4 => TRAF6_PP4; TRAF6, PP4, Rate Law: kinhibTRAF6*TRAF6*PP4
kinhibADAMTS4TIMP3 = 5.0E-4Reaction: TIMP3 + ADAMTS4 => ADAMTS4_TIMP3; TIMP3, ADAMTS4, Rate Law: kinhibADAMTS4TIMP3*TIMP3*ADAMTS4
kphosJNK = 1.0E-4Reaction: JNK + IRAK2_TRAF6 => JNK_P + IRAK2_TRAF6; JNK, IRAK2_TRAF6, Rate Law: kphosJNK*JNK*IRAK2_TRAF6
kdegcJunmRNA = 0.003Reaction: cJun_mRNA => Sink; cJun_mRNA, Rate Law: kdegcJunmRNA*cJun_mRNA
knuc2cytSTAT3 = 0.001Reaction: STAT3_nuc => STAT3_cyt; STAT3_nuc, Rate Law: knuc2cytSTAT3*STAT3_nuc
ksynMMP1mRNA = 0.005; kAP1activity = 1.0Reaction: cFos_cJun => cFos_cJun + MMP1_mRNA; cFos_cJun, Rate Law: ksynMMP1mRNA*cFos_cJun*kAP1activity
kdephosJAK1PTPRT = 0.004Reaction: JAK1_P + PTPRT => JAK1 + PTPRT; JAK1_P, PTPRT, Rate Law: kdephosJAK1PTPRT*JAK1_P*PTPRT
ksynbasalcJunmRNA = 0.015Reaction: Source => cJun_mRNA; Source, Rate Law: ksynbasalcJunmRNA*Source
kdephoscJun = 0.01Reaction: cJun_P => cJun; cJun_P, Rate Law: kdephoscJun*cJun_P
kdephosJAK1 = 4.0E-4Reaction: JAK1_P => JAK1; JAK1_P, Rate Law: kdephosJAK1*JAK1_P
kdegMMP1 = 1.0E-6Reaction: MMP1 => Sink; MMP1, Rate Law: kdegMMP1*MMP1
kdegCollagen2mmp1 = 5.0E-12Reaction: Collagen2 + MMP1 => MMP1 + ColFrag; Collagen2, MMP1, Rate Law: kdegCollagen2mmp1*Collagen2*MMP1
kbinSOCS3OSMR = 0.005Reaction: SOCS3 + OSMR => OSMR_SOCS3; SOCS3, OSMR, Rate Law: kbinSOCS3OSMR*SOCS3*OSMR
kdephosp38MKP1 = 1.0E-5Reaction: p38_P + MKP1 => p38 + MKP1; p38_P, MKP1, Rate Law: kdephosp38MKP1*p38_P*MKP1
ksynMMP13mRNA = 5.0E-4; kAP1activity = 1.0Reaction: cFos_cJun => cFos_cJun + MMP13_mRNA; cFos_cJun, Rate Law: ksynMMP13mRNA*cFos_cJun*kAP1activity
ksynTIMP1mRNA = 5.0E-7; kAP1activity = 1.0Reaction: cFos_cJun + TIMP1_DNA => cFos_cJun + TIMP1_mRNA + TIMP1_DNA; cFos_cJun, TIMP1_DNA, Rate Law: ksynTIMP1mRNA*cFos_cJun*TIMP1_DNA*kAP1activity
kdegPP4 = 1.0E-4Reaction: PP4 => Sink; PP4, Rate Law: kdegPP4*PP4
kdephosJNK = 0.001Reaction: JNK_P => JNK; JNK_P, Rate Law: kdephosJNK*JNK_P
kdegMMP1mRNA = 6.4E-6Reaction: MMP1_mRNA => Sink; MMP1_mRNA, Rate Law: kdegMMP1mRNA*MMP1_mRNA
ksynTIMP3mRNA = 5.0E-7; kAP1activity = 1.0Reaction: cFos_cJun => cFos_cJun + TIMP3_mRNA; cFos_cJun, Rate Law: ksynTIMP3mRNA*cFos_cJun*kAP1activity
kAP1activity = 1.0; ksyncFosmRNA = 5.0E-6Reaction: cFos_cJun => cFos_cJun + cFos_mRNA; cFos_cJun, Rate Law: ksyncFosmRNA*cFos_cJun*kAP1activity
krelADAMTS4TIMP3 = 0.001Reaction: ADAMTS4_TIMP3 => ADAMTS4 + TIMP3; ADAMTS4_TIMP3, Rate Law: krelADAMTS4TIMP3*ADAMTS4_TIMP3
kdegSOCS3 = 8.0E-4Reaction: SOCS3 => Sink; SOCS3, Rate Law: kdegSOCS3*SOCS3
ksynTIMP3 = 4.0E-4Reaction: TIMP3_mRNA => TIMP3_mRNA + TIMP3; TIMP3_mRNA, Rate Law: ksynTIMP3*TIMP3_mRNA

States:

NameDescription
Aggrecan Collagen2[Collagen alpha-1(II) chain; AggrecanAggrecan core protein]
cFos[Proto-oncogene c-Fos]
cJun[Transcription factor AP-1]
cJun mRNA[Transcription factor AP-1; JUN]
cFos mRNA[Proto-oncogene c-Fos; FOS]
p38 P[Mitogen-activated protein kinase 11; 3842]
TRAF6 PP4[Serine/threonine-protein phosphatase 4 catalytic subunit; TNF receptor-associated factor 6]
ColFrag[Collagen alpha-1(II) chain]
ADAMTS4[A disintegrin and metalloproteinase with thrombospondin motifs 4]
TIMP1 mRNA[TIMP1; Metalloproteinase inhibitor 1]
MKP1[Dual specificity protein phosphatase 1]
STAT3 cyt[cytoplasm; Signal transducer and activator of transcription 3]
JAK1[Tyrosine-protein kinase JAK1]
DUSP16[Dual specificity protein phosphatase 16]
MMP13 mRNA[MMP13; Collagenase 3]
STAT3 nuc[Signal transducer and activator of transcription 3; nucleus]
JNK P[Mitogen-activated protein kinase 8; 3842]
STAT3 P nuc[Signal transducer and activator of transcription 3; 3842; nucleus]
SOCS3[Suppressor of cytokine signaling 3]
ADAMTS4 TIMP1[Metalloproteinase inhibitor 1; A disintegrin and metalloproteinase with thrombospondin motifs 4]
TIMP3 mRNA[TIMP3; Metalloproteinase inhibitor 3]
SOCS3 mRNA[SOCS3; Suppressor of cytokine signaling 3]
STAT3 P cyt[cytoplasm; Signal transducer and activator of transcription 3; 3842]
p38[Mitogen-activated protein kinase 11]
IRAK2 TRAF6[TNF receptor-associated factor 6; Interleukin-1 receptor-associated kinase-like 2]
IRAK2 TRAF6 PP4[Serine/threonine-protein phosphatase 4 catalytic subunit; Interleukin-1 receptor-associated kinase-like 2; TNF receptor-associated factor 6]
ADAMTS4 TIMP3[Metalloproteinase inhibitor 1; A disintegrin and metalloproteinase with thrombospondin motifs 4]
proMMP1[Interstitial collagenase]
TIMP1 DNA[deoxyribonucleic acid; Metalloproteinase inhibitor 1]
JAK1 P[Tyrosine-protein kinase JAK1; 3842]
MMP1[Interstitial collagenase]
SinkSink
MMP1 mRNA[Interstitial collagenase; MMP1]
TRAF6[TNF receptor-associated factor 6]
PP4[Serine/threonine-protein phosphatase 4 catalytic subunit]

Proctor2013 - Effect of Aβ immunisation in Alzheimer's disease (deterministic version): BIOMD0000000488v0.0.1

Proctor2013 - Effect of Aβ immunisation in Alzheimer's disease (deterministic version)Extension of a previously publishe…

Details

Progress in the development of therapeutic interventions to treat or slow the progression of Alzheimer's disease has been hampered by lack of efficacy and unforeseen side effects in human clinical trials. This setback highlights the need for new approaches for pre-clinical testing of possible interventions. Systems modelling is becoming increasingly recognised as a valuable tool for investigating molecular and cellular mechanisms involved in ageing and age-related diseases. However, there is still a lack of awareness of modelling approaches in many areas of biomedical research. We previously developed a stochastic computer model to examine some of the key pathways involved in the aggregation of amyloid-beta (Aβ) and the micro-tubular binding protein tau. Here we show how we extended this model to include the main processes involved in passive and active immunisation against Aβ and then demonstrate the effects of this intervention on soluble Aβ, plaques, phosphorylated tau and tangles. The model predicts that immunisation leads to clearance of plaques but only results in small reductions in levels of soluble Aβ, phosphorylated tau and tangles. The behaviour of this model is supported by neuropathological observations in Alzheimer patients immunised against Aβ. Since, soluble Aβ, phosphorylated tau and tangles more closely correlate with cognitive decline than plaques, our model suggests that immunotherapy against Aβ may not be effective unless it is performed very early in the disease process or combined with other therapies. link: http://identifiers.org/pubmed/24098635

Parameters:

NameDescription
kdisaggAbeta2 = 1.0E-6Reaction: AbetaPlaque + antiAb => AbetaDimer + antiAb + disaggPlaque2; antiAb, AbetaPlaque, Rate Law: kdisaggAbeta2*antiAb*AbetaPlaque
kactDUBp53 = 1.0E-7Reaction: Mdm2_p53_Ub + p53DUB => Mdm2_p53 + p53DUB + Ub; Mdm2_p53_Ub, p53DUB, Rate Law: kactDUBp53*Mdm2_p53_Ub*p53DUB
kremROS = 7.0E-5Reaction: ROS => Sink; ROS, Rate Law: kremROS*ROS
kinactglia2 = 5.0E-6Reaction: GliaM2 => GliaM1; GliaM2, Rate Law: kinactglia2*GliaM2
kprodAbeta = 1.86E-5Reaction: Source => Abeta; Source, Rate Law: kprodAbeta*Source
krelMTTau = 1.0E-4Reaction: MT_Tau => Tau; MT_Tau, Rate Law: krelMTTau*MT_Tau
krepair = 2.0E-5Reaction: damDNA => Sink; damDNA, Rate Law: krepair*damDNA
kinhibprot = 1.0E-7Reaction: AbetaDimer + Proteasome => AggAbeta_Proteasome; AbetaDimer, Proteasome, Rate Law: kinhibprot*AbetaDimer*Proteasome
kbinAbantiAb = 1.0E-6Reaction: AbetaDimer + antiAb => AbetaDimer_antiAb; AbetaDimer, antiAb, Rate Law: kbinAbantiAb*AbetaDimer*antiAb
kactglia1 = 6.0E-7Reaction: GliaM1 + AbetaPlaque => GliaM2 + AbetaPlaque; GliaM1, AbetaPlaque, Rate Law: kactglia1*GliaM1*AbetaPlaque
kaggTauP1 = 1.0E-8Reaction: Tau_P1 => AggTau; Tau_P1, Rate Law: kaggTauP1*Tau_P1^2*0.5
kaggTauP2 = 1.0E-7Reaction: Tau_P2 => AggTau; Tau_P2, Rate Law: kaggTauP2*Tau_P2^2*0.5
kdephosMdm2 = 0.5Reaction: Mdm2_P => Mdm2; Mdm2_P, Rate Law: kdephosMdm2*Mdm2_P
kdephosp53 = 0.5Reaction: p53_P => p53; p53_P, Rate Law: kdephosp53*p53_P
kbinMdm2p53 = 0.001155Reaction: p53 + Mdm2 => Mdm2_p53; p53, Mdm2, Rate Law: kbinMdm2p53*p53*Mdm2
krelGSK3bp53 = 0.002Reaction: GSK3b_p53 => GSK3b + p53; GSK3b_p53, Rate Law: krelGSK3bp53*GSK3b_p53
kdegTau20SProt = 0.01Reaction: Proteasome_Tau => Proteasome; Proteasome_Tau, Rate Law: kdegTau20SProt*Proteasome_Tau
krelMdm2p53 = 1.155E-5Reaction: Mdm2_p53 => p53 + Mdm2; Mdm2_p53, Rate Law: krelMdm2p53*Mdm2_p53
kdisaggAbeta = 1.0E-6Reaction: AbetaDimer => Abeta; AbetaDimer, Rate Law: kdisaggAbeta*AbetaDimer
kaggTau = 1.0E-8Reaction: Tau => AggTau; Tau, Rate Law: kaggTau*Tau^2*0.5
kinactATM = 5.0E-4Reaction: ATMA => ATMI; ATMA, Rate Law: kinactATM*ATMA
kdisaggAbeta1 = 2.0E-4Reaction: AbetaPlaque => AbetaDimer + disaggPlaque1; AbetaPlaque, Rate Law: kdisaggAbeta1*AbetaPlaque
kdegAbetaGlia = 0.005Reaction: AbetaPlaque_GliaA => GliaA + degAbetaGlia; AbetaPlaque_GliaA, Rate Law: kdegAbetaGlia*AbetaPlaque_GliaA
kbinGSK3bp53 = 2.0E-6Reaction: GSK3b + p53 => GSK3b_p53; GSK3b, p53, Rate Law: kbinGSK3bp53*GSK3b*p53
kgenROSGlia = 1.0E-5Reaction: AbetaPlaque_GliaA => AbetaPlaque_GliaA + ROS; AbetaPlaque_GliaA, Rate Law: kgenROSGlia*AbetaPlaque_GliaA
kMdm2PolyUb = 0.00456Reaction: Mdm2_Ub2 + E2_Ub => Mdm2_Ub3 + E2; Mdm2_Ub2, E2_Ub, Rate Law: kMdm2PolyUb*Mdm2_Ub2*E2_Ub
ksynMdm2mRNAGSK3bp53 = 7.0E-4Reaction: GSK3b_p53 => GSK3b_p53 + Mdm2_mRNA; GSK3b_p53, Rate Law: ksynMdm2mRNAGSK3bp53*GSK3b_p53
kbinProt = 2.0E-6Reaction: Mdm2_Ub4 + Proteasome => Mdm2_Ub4_Proteasome; Mdm2_Ub4, Proteasome, Rate Law: kbinProt*Mdm2_Ub4*Proteasome
kphosMdm2GSK3b = 0.005Reaction: Mdm2_p53_Ub4 + GSK3b => Mdm2_P1_p53_Ub4 + GSK3b; Mdm2_p53_Ub4, GSK3b, Rate Law: kphosMdm2GSK3b*Mdm2_p53_Ub4*GSK3b
kbinE1Ub = 2.0E-4Reaction: E1 + Ub + ATP => E1_Ub + AMP; E1, Ub, ATP, Rate Law: kbinE1Ub*E1*Ub*ATP/(5000+ATP)
kpghalf = 10.0; kpg = 0.15Reaction: AbetaDimer + AbetaPlaque => AbetaPlaque; AbetaDimer, AbetaPlaque, Rate Law: kpg*AbetaDimer*AbetaPlaque^2/(kpghalf^2+AbetaPlaque^2)
kaggAbeta = 3.0E-6Reaction: Abeta => AbetaDimer; Abeta, Rate Law: kaggAbeta*Abeta^2*0.5
krelAbetaGlia = 5.0E-5Reaction: AbetaPlaque_GliaA => AbetaPlaque + GliaA; AbetaPlaque_GliaA, Rate Law: krelAbetaGlia*AbetaPlaque_GliaA
kp53Ub = 5.0E-5Reaction: E2_Ub + Mdm2_p53 => Mdm2_p53_Ub + E2; E2_Ub, Mdm2_p53, Rate Law: kp53Ub*E2_Ub*Mdm2_p53
kbinAbetaGlia = 1.0E-5Reaction: AbetaPlaque + GliaA => AbetaPlaque_GliaA; AbetaPlaque, GliaA, Rate Law: kbinAbetaGlia*AbetaPlaque*GliaA
kp53PolyUb = 0.01Reaction: Mdm2_p53_Ub2 + E2_Ub => Mdm2_p53_Ub3 + E2; Mdm2_p53_Ub2, E2_Ub, Rate Law: kp53PolyUb*Mdm2_p53_Ub2*E2_Ub
kdamROS = 1.0E-5Reaction: ROS => ROS + damDNA; ROS, Rate Law: kdamROS*ROS
kdam = 0.08Reaction: IR => IR + damDNA; IR, Rate Law: kdam*IR
ksynMdm2mRNA = 5.0E-4Reaction: p53_P => p53_P + Mdm2_mRNA; p53_P, Rate Law: ksynMdm2mRNA*p53_P
kgenROSPlaque = 1.0E-5Reaction: AbetaPlaque => AbetaPlaque + ROS; AbetaPlaque, Rate Law: kgenROSPlaque*AbetaPlaque
kactATM = 1.0E-4Reaction: damDNA + ATMI => damDNA + ATMA; damDNA, ATMI, Rate Law: kactATM*damDNA*ATMI
kphospTauGSK3b = 2.0E-4Reaction: GSK3b + Tau => GSK3b + Tau_P1; GSK3b, Tau, Rate Law: kphospTauGSK3b*GSK3b*Tau
kdegMdm2 = 0.01; kproteff = 1.0Reaction: Mdm2_Ub4_Proteasome => Proteasome + Ub; Mdm2_Ub4_Proteasome, Rate Law: kdegMdm2*Mdm2_Ub4_Proteasome*kproteff
kactDUBMdm2 = 1.0E-7Reaction: Mdm2_Ub + Mdm2DUB => Mdm2 + Mdm2DUB + Ub; Mdm2_Ub, Mdm2DUB, Rate Law: kactDUBMdm2*Mdm2_Ub*Mdm2DUB
kdegAbeta = 1.5E-5Reaction: Abeta_antiAb => antiAb; Abeta_antiAb, Rate Law: 10*kdegAbeta*Abeta_antiAb
kgenROSAbeta = 2.0E-5Reaction: AggAbeta_Proteasome => AggAbeta_Proteasome + ROS; AggAbeta_Proteasome, Rate Law: kgenROSAbeta*AggAbeta_Proteasome
kphosMdm2 = 2.0Reaction: Mdm2 + ATMA => Mdm2_P + ATMA; Mdm2, ATMA, Rate Law: kphosMdm2*Mdm2*ATMA
kactglia2 = 6.0E-7Reaction: GliaM2 + antiAb => GliaA + antiAb; GliaM2, antiAb, Rate Law: kactglia2*GliaM2*antiAb
kMdm2PUb = 6.84E-6Reaction: Mdm2_P + E2_Ub => Mdm2_P_Ub + E2; Mdm2_P, E2_Ub, Rate Law: kMdm2PUb*Mdm2_P*E2_Ub
kinactglia1 = 5.0E-6Reaction: GliaA => GliaM2; GliaA, Rate Law: kinactglia1*GliaA
kdegp53mRNA = 1.0E-4Reaction: p53_mRNA => Sink; p53_mRNA, Rate Law: kdegp53mRNA*p53_mRNA
ktangfor = 0.001Reaction: AggTau => NFT; AggTau, Rate Law: ktangfor*AggTau^2*0.5
ksynp53 = 0.007Reaction: p53_mRNA => p53 + p53_mRNA; p53_mRNA, Rate Law: ksynp53*p53_mRNA
kbinMTTau = 0.1Reaction: Tau => MT_Tau; Tau, Rate Law: kbinMTTau*Tau

States:

NameDescription
Mdm2 P[E3 ubiquitin-protein ligase Mdm2; phosphoprotein]
antiAb[Immunoglobulin]
Mdm2 p53 Ub2[Cellular tumor antigen p53; E3 ubiquitin-protein ligase Mdm2; Polyubiquitin-B]
AbetaPlaque[Amyloid beta A4 protein; urn:miriam:sbo:SBO%3A0000543]
MT Tau[IPR015562]
Ub[Polyubiquitin-B]
AMP[AMP]
p53[Cellular tumor antigen p53]
disaggPlaque2disaggPlaque2
Mdm2 Ub2[E3 ubiquitin-protein ligase Mdm2; Polyubiquitin-B]
SourceSource
GliaA[microglial cell]
p53 P[Cellular tumor antigen p53; phosphoprotein]
IRIR
Mdm2 P Ub[Polyubiquitin-B; E3 ubiquitin-protein ligase Mdm2; phosphoprotein]
Abeta[Amyloid beta A4 protein]
Mdm2[E3 ubiquitin-protein ligase Mdm2]
GliaM2[microglial cell]
ROS[reactive oxygen species]
Proteasome[proteasome complex]
GSK3b p53[Cellular tumor antigen p53; Glycogen synthase kinase-3 beta]
Mdm2 Ub3[Polyubiquitin-B; E3 ubiquitin-protein ligase Mdm2]
AbetaDimer[Amyloid beta A4 protein]
damDNA[deoxyribonucleic acid]
AbetaDimer antiAb[Amyloid beta A4 protein; Immunoglobulin]
Mdm2 P Ub2[Polyubiquitin-B; E3 ubiquitin-protein ligase Mdm2; phosphoprotein]
p53 mRNA[Cellular tumor antigen p53]
GliaM1[microglial cell]
AggTau[IPR002955; urn:miriam:sbo:SBO%3A0000543]
ATMA[Serine-protein kinase ATM; urn:miriam:pato:PATO%3A000234]
Mdm2 Ub4[Polyubiquitin-B; E3 ubiquitin-protein ligase Mdm2]
Mdm2 p53[E3 ubiquitin-protein ligase Mdm2; Cellular tumor antigen p53]
AbetaPlaque GliaA[Amyloid beta A4 protein; microglial cell; urn:miriam:sbo:SBO%3A0000543]
Abeta antiAb[Amyloid beta A4 protein; Immunoglobulin]
GSK3b[Glycogen synthase kinase-3 beta]
degAbetaGliadegAbetaGlia
SinkSink
Mdm2 P Ub4 Proteasome[Polyubiquitin-B; E3 ubiquitin-protein ligase Mdm2; proteasome complex; phosphoprotein]
Mdm2 Ub4 Proteasome[Polyubiquitin-B; E3 ubiquitin-protein ligase Mdm2; proteasome complex]
Mdm2 mRNA[E3 ubiquitin-protein ligase Mdm2]

Proctor2013 - Effect of Aβ immunisation in Alzheimer's disease (stochastic version): BIOMD0000000634v0.0.1

Proctor2013 - Effect of Aβ immunisation in Alzheimer's disease (stochastic version)Extension of a previously published s…

Details

Progress in the development of therapeutic interventions to treat or slow the progression of Alzheimer's disease has been hampered by lack of efficacy and unforeseen side effects in human clinical trials. This setback highlights the need for new approaches for pre-clinical testing of possible interventions. Systems modelling is becoming increasingly recognised as a valuable tool for investigating molecular and cellular mechanisms involved in ageing and age-related diseases. However, there is still a lack of awareness of modelling approaches in many areas of biomedical research. We previously developed a stochastic computer model to examine some of the key pathways involved in the aggregation of amyloid-beta (Aβ) and the micro-tubular binding protein tau. Here we show how we extended this model to include the main processes involved in passive and active immunisation against Aβ and then demonstrate the effects of this intervention on soluble Aβ, plaques, phosphorylated tau and tangles. The model predicts that immunisation leads to clearance of plaques but only results in small reductions in levels of soluble Aβ, phosphorylated tau and tangles. The behaviour of this model is supported by neuropathological observations in Alzheimer patients immunised against Aβ. Since, soluble Aβ, phosphorylated tau and tangles more closely correlate with cognitive decline than plaques, our model suggests that immunotherapy against Aβ may not be effective unless it is performed very early in the disease process or combined with other therapies. link: http://identifiers.org/pubmed/24098635

Parameters:

NameDescription
kdisaggAbeta2 = 1.0E-6Reaction: AbetaPlaque + antiAb => AbetaDimer + antiAb + disaggPlaque2; antiAb, AbetaPlaque, Rate Law: kdisaggAbeta2*antiAb*AbetaPlaque
kremROS = 7.0E-5Reaction: ROS => Sink; ROS, Rate Law: kremROS*ROS
kinactglia2 = 5.0E-6Reaction: GliaM1 => GliaI; GliaM1, Rate Law: kinactglia2*GliaM1
krelMTTau = 1.0E-4Reaction: MT_Tau => Tau; MT_Tau, Rate Law: krelMTTau*MT_Tau
krepair = 2.0E-5Reaction: damDNA => Sink; damDNA, Rate Law: krepair*damDNA
kinhibprot = 1.0E-7Reaction: AggTau + Proteasome => AggTau_Proteasome; AggTau, Proteasome, Rate Law: kinhibprot*AggTau*Proteasome
kbinAbantiAb = 1.0E-6Reaction: AbetaDimer + antiAb => AbetaDimer_antiAb; AbetaDimer, antiAb, Rate Law: kbinAbantiAb*AbetaDimer*antiAb
kactglia1 = 6.0E-7Reaction: GliaI + AbetaPlaque => GliaM1 + AbetaPlaque; GliaI, AbetaPlaque, Rate Law: kactglia1*GliaI*AbetaPlaque
kphosMdm2GSK3bp53 = 0.5Reaction: Mdm2_p53_Ub4 + GSK3b_p53_P => Mdm2_P1_p53_Ub4 + GSK3b_p53_P; Mdm2_p53_Ub4, GSK3b_p53_P, Rate Law: kphosMdm2GSK3bp53*Mdm2_p53_Ub4*GSK3b_p53_P
kaggTauP1 = 1.0E-8Reaction: Tau_P1 => AggTau; Tau_P1, Rate Law: kaggTauP1*Tau_P1*(Tau_P1-1)*0.5
kaggTauP2 = 1.0E-7Reaction: Tau_P2 => AggTau; Tau_P2, Rate Law: kaggTauP2*Tau_P2*(Tau_P2-1)*0.5
kdephosp53 = 0.5Reaction: p53_P => p53; p53_P, Rate Law: kdephosp53*p53_P
kbinMdm2p53 = 0.001155Reaction: p53 + Mdm2 => Mdm2_p53; p53, Mdm2, Rate Law: kbinMdm2p53*p53*Mdm2
kbinTauProt = 1.925E-7Reaction: Tau + Proteasome => Proteasome_Tau; Tau, Proteasome, Rate Law: kbinTauProt*Tau*Proteasome
krelGSK3bp53 = 0.002Reaction: GSK3b_p53_P => GSK3b + p53_P; GSK3b_p53_P, Rate Law: krelGSK3bp53*GSK3b_p53_P
kdegTau20SProt = 0.01Reaction: Proteasome_Tau => Proteasome; Proteasome_Tau, Rate Law: kdegTau20SProt*Proteasome_Tau
krelMdm2p53 = 1.155E-5Reaction: Mdm2_p53 => p53 + Mdm2; Mdm2_p53, Rate Law: krelMdm2p53*Mdm2_p53
kaggTau = 1.0E-8Reaction: Tau => AggTau; Tau, Rate Law: kaggTau*Tau*(Tau-1)*0.5
kdisaggAbeta = 1.0E-6Reaction: AbetaDimer => Abeta; AbetaDimer, Rate Law: kdisaggAbeta*AbetaDimer
kphosp53 = 2.0E-4Reaction: p53 + ATMA => p53_P + ATMA; p53, ATMA, Rate Law: kphosp53*p53*ATMA
kinactATM = 5.0E-4Reaction: ATMA => ATMI; ATMA, Rate Law: kinactATM*ATMA
kdisaggAbeta1 = 2.0E-4Reaction: AbetaPlaque => AbetaDimer + disaggPlaque1; AbetaPlaque, Rate Law: kdisaggAbeta1*AbetaPlaque
kdegAbetaGlia = 0.005Reaction: AbetaPlaque_GliaA => GliaA + degAbetaGlia; AbetaPlaque_GliaA, Rate Law: kdegAbetaGlia*AbetaPlaque_GliaA
kprodAbeta2 = 1.86E-5Reaction: GSK3b_p53_P => Abeta + GSK3b_p53_P; GSK3b_p53_P, Rate Law: kprodAbeta2*GSK3b_p53_P
kbinGSK3bp53 = 2.0E-6Reaction: GSK3b + p53_P => GSK3b_p53_P; GSK3b, p53_P, Rate Law: kbinGSK3bp53*GSK3b*p53_P
kgenROSGlia = 1.0E-5Reaction: AbetaPlaque_GliaA => AbetaPlaque_GliaA + ROS; AbetaPlaque_GliaA, Rate Law: kgenROSGlia*AbetaPlaque_GliaA
kMdm2PolyUb = 0.00456Reaction: Mdm2_Ub + E2_Ub => Mdm2_Ub2 + E2; Mdm2_Ub, E2_Ub, Rate Law: kMdm2PolyUb*Mdm2_Ub*E2_Ub
ksynMdm2mRNAGSK3bp53 = 7.0E-4Reaction: GSK3b_p53 => GSK3b_p53 + Mdm2_mRNA; GSK3b_p53, Rate Law: ksynMdm2mRNAGSK3bp53*GSK3b_p53
kbinProt = 2.0E-6Reaction: Mdm2_P1_p53_Ub4 + Proteasome => p53_Ub4_Proteasome + Mdm2; Mdm2_P1_p53_Ub4, Proteasome, Rate Law: kbinProt*Mdm2_P1_p53_Ub4*Proteasome
kbinE1Ub = 2.0E-4Reaction: E1 + Ub + ATP => E1_Ub + AMP; E1, Ub, ATP, Rate Law: kbinE1Ub*E1*Ub*ATP/(5000+ATP)
kpghalf = 10.0; kpg = 0.15Reaction: AbetaDimer + AbetaPlaque => AbetaPlaque; AbetaDimer, AbetaPlaque, Rate Law: kpg*AbetaDimer*AbetaPlaque^2/(kpghalf^2+AbetaPlaque^2)
ksynMdm2 = 4.95E-4Reaction: Mdm2_mRNA => Mdm2_mRNA + Mdm2; Mdm2_mRNA, Rate Law: ksynMdm2*Mdm2_mRNA
krelAbetaGlia = 5.0E-5Reaction: AbetaPlaque_GliaA => AbetaPlaque + GliaA; AbetaPlaque_GliaA, Rate Law: krelAbetaGlia*AbetaPlaque_GliaA
kaggAbeta = 3.0E-6Reaction: Abeta => AbetaDimer; Abeta, Rate Law: kaggAbeta*Abeta*(Abeta-1)*0.5
kbinAbetaGlia = 1.0E-5Reaction: AbetaPlaque + GliaA => AbetaPlaque_GliaA; AbetaPlaque, GliaA, Rate Law: kbinAbetaGlia*AbetaPlaque*GliaA
kphospTauGSK3bp53 = 0.1Reaction: GSK3b_p53 + Tau_P1 => GSK3b_p53 + Tau_P2; GSK3b_p53, Tau_P1, Rate Law: kphospTauGSK3bp53*GSK3b_p53*Tau_P1
kpf = 0.2Reaction: AbetaDimer => AbetaPlaque; AbetaDimer, Rate Law: kpf*AbetaDimer*(AbetaDimer-1)*0.5
kproteff = 1.0; kdegp53 = 0.005Reaction: p53_Ub4_Proteasome + ATP => Ub + Proteasome + ADP; p53_Ub4_Proteasome, ATP, Rate Law: kdegp53*kproteff*p53_Ub4_Proteasome*ATP/(5000+ATP)
kdephospTau = 0.01Reaction: Tau_P1 + PP1 => Tau + PP1; Tau_P1, PP1, Rate Law: kdephospTau*Tau_P1*PP1
ksynTau = 8.0E-5Reaction: Source => Tau; Source, Rate Law: ksynTau*Source
kdamROS = 1.0E-5Reaction: ROS => ROS + damDNA; ROS, Rate Law: kdamROS*ROS
kdam = 0.08Reaction: IR => IR + damDNA; IR, Rate Law: kdam*IR
ksynMdm2mRNA = 5.0E-4Reaction: p53_P => p53_P + Mdm2_mRNA; p53_P, Rate Law: ksynMdm2mRNA*p53_P
kgenROSPlaque = 1.0E-5Reaction: AbetaPlaque => AbetaPlaque + ROS; AbetaPlaque, Rate Law: kgenROSPlaque*AbetaPlaque
kMdm2Ub = 4.56E-6Reaction: Mdm2 + E2_Ub => Mdm2_Ub + E2; Mdm2, E2_Ub, Rate Law: kMdm2Ub*Mdm2*E2_Ub
kphospTauGSK3b = 2.0E-4Reaction: GSK3b + Tau_P1 => GSK3b + Tau_P2; GSK3b, Tau_P1, Rate Law: kphospTauGSK3b*GSK3b*Tau_P1
kdegAntiAb = 2.75E-6Reaction: antiAb => Sink; antiAb, Rate Law: kdegAntiAb*antiAb
kdegMdm2 = 0.01; kproteff = 1.0Reaction: Mdm2_Ub4_Proteasome => Proteasome + Ub; Mdm2_Ub4_Proteasome, Rate Law: kdegMdm2*Mdm2_Ub4_Proteasome*kproteff
kactATM = 1.0E-4Reaction: damDNA + ATMI => damDNA + ATMA; damDNA, ATMI, Rate Law: kactATM*damDNA*ATMI
kactDUBMdm2 = 1.0E-7Reaction: Mdm2_Ub + Mdm2DUB => Mdm2 + Mdm2DUB + Ub; Mdm2_Ub, Mdm2DUB, Rate Law: kactDUBMdm2*Mdm2_Ub*Mdm2DUB
kdegAbeta = 1.5E-5Reaction: AbetaDimer_antiAb => antiAb; AbetaDimer_antiAb, Rate Law: 10*kdegAbeta*AbetaDimer_antiAb
kgenROSAbeta = 2.0E-5Reaction: Abeta => Abeta + ROS; Abeta, Rate Law: kgenROSAbeta*Abeta
kphosMdm2 = 2.0Reaction: Mdm2 + ATMA => Mdm2_P + ATMA; Mdm2, ATMA, Rate Law: kphosMdm2*Mdm2*ATMA
ksynp53mRNA = 0.001Reaction: Source => p53_mRNA; Source, Rate Law: ksynp53mRNA*Source
kdegp53mRNA = 1.0E-4Reaction: p53_mRNA => Sink; p53_mRNA, Rate Law: kdegp53mRNA*p53_mRNA
ktangfor = 0.001Reaction: AggTau => NFT; AggTau, Rate Law: ktangfor*AggTau*(AggTau-1)*0.5
ksynp53 = 0.007Reaction: p53_mRNA => p53 + p53_mRNA; p53_mRNA, Rate Law: ksynp53*p53_mRNA
kbinMTTau = 0.1Reaction: Tau => MT_Tau; Tau, Rate Law: kbinMTTau*Tau
kdegMdm2mRNA = 5.0E-4Reaction: Mdm2_mRNA => Sink; Mdm2_mRNA, Rate Law: kdegMdm2mRNA*Mdm2_mRNA

States:

NameDescription
Mdm2 P[E3 ubiquitin-protein ligase Mdm2; phosphoprotein]
AggTau Proteasome[urn:miriam:sbo:SBO%3A0000543; IPR002955; proteasome complex]
MT Tau[IPR002955]
Proteasome Tau[IPR002955; proteasome complex]
AMP[AMP]
p53[Cellular tumor antigen p53]
Mdm2 Ub2[E3 ubiquitin-protein ligase Mdm2; Polyubiquitin-B]
SourceSource
p53 P[Cellular tumor antigen p53; phosphoprotein]
IRIR
E1[IPR000011]
GSK3b p53 P[Cellular tumor antigen p53; Glycogen synthase kinase-3 beta; phosphoprotein]
Abeta[Amyloid beta A4 protein]
Mdm2[E3 ubiquitin-protein ligase Mdm2]
ROS[reactive oxygen species]
Tau P1[IPR002955]
Proteasome[proteasome complex]
damDNA[deoxyribonucleic acid]
AggTau[urn:miriam:sbo:SBO%3A0000543; IPR002955]
AbetaDimer[Amyloid beta A4 protein]
AbetaDimer antiAb[Amyloid beta A4 protein; Immunoglobulin]
GliaM1[microglial cell]
p53 mRNA[Cellular tumor antigen p53]
PP1[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit]
ATMA[Serine-protein kinase ATM; active]
AbetaPlaque GliaA[Amyloid beta A4 protein; microglial cell; urn:miriam:sbo:SBO%3A0000543]
Mdm2 Ub[E3 ubiquitin-protein ligase Mdm2; Polyubiquitin-B]
ATMI[Serine-protein kinase ATM; inactive]
ADP[ADP]
Tau P2[IPR002955]
GliaI[microglial cell]
SinkSink
Mdm2 mRNA[E3 ubiquitin-protein ligase Mdm2]

Proctor2016 - Circadian rhythm of PTH and the dynamics of signaling molecules on bone remodeling: BIOMD0000000612v0.0.1

Proctor2016 - Circadian rhythm of PTH and the dynamics of signaling molecules on bone remodelingThis model is described…

Details

Bone remodeling is the continuous process of bone resorption by osteoclasts and bone formation by osteoblasts, in order to maintain homeostasis. The activity of osteoclasts and osteoblasts is regulated by a network of signaling pathways, including Wnt, parathyroid hormone (PTH), RANK ligand/osteoprotegrin, and TGF-β, in response to stimuli, such as mechanical loading. During aging there is a gradual loss of bone mass due to dysregulation of signaling pathways. This may be due to a decline in physical activity with age and/or changes in hormones and other signaling molecules. In particular, hormones, such as PTH, have a circadian rhythm, which may be disrupted in aging. Due to the complexity of the molecular and cellular networks involved in bone remodeling, several mathematical models have been proposed to aid understanding of the processes involved. However, to date, there are no models, which explicitly consider the effects of mechanical loading, the circadian rhythm of PTH, and the dynamics of signaling molecules on bone remodeling. Therefore, we have constructed a network model of the system using a modular approach, which will allow further modifications as required in future research. The model was used to simulate the effects of mechanical loading and also the effects of different interventions, such as continuous or intermittent administration of PTH. Our model predicts that the absence of regular mechanical loading and/or an impaired PTH circadian rhythm leads to a gradual decrease in bone mass over time, which can be restored by simulated interventions and that the effectiveness of some interventions may depend on their timing. link: http://identifiers.org/pubmed/27379013

Parameters:

NameDescription
kdegSost = 0.004Reaction: Sost => Sink; Sost, Rate Law: kdegSost*Sost
ksecRANKLbyOcyI = 1.0E-7Reaction: Ocy_I => Ocy_I + RANKL; Ocy_I, Rate Law: ksecRANKLbyOcyI*Ocy_I
kdiffHSC = 5.5E-5Reaction: HSC + MCSF => HSC + MCSF + Ocl_p; HSC, MCSF, Rate Law: kdiffHSC*HSC*MCSF^2/(50^2+MCSF^2)
kbinOclpRANKL = 0.001Reaction: RANKL + Ocl_p => Ocl_p_RANKL; Ocl_p, RANKL, Rate Law: kbinOclpRANKL*Ocl_p*RANKL
ksecMCSFbyObp = 1.0E-5Reaction: Ob_p => Ob_p + MCSF; Ob_p, Rate Law: ksecMCSFbyObp*Ob_p
ksecRANKLbyObpTgfb = 4.0E-6Reaction: Ob_p_Tgfb_A => Ob_p_Tgfb_A + RANKL; Ob_p_Tgfb_A, Rate Law: ksecRANKLbyObpTgfb*Ob_p_Tgfb_A
kdiffMSC = 6.5E-4Reaction: MSC + Wnt_A => MSC + Wnt_A + Ob_pro; MSC, Wnt_A, Rate Law: kdiffMSC*MSC*Wnt_A^2/(50^2+Wnt_A^2)
kbinBaxBcl2 = 0.01Reaction: Bax + Bcl2 => Bax_Bcl2; Bax, Bcl2, Rate Law: kbinBaxBcl2*Bax*Bcl2
krelCrebRunx2 = 0.01Reaction: CREB_Runx2 => CREB_P + Runx2; CREB_Runx2, Rate Law: krelCrebRunx2*CREB_Runx2
krelObmPTH = 0.005Reaction: Ob_m_PTH => Ob_m + PTH; Ob_m_PTH, Rate Law: krelObmPTH*Ob_m_PTH
ksecMCSFbyObpro = 1.0E-5Reaction: Ob_pro => Ob_pro + MCSF; Ob_pro, Rate Law: ksecMCSFbyObpro*Ob_pro
kactWnt = 0.03Reaction: Wnt_I => Wnt_A; Wnt_I, Rate Law: kactWnt*Wnt_I
ksecRANKLbyObp = 3.0E-6Reaction: Ob_p => Ob_p + RANKL; Ob_p, Rate Law: ksecRANKLbyObp*Ob_p
kdegBone = 6.5E-9Reaction: Ocl_m + Bone => Ocl_m; Ocl_m, Bone, Rate Law: kdegBone*Ocl_m*Bone
ksecRANKLbyObm = 1.0E-7Reaction: Ob_m => Ob_m + RANKL; Ob_m, Rate Law: ksecRANKLbyObm*Ob_m
kdiffObP = 1.0E-4Reaction: Ob_p => Ob_m; Ob_p, Rate Law: kdiffObP*Ob_p
kmatOb = 2.0E-9Reaction: Ob_m => Ocy_I; Ob_m, Rate Law: kmatOb*Ob_m
kdeathOb = 2.4E-4Reaction: Ob_m_PTH + Bax => Bax + PTH; Ob_m_PTH, Bax, Rate Law: kdeathOb*Ob_m_PTH*Bax^2/(50^2+Bax^2)
kformBone = 3.07E-6Reaction: Ob_m_PTH => Ob_m_PTH + Bone + newbone; Ob_m_PTH, Rate Law: kformBone*Ob_m_PTH
ksecRANKLbyObpro = 7.0E-6Reaction: Ob_pro => Ob_pro + RANKL; Ob_pro, Rate Law: ksecRANKLbyObpro*Ob_pro
kactCreb = 0.009Reaction: Ob_m_PTH + CREB => Ob_m_PTH + CREB_P; CREB, Ob_m_PTH, Rate Law: kactCreb*CREB*Ob_m_PTH^2/(100^2+Ob_m_PTH^2)
ksynPTH = 0.02Reaction: Source => PTH; Source, Rate Law: ksynPTH*Source
ksecRANKLbyOcy = 1.0E-6Reaction: Ocy_A => Ocy_A + RANKL; Ocy_A, Rate Law: ksecRANKLbyOcy*Ocy_A
kdeathOcy = 1.0E-8Reaction: Ocy_I => Sink; Ocy_I, Rate Law: kdeathOcy*Ocy_I
kdiffObproTgfb = 0.05Reaction: Ob_pro + Tgfb_A => Ob_p + Tgfb_A; Ob_pro, Tgfb_A, Rate Law: kdiffObproTgfb*Ob_pro*Tgfb_A^2/(50^2+Tgfb_A^2)
krelOclpRANKL = 0.001Reaction: Ocl_p_RANKL => Ocl_p + RANKL; Ocl_p_RANKL, Rate Law: krelOclpRANKL*Ocl_p_RANKL
kdeathOclp = 1.0E-5Reaction: Ocl_p => Sink; Ocl_p, Rate Law: kdeathOclp*Ocl_p
kdegRANKL = 3.0E-5Reaction: RANKL => Sink; RANKL, Rate Law: kdegRANKL*RANKL
kinactCreb = 1.0E-4Reaction: CREB_P => CREB; CREB_P, Rate Law: kinactCreb*CREB_P
krelOcyPTH = 0.005Reaction: Ocy_I_PTH => Ocy_I + PTH; Ocy_I_PTH, Rate Law: krelOcyPTH*Ocy_I_PTH
kdegRunx2PTH = 0.003Reaction: Ob_m_PTH + Runx2 => Ob_m_PTH; Runx2, Ob_m_PTH, Rate Law: kdegRunx2PTH*Runx2*Ob_m_PTH
ksecRANKLbyObmPTH = 1.0E-6Reaction: Ob_m_PTH => Ob_m_PTH + RANKL; Ob_m_PTH, Rate Law: ksecRANKLbyObmPTH*Ob_m_PTH
ksecTgfb = 5.0E-5Reaction: Ob_m => Ob_m + Tgfb_I; Ob_m, Rate Law: ksecTgfb*Ob_m
kinhibRANKL = 0.001Reaction: OPG + RANKL => OPG_RANKL; OPG, RANKL, Rate Law: kinhibRANKL*OPG*RANKL
ksynX = 0.01157Reaction: Source => X; Source, Rate Law: ksynX*Source
krelBaxBcl2 = 0.5Reaction: Bax_Bcl2 => Bax + Bcl2; Bax_Bcl2, Rate Law: krelBaxBcl2*Bax_Bcl2
kdegPTH = 0.002Reaction: PTH => Sink; PTH, Rate Law: kdegPTH*PTH
kdegMCSF = 1.0E-4Reaction: MCSF => Sink; MCSF, Rate Law: kdegMCSF*MCSF
kbinObpTgfb = 2.0E-4Reaction: Ob_p + Tgfb_A => Ob_p_Tgfb_A; Ob_p, Tgfb_A, Rate Law: kbinObpTgfb*Ob_p*Tgfb_A
ksynRunx2 = 0.005Reaction: Source => Runx2; Source, Rate Law: ksynRunx2*Source
kdeathOcl = 6.5E-5Reaction: Ocl_m => Sink; Ocl_m, Rate Law: kdeathOcl*Ocl_m
kdegTgfb = 5.0E-5Reaction: Tgfb_A => Sink; Tgfb_A, Rate Law: kdegTgfb*Tgfb_A
kactWntPth = 0.001Reaction: Wnt_I + Ob_m_PTH => Wnt_A + Ob_m_PTH; Wnt_I, Ob_m_PTH, Rate Law: kactWntPth*Wnt_I*Ob_m_PTH
ksecMCSFbyMSC = 1.0E-5Reaction: MSC => MSC + MCSF; MSC, Rate Law: ksecMCSFbyMSC*MSC
kdegOPG = 4.0E-6Reaction: OPG => Sink; OPG, Rate Law: kdegOPG*OPG
krelRANKL = 0.001Reaction: OPG_RANKL => OPG + RANKL; OPG_RANKL, Rate Law: krelRANKL*OPG_RANKL
kbinCrebRunx2 = 0.01Reaction: CREB_P + Runx2 => CREB_Runx2; CREB_P, Runx2, Rate Law: kbinCrebRunx2*CREB_P*Runx2
kdegOPGRANKL = 1.0E-5Reaction: OPG_RANKL => Sink; OPG_RANKL, Rate Law: kdegOPGRANKL*OPG_RANKL
kinactWnt = 0.8Reaction: Wnt_A + Sost => Wnt_I + Sost; Wnt_A, Sost, Rate Law: kinactWnt*Wnt_A*Sost^2/(50^2+Sost^2)
kactTgfb = 2.0E-7Reaction: Tgfb_I + Ocl_m => Tgfb_A + Ocl_m; Tgfb_I, Ocl_m, Rate Law: kactTgfb*Tgfb_I*Ocl_m
kdegRunx2 = 1.0E-4Reaction: Runx2 => Sink; Runx2, Rate Law: kdegRunx2*Runx2
ksecOPGbyObp = 2.0E-6Reaction: Ob_p => Ob_p + OPG; Ob_p, Rate Law: ksecOPGbyObp*Ob_p
kmatObTgfb = 1.0E-8Reaction: Ob_m + Tgfb_A => Ocy_I + Tgfb_A; Ob_m, Tgfb_A, Rate Law: kmatObTgfb*Ob_m*Tgfb_A^2/(50^2+Tgfb_A^2)
kdegTgfbPTH = 1.7E-5Reaction: Tgfb_A + Ob_m_PTH => Ob_m_PTH; Tgfb_A, Ob_m_PTH, Rate Law: kdegTgfbPTH*Tgfb_A*Ob_m_PTH
ksecMCSFbyObm = 1.0E-5Reaction: Ob_m_PTH => Ob_m_PTH + MCSF; Ob_m_PTH, Rate Law: ksecMCSFbyObm*Ob_m_PTH
kdegBcl2 = 0.0025Reaction: Bcl2 => Sink; Bcl2, Rate Law: kdegBcl2*Bcl2
kbinObpPTH = 3.0E-4Reaction: Ob_p + PTH => Ob_p_PTH; Ob_p, PTH, Rate Law: kbinObpPTH*Ob_p*PTH^2/(100^2+PTH^2)
kbinObmPTH = 0.02Reaction: Ob_m + PTH => Ob_m_PTH; Ob_m, PTH, Rate Law: kbinObmPTH*Ob_m*PTH^2/(100^2+PTH^2)
ksynBcl2 = 0.005Reaction: CREB_Runx2 => CREB_Runx2 + Bcl2; CREB_Runx2, Rate Law: ksynBcl2*CREB_Runx2
krelObpPTH = 0.005Reaction: Ob_p_PTH => Ob_p + PTH; Ob_p_PTH, Rate Law: krelObpPTH*Ob_p_PTH
ksecRANKLbyMSC = 1.0E-6Reaction: MSC => MSC + RANKL; MSC, Rate Law: ksecRANKLbyMSC*MSC
kunload = 3.5E-4Reaction: LOAD => Sink; LOAD, Rate Law: kunload*LOAD
ksecSost = 7.5E-4Reaction: Ocy_I => Ocy_I + Sost; Ocy_I, Rate Law: ksecSost*Ocy_I
ksecRANKLbyObpPTH = 2.0E-5Reaction: Ob_p_PTH => Ob_p_PTH + RANKL; Ob_p_PTH, Rate Law: ksecRANKLbyObpPTH*Ob_p_PTH

States:

NameDescription
Sost[Sclerostin]
CREB Runx2[Cyclic AMP-responsive element-binding protein 1; Runt-related transcription factor 2]
BoneBone
Ob p[preosteoblast]
Ob m[terminally differentiated osteoblast]
Wnt I[Proto-oncogene Wnt-1]
PTH[Parathyroid hormone]
MSC[mesenchymal stem cell]
HSC[bone marrow hematopoietic cell]
Bax[Apoptosis regulator BAX]
newbonenewbone
SourceSource
Tgfb I[Transforming growth factor beta-1]
CREB P[Cyclic AMP-responsive element-binding protein 1; phosphoprotein]
Bcl2[Apoptosis regulator Bcl-2]
CREB[Cyclic AMP-responsive element-binding protein 1]
XX
Ob p PTH[Parathyroid hormone; preosteoblast]
Ocy I PTH[Parathyroid hormone; osteocyte]
Runx2[Runt-related transcription factor 2]
Ob m PTH[Parathyroid hormone; terminally differentiated osteoblast]
Ob p Tgfb A[Transforming growth factor beta-1; preosteoblast]
Wnt A[Proto-oncogene Wnt-1; TGF-beta 1 isoform 1 cleaved 1]
LOADLOAD
Ob pro[non-terminally differentiated osteoblast]
RANKL[Tumor necrosis factor ligand superfamily member 11]
SinkSink
Bax Bcl2[Apoptosis regulator Bcl-2; Apoptosis regulator BAX]
MCSF[Macrophage colony-stimulating factor 1]

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mir181_in_muscle): MODEL1704110001v0.0.1

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mir181_in_muscle)This model is described in th…

Details

MicroRNAs (miRNAs) regulate gene expression through interactions with target sites within mRNAs, leading to enhanced degradation of the mRNA or inhibition of translation. Skeletal muscle expresses many different miRNAs with important roles in adulthood myogenesis (regeneration) and myofibre hypertrophy and atrophy, processes associated with muscle ageing. However, the large number of miRNAs and their targets mean that a complex network of pathways exists, making it difficult to predict the effect of selected miRNAs on age-related muscle wasting. Computational modelling has the potential to aid this process as it is possible to combine models of individual miRNA:target interactions to form an integrated network. As yet, no models of these interactions in muscle exist. We created the first model of miRNA:target interactions in myogenesis based on experimental evidence of individual miRNAs which were next validated and used to make testable predictions. Our model confirms that miRNAs regulate key interactions during myogenesis and can act by promoting the switch between quiescent/proliferating/differentiating myoblasts and by maintaining the differentiation process. We propose that a threshold level of miR-1 acts in the initial switch to differentiation, with miR-181 keeping the switch on and miR-378 maintaining the differentiation and miR-143 inhibiting myogenesis. link: http://identifiers.org/doi/10.1038/s41598-017-12538-6

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mirs_in_muscle): MODEL1704110004v0.0.1

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mirs_in_muscle)This model is described in the…

Details

MicroRNAs (miRNAs) regulate gene expression through interactions with target sites within mRNAs, leading to enhanced degradation of the mRNA or inhibition of translation. Skeletal muscle expresses many different miRNAs with important roles in adulthood myogenesis (regeneration) and myofibre hypertrophy and atrophy, processes associated with muscle ageing. However, the large number of miRNAs and their targets mean that a complex network of pathways exists, making it difficult to predict the effect of selected miRNAs on age-related muscle wasting. Computational modelling has the potential to aid this process as it is possible to combine models of individual miRNA:target interactions to form an integrated network. As yet, no models of these interactions in muscle exist. We created the first model of miRNA:target interactions in myogenesis based on experimental evidence of individual miRNAs which were next validated and used to make testable predictions. Our model confirms that miRNAs regulate key interactions during myogenesis and can act by promoting the switch between quiescent/proliferating/differentiating myoblasts and by maintaining the differentiation process. We propose that a threshold level of miR-1 acts in the initial switch to differentiation, with miR-181 keeping the switch on and miR-378 maintaining the differentiation and miR-143 inhibiting myogenesis. link: http://identifiers.org/doi/10.1038/s41598-017-12538-6

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mir1_in_muscle): MODEL1704110000v0.0.1

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mir1_in_muscle)This model is described in the…

Details

MicroRNAs (miRNAs) regulate gene expression through interactions with target sites within mRNAs, leading to enhanced degradation of the mRNA or inhibition of translation. Skeletal muscle expresses many different miRNAs with important roles in adulthood myogenesis (regeneration) and myofibre hypertrophy and atrophy, processes associated with muscle ageing. However, the large number of miRNAs and their targets mean that a complex network of pathways exists, making it difficult to predict the effect of selected miRNAs on age-related muscle wasting. Computational modelling has the potential to aid this process as it is possible to combine models of individual miRNA:target interactions to form an integrated network. As yet, no models of these interactions in muscle exist. We created the first model of miRNA:target interactions in myogenesis based on experimental evidence of individual miRNAs which were next validated and used to make testable predictions. Our model confirms that miRNAs regulate key interactions during myogenesis and can act by promoting the switch between quiescent/proliferating/differentiating myoblasts and by maintaining the differentiation process. We propose that a threshold level of miR-1 acts in the initial switch to differentiation, with miR-181 keeping the switch on and miR-378 maintaining the differentiation and miR-143 inhibiting myogenesis. link: http://identifiers.org/doi/10.1038/s41598-017-12538-6

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mir143_in_muscle): MODEL1704110003v0.0.1

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mir143_in_muscle)This model is described in th…

Details

MicroRNAs (miRNAs) regulate gene expression through interactions with target sites within mRNAs, leading to enhanced degradation of the mRNA or inhibition of translation. Skeletal muscle expresses many different miRNAs with important roles in adulthood myogenesis (regeneration) and myofibre hypertrophy and atrophy, processes associated with muscle ageing. However, the large number of miRNAs and their targets mean that a complex network of pathways exists, making it difficult to predict the effect of selected miRNAs on age-related muscle wasting. Computational modelling has the potential to aid this process as it is possible to combine models of individual miRNA:target interactions to form an integrated network. As yet, no models of these interactions in muscle exist. We created the first model of miRNA:target interactions in myogenesis based on experimental evidence of individual miRNAs which were next validated and used to make testable predictions. Our model confirms that miRNAs regulate key interactions during myogenesis and can act by promoting the switch between quiescent/proliferating/differentiating myoblasts and by maintaining the differentiation process. We propose that a threshold level of miR-1 acts in the initial switch to differentiation, with miR-181 keeping the switch on and miR-378 maintaining the differentiation and miR-143 inhibiting myogenesis. link: http://identifiers.org/doi/10.1038/s41598-017-12538-6

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mir378_in_muscle): MODEL1704110002v0.0.1

Proctor2017 - Identifying microRNA for muscle regeneration during ageing (Mir378_in_muscle)This model is described in th…

Details

MicroRNAs (miRNAs) regulate gene expression through interactions with target sites within mRNAs, leading to enhanced degradation of the mRNA or inhibition of translation. Skeletal muscle expresses many different miRNAs with important roles in adulthood myogenesis (regeneration) and myofibre hypertrophy and atrophy, processes associated with muscle ageing. However, the large number of miRNAs and their targets mean that a complex network of pathways exists, making it difficult to predict the effect of selected miRNAs on age-related muscle wasting. Computational modelling has the potential to aid this process as it is possible to combine models of individual miRNA:target interactions to form an integrated network. As yet, no models of these interactions in muscle exist. We created the first model of miRNA:target interactions in myogenesis based on experimental evidence of individual miRNAs which were next validated and used to make testable predictions. Our model confirms that miRNAs regulate key interactions during myogenesis and can act by promoting the switch between quiescent/proliferating/differentiating myoblasts and by maintaining the differentiation process. We propose that a threshold level of miR-1 acts in the initial switch to differentiation, with miR-181 keeping the switch on and miR-378 maintaining the differentiation and miR-143 inhibiting myogenesis. link: http://identifiers.org/doi/10.1038/s41598-017-12538-6

Proctor2017- Role of microRNAs in osteoarthritis (miR140 in osteoarthritis): MODEL1705170005v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (miR140 in osteoarthritis)This model is described in the article: [Com…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Proctor2017- Role of microRNAs in osteoarthritis (Mir140-IGFBP5 incoherent feed forward): MODEL1705170004v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (Mir140-IGFBP5 incoherent feed forward)This model is described in the a…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Proctor2017- Role of microRNAs in osteoarthritis (miR140-IL1 coherent feed forward): MODEL1705170001v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (miR140-IL1 coherent feed forward)This model is described in the articl…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Proctor2017- Role of microRNAs in osteoarthritis (miR140-IL1 incoherent feed forward): MODEL1705170002v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (miR140-IL1 incoherent feed forward)This model is described in the arti…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Proctor2017- Role of microRNAs in osteoarthritis (miR140-SMAD3 double negative feedback): MODEL1705170000v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (miR140-SMAD3 double negative feedback)This model is described in the a…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Proctor2017- Role of microRNAs in osteoarthritis (miR140-SOX9 incoherent feed forward): MODEL1705170003v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (miR140-SOX9 incoherent feed forward)This model is described in the art…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Proctor2017- Role of microRNAs in osteoarthritis (Negative Feedback By MicroRNA with Delay): MODEL1610100002v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (Negative Feedback By MicroRNA with Delay)This model is described in th…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Proctor2017- Role of microRNAs in osteoarthritis (Negative Feedback By MicroRNA): BIOMD0000000864v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (Negative Feedback By MicroRNA)This model is described in the article:…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Parameters:

NameDescription
kbinTF1miRgene = 0.005Reaction: miR_gene + TF1 => miR_gene_TF1, Rate Law: cell*kbinTF1miRgene*miR_gene*cell*TF1*cell/cell
krelTF1miRgene = 5.0Reaction: miR_gene_TF1 => miR_gene + TF1, Rate Law: cell*krelTF1miRgene*miR_gene_TF1*cell/cell
ksynMiR = 5.0Reaction: miR_gene_TF1 => miR_gene_TF1 + miR, Rate Law: cell*ksynMiR*miR_gene_TF1*cell/cell
ksynTF1mRNA = 10.0Reaction: Signal => Signal + TF1_mRNA, Rate Law: cell*ksynTF1mRNA*Signal*cell/cell
kdegMiR = 0.008Reaction: miR => Sink, Rate Law: cell*kdegMiR*miR*cell/cell
ksynTF1 = 0.05Reaction: TF1_mRNA => TF1_mRNA + TF1, Rate Law: cell*ksynTF1*TF1_mRNA*cell/cell
kdegTF1 = 0.005Reaction: TF1 => Sink, Rate Law: cell*kdegTF1*TF1*cell/cell
kdegTF1mRNA = 1.0E-4Reaction: TF1_mRNA => Sink, Rate Law: cell*kdegTF1mRNA*TF1_mRNA*cell/cell
kdegTF1mRNAbyMiR = 0.001Reaction: TF1_mRNA + miR => miR, Rate Law: cell*kdegTF1mRNAbyMiR*TF1_mRNA*cell*miR*cell/cell

States:

NameDescription
miR gene TF1miR_gene_TF1
TF1 mRNATF1_mRNA
miRmiR
SinkSink
miR genemiR_gene
TF1TF1
SignalSignal

Proctor2017- Role of microRNAs in osteoarthritis (Positive Feedback By Micro RNA): BIOMD0000000862v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (Positive Feedback By Micro RNA)This model is described in the article:…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Parameters:

NameDescription
krelTF2miRgene = 0.001Reaction: miR_gene_TF2 => miR_gene + TF2, Rate Law: cell*krelTF2miRgene*miR_gene_TF2*cell/cell
kdegTF1 = 1.0E-5Reaction: TF1 => Sink, Rate Law: cell*kdegTF1*TF1*cell/cell
kbinTF1miRgene = 0.002Reaction: miR_gene + TF1 => miR_gene_TF1, Rate Law: cell*kbinTF1miRgene*miR_gene*cell*TF1*cell/cell
kdegTF1mRNA = 1.0E-4Reaction: TF1_mRNA => Sink, Rate Law: cell*kdegTF1mRNA*TF1_mRNA*cell/cell
ksynMiR = 0.2Reaction: miR_gene_TF2 => miR_gene_TF2 + miR, Rate Law: cell*ksynMiR*miR_gene_TF2*cell/cell
krelTF1miRgene = 0.001Reaction: miR_gene_TF1 => miR_gene + TF1, Rate Law: cell*krelTF1miRgene*miR_gene_TF1*cell/cell
kbinTF2miRgene = 1.0E-4Reaction: miR_gene + TF2 => miR_gene_TF2, Rate Law: cell*kbinTF2miRgene*miR_gene*cell*TF2*cell/cell
kdegMiR = 4.0E-4Reaction: miR => Sink, Rate Law: cell*kdegMiR*miR*cell/cell
kdegTF1mRNAbyMiR = 1.0E-6Reaction: TF1_mRNA + miR => miR, Rate Law: cell*kdegTF1mRNAbyMiR*TF1_mRNA*cell*miR*cell/cell
ksynTF1mRNA = 0.01Reaction: Signal => Signal + TF1_mRNA, Rate Law: cell*ksynTF1mRNA*Signal*cell/cell
ksynTF1 = 3.0E-4Reaction: TF1_mRNA => TF1_mRNA + TF1, Rate Law: cell*ksynTF1*TF1_mRNA*cell/cell

States:

NameDescription
miR gene TF1miR_gene_TF1
miR gene TF2miR_gene_TF2
TF2TF2
TF1 mRNATF1_mRNA
miRmiR
miR genemiR_gene
SinkSink
SignalSignal
TF1TF1

Proctor2017- Role of microRNAs in osteoarthritis (Positive Feedforward Coherent By MicroRNA): MODEL1610100003v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (Positive Feedforward Coherent By MicroRNA)This model is described in t…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Proctor2017- Role of microRNAs in osteoarthritis (Positive Feedforward Incoherent By MicroRNA)_1: BIOMD0000000860v0.0.1

Proctor2017- Role of microRNAs in osteoarthritis (Positive Feedforward Incoherent By MicroRNA)This model is described in…

Details

The aim of this study was to show how computational models can be used to increase our understanding of the role of microRNAs in osteoarthritis (OA) using miR-140 as an example. Bioinformatics analysis and experimental results from the literature were used to create and calibrate models of gene regulatory networks in OA involving miR-140 along with key regulators such as NF-κB, SMAD3, and RUNX2. The individual models were created with the modelling standard, Systems Biology Markup Language, and integrated to examine the overall effect of miR-140 on cartilage homeostasis. Down-regulation of miR-140 may have either detrimental or protective effects for cartilage, indicating that the role of miR-140 is complex. Studies of individual networks in isolation may therefore lead to different conclusions. This indicated the need to combine the five chosen individual networks involving miR-140 into an integrated model. This model suggests that the overall effect of miR-140 is to change the response to an IL-1 stimulus from a prolonged increase in matrix degrading enzymes to a pulse-like response so that cartilage degradation is temporary. Our current model can easily be modified and extended as more experimental data become available about the role of miR-140 in OA. In addition, networks of other microRNAs that are important in OA could be incorporated. A fully integrated model could not only aid our understanding of the mechanisms of microRNAs in ageing cartilage but could also provide a useful tool to investigate the effect of potential interventions to prevent cartilage loss. link: http://identifiers.org/pubmed/29095952

Parameters:

NameDescription
kdegTF1targetmRNAbyMiR = 5.0E-5 1/ (mol *s)Reaction: TF1target_mRNA + miR => Sink + miR, Rate Law: cell*kdegTF1targetmRNAbyMiR*TF1target_mRNA*cell*miR*cell/cell
kdegMiR = 4.0E-4 1/sReaction: miR => Sink, Rate Law: cell*kdegMiR*miR*cell/cell
ksynTF1targetmRNA = 0.004 1/sReaction: TF1 => TF1 + TF1target_mRNA, Rate Law: cell*ksynTF1targetmRNA*TF1*cell/cell
ksynMiR = 2.0E-4 1/sReaction: TF1 => TF1 + miR, Rate Law: cell*ksynMiR*TF1*cell/cell
kdegTF1targetmRNA = 0.001 1/sReaction: TF1target_mRNA => Sink, Rate Law: cell*kdegTF1targetmRNA*TF1target_mRNA*cell/cell

States:

NameDescription
miR[C25966]
SinkSink
TF1[1,4-beta-D-Mannooligosaccharide]
TF1target mRNATF1target_mRNA

Puchalka2008 - Genome-scale metabolic network of Pseudomonas putida (iJP815): MODEL1507180044v0.0.1

Puchalka2008 - Genome-scale metabolic network of Pseudomonas putida (iJP815)This model is described in the article: [Ge…

Details

A cornerstone of biotechnology is the use of microorganisms for the efficient production of chemicals and the elimination of harmful waste. Pseudomonas putida is an archetype of such microbes due to its metabolic versatility, stress resistance, amenability to genetic modifications, and vast potential for environmental and industrial applications. To address both the elucidation of the metabolic wiring in P. putida and its uses in biocatalysis, in particular for the production of non-growth-related biochemicals, we developed and present here a genome-scale constraint-based model of the metabolism of P. putida KT2440. Network reconstruction and flux balance analysis (FBA) enabled definition of the structure of the metabolic network, identification of knowledge gaps, and pin-pointing of essential metabolic functions, facilitating thereby the refinement of gene annotations. FBA and flux variability analysis were used to analyze the properties, potential, and limits of the model. These analyses allowed identification, under various conditions, of key features of metabolism such as growth yield, resource distribution, network robustness, and gene essentiality. The model was validated with data from continuous cell cultures, high-throughput phenotyping data, (13)C-measurement of internal flux distributions, and specifically generated knock-out mutants. Auxotrophy was correctly predicted in 75% of the cases. These systematic analyses revealed that the metabolic network structure is the main factor determining the accuracy of predictions, whereas biomass composition has negligible influence. Finally, we drew on the model to devise metabolic engineering strategies to improve production of polyhydroxyalkanoates, a class of biotechnologically useful compounds whose synthesis is not coupled to cell survival. The solidly validated model yields valuable insights into genotype-phenotype relationships and provides a sound framework to explore this versatile bacterium and to capitalize on its vast biotechnological potential. link: http://identifiers.org/pubmed/18974823

Puri2010 - Mathematical Modeling for the Pathogenesis of Alzheimer's Disease: MODEL1409240001v0.0.1

Puri2010 - Mathematical Modeling for the Pathogenesis of Alzheimer's DiseasePuri2010 - Mathematical Modeling for the Pat…

Details

Despite extensive research, the pathogenesis of neurodegenerative Alzheimer's disease (AD) still eludes our comprehension. This is largely due to complex and dynamic cross-talks that occur among multiple cell types throughout the aging process. We present a mathematical model that helps define critical components of AD pathogenesis based on differential rate equations that represent the known cross-talks involving microglia, astroglia, neurons, and amyloid-β (Aβ). We demonstrate that the inflammatory activation of microglia serves as a key node for progressive neurodegeneration. Our analysis reveals that targeting microglia may hold potential promise in the prevention and treatment of AD. link: http://identifiers.org/pubmed/21179474

Purvis2005_IonicCurrentModel: MODEL7980735163v0.0.1

This a model from the article: Ionic current model of a hypoglossal motoneuron. Purvis LK, Butera RJ. J Neurophysiol…

Details

We have developed a single-compartment, electrophysiological, hypoglossal motoneuron (HM) model based primarily on experimental data from neonatal rat HMs. The model is able to reproduce the fine features of the HM action potential: the fast afterhyperpolarization, the afterdepolarization, and the medium-duration afterhyperpolarization (mAHP). The model also reproduces the repetitive firing properties seen in neonatal HMs and replicates the neuron's response to pharmacological experiments. The model was used to study the role of specific ionic currents in HM firing and how variations in the densities of these currents may account for age-dependent changes in excitability seen in HMs. By varying the density of a fast inactivating calcium current, the model alternates between accelerating and adapting firing patterns. Modeling the age-dependent increase in H current density accounts for the decrease in mAHP duration observed experimentally, but does not fully account for the decrease in input resistance. An increase in the density of the voltage-dependent potassium currents and the H current is required to account for the decrease in input resistance. These changes also account for the age-dependent decrease in action potential duration. link: http://identifiers.org/pubmed/15653786

Purvis2008 - A molecular signaling model of platelet phosphoinositide and calcium regulation during homeostasis and P2Y1 activation: MODEL1805150001v0.0.1

Mathematical model

Details

To quantify how various molecular mechanisms are integrated to maintain platelet homeostasis and allow responsiveness to adenosine diphosphate (ADP), we developed a computational model of the human platelet. Existing kinetic information for 77 reactions, 132 fixed kinetic rate constants, and 70 species was combined with electrochemical calculations, measurements of platelet ultrastructure, novel experimental results, and published single-cell data. The model accurately predicted: (1) steady-state resting concentrations for intracellular calcium, inositol 1,4,5-trisphosphate, diacylglycerol, phosphatidic acid, phosphatidylinositol, phosphatidylinositol phosphate, and phosphatidylinositol 4,5-bisphosphate; (2) transient increases in intracellular calcium, inositol 1,4,5-trisphosphate, and G(q)-GTP in response to ADP; and (3) the volume of the platelet dense tubular system. A more stringent test of the model involved stochastic simulation of individual platelets, which display an asynchronous calcium spiking behavior in response to ADP. Simulations accurately reproduced the broad frequency distribution of measured spiking events and demonstrated that asynchronous spiking was a consequence of stochastic fluctuations resulting from the small volume of the platelet. The model also provided insights into possible mechanisms of negative-feedback signaling, the relative potency of platelet agonists, and cell-to-cell variation across platelet populations. This integrative approach to platelet biology offers a novel and complementary strategy to traditional reductionist methods. link: http://identifiers.org/pubmed/18596227

Q


Qi2013 - IL-6 and IFN crosstalk model: BIOMD0000000544v0.0.1

Qi2013 - IL-6 and IFN crosstalk modelThis model [[BIOMD0000000544]](http://www.ebi.ac.uk/biomodels-main/BIOMD0000000544…

Details

BACKGROUND: Interferon-gamma (IFN-gamma) and interleukin-6 (IL-6) are multifunctional cytokines that regulate immune responses, cell proliferation, and tumour development and progression, which frequently have functionally opposing roles. The cellular responses to both cytokines are activated via the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. During the past 10 years, the crosstalk mechanism between the IFN-gamma and IL-6 pathways has been studied widely and several biological hypotheses have been proposed, but the kinetics and detailed crosstalk mechanism remain unclear. RESULTS: Using established mathematical models and new experimental observations of the crosstalk between the IFN-gamma and IL-6 pathways, we constructed a new crosstalk model that considers three possible crosstalk levels: (1) the competition between STAT1 and STAT3 for common receptor docking sites; (2) the mutual negative regulation between SOCS1 and SOCS3; and (3) the negative regulatory effects of the formation of STAT1/3 heterodimers. A number of simulations were tested to explore the consequences of cross-regulation between the two pathways. The simulation results agreed well with the experimental data, thereby demonstrating the effectiveness and correctness of the model. CONCLUSION: In this study, we developed a crosstalk model of the IFN-gamma and IL-6 pathways to theoretically investigate their cross-regulation mechanism. The simulation experiments showed the importance of the three crosstalk levels between the two pathways. In particular, the unbalanced competition between STAT1 and STAT3 for IFNR and gp130 led to preferential activation of IFN-gamma and IL-6, while at the same time the formation of STAT1/3 heterodimers enhanced preferential signal transduction by sequestering a fraction of the activated STATs. The model provided a good explanation of the experimental observations and provided insights that may inform further research to facilitate a better understanding of the cross-regulation mechanism between the two pathways. link: http://identifiers.org/pubmed/23384097

Parameters:

NameDescription
parameter_94 = 0.064; parameter_93 = 0.03Reaction: species_35 + species_36 => species_46; species_35, species_36, species_46, Rate Law: compartment_1*(parameter_93*species_35*species_36-parameter_94*species_46)
parameter_48 = 0.005Reaction: species_25 => species_24 + species_29; species_25, Rate Law: c3*parameter_48*species_25
parameter_153 = 0.2; parameter_152 = 0.001Reaction: species_95 + species_24 => species_94; species_24, species_94, species_95, Rate Law: c2*(parameter_152*species_24*species_95-parameter_153*species_94)
parameter_221 = 0.001; parameter_222 = 7.99942179Reaction: species_82 + species_11 => s118; species_82, species_11, s118, Rate Law: compartment_1*(parameter_221*species_82*species_11-parameter_222*s118)
parameter_123 = 0.3Reaction: species_66 => species_64 + species_59; species_66, Rate Law: compartment_1*parameter_123*species_66
parameter_145 = 0.003Reaction: species_88 => species_81 + species_108; species_88, Rate Law: compartment_1*parameter_145*species_88
parameter_120 = 0.27Reaction: species_65 => species_64 + species_61; species_65, Rate Law: compartment_1*parameter_120*species_65
parameter_109 = 2.5E-4; parameter_110 = 0.5Reaction: species_53 + species_57 => species_58; species_53, species_57, species_58, Rate Law: compartment_1*(parameter_109*species_53*species_57-parameter_110*species_58)
parameter_51 = 0.05Reaction: species_28 => species_11; species_28, Rate Law: parameter_51*species_28
parameter_166 = 0.003Reaction: species_101 => species_91 + species_20; species_101, Rate Law: compartment_1*parameter_166*species_101
parameter_155 = 0.005Reaction: species_94 => species_96 + species_24; species_94, Rate Law: c2*parameter_155*species_94
parameter_150 = 0.2; parameter_149 = 2.0E-7Reaction: species_84 + species_85 => species_91; species_84, species_85, species_91, Rate Law: compartment_1*(parameter_149*species_84*species_85-parameter_150*species_91)
parameter_161 = 0.1; parameter_160 = 0.02Reaction: species_99 + species_82 => species_100; species_99, species_82, species_100, Rate Law: compartment_1*(parameter_160*species_99*species_82-parameter_161*species_100)
parameter_129 = 0.1; parameter_130 = 0.05Reaction: species_5 + species_107 => species_78; species_5, species_107, species_78, Rate Law: compartment_1*(parameter_129*species_5*species_107-parameter_130*species_78)
parameter_241 = 0.2; parameter_240 = 0.001Reaction: s122 + species_24 => s126; s122, species_24, s126, Rate Law: parameter_240*s122*species_24-parameter_241*s126
parameter_238 = 0.001; parameter_239 = 0.2Reaction: species_20 + s120 => s135; species_20, s120, s135, Rate Law: compartment_1*(parameter_238*species_20*s120-parameter_239*s135)
parameter_61 = 6.0; parameter_62 = 0.06Reaction: species_16 => species_33; species_16, species_33, Rate Law: compartment_1*(parameter_61*species_16-parameter_62*species_33)
parameter_126 = 0.0388Reaction: species_75 => species_74; species_75, Rate Law: compartment_1*parameter_126*species_75
parameter_175 = 0.8; parameter_174 = 0.008Reaction: species_84 + species_100 => species_104; species_84, species_100, species_104, Rate Law: compartment_1*(parameter_174*species_84*species_100-parameter_175*species_104)
parameter_100 = 0.011; parameter_101 = 0.001833Reaction: species_44 + species_51 => species_52; species_44, species_51, species_52, Rate Law: compartment_1*(parameter_100*species_44*species_51-parameter_101*species_52)
parameter_177 = 0.2; parameter_176 = 0.001Reaction: species_108 + species_104 => species_105; species_108, species_104, species_105, Rate Law: compartment_1*(parameter_176*species_108*species_104-parameter_177*species_105)
parameter_88 = 9.0E-4; parameter_87 = 0.3Reaction: species_33 => species_9 + species_48; species_33, species_9, species_48, Rate Law: compartment_1*(parameter_87*species_33-parameter_88*species_9*species_48)
parameter_40 = 0.005Reaction: species_14 => species_23; species_14, Rate Law: parameter_40*species_14
parameter_131 = 0.02; parameter_132 = 0.02Reaction: species_79 + species_78 => species_80; species_79, species_78, species_80, Rate Law: parameter_131*species_79*species_78-parameter_132*species_80
parameter_99 = 1.0Reaction: species_50 => species_41 + species_49; species_50, Rate Law: compartment_1*parameter_99*species_50
parameter_243 = 0.0015Reaction: s135 => species_85 + species_11 + species_20; s135, Rate Law: compartment_1*parameter_243*s135
parameter_244 = 0.0025Reaction: s126 => species_26 + species_24 + species_96; s126, Rate Law: parameter_244*s126
parameter_96 = 0.0429; parameter_95 = 0.03Reaction: species_33 + species_47 => species_37; species_33, species_47, species_37, Rate Law: compartment_1*(parameter_95*species_33*species_47-parameter_96*species_37)
parameter_85 = 1.7; parameter_86 = 340.0Reaction: species_48 => species_108; species_48, Rate Law: compartment_1*parameter_85*species_48/(parameter_86+species_48)
parameter_53 = 400.0; parameter_52 = 0.01Reaction: => species_30; species_23, species_23, Rate Law: c3*parameter_52*species_23/(parameter_53+species_23)
parameter_224 = 5.09534E-4; parameter_225 = 4.982769238Reaction: species_12 + species_82 => s119; species_12, species_82, s119, Rate Law: compartment_1*(parameter_224*species_12*species_82-parameter_225*s119)
parameter_178 = 0.003Reaction: species_105 => species_99 + species_81 + species_84 + species_108; species_105, Rate Law: compartment_1*parameter_178*species_105
parameter_97 = 0.0717; parameter_98 = 0.2Reaction: species_49 + species_44 => species_50; species_49, species_44, species_50, Rate Law: compartment_1*(parameter_97*species_49*species_44-parameter_98*species_50)
parameter_63 = 0.01; parameter_64 = 0.55Reaction: species_33 + species_32 => species_34; species_33, species_32, species_34, Rate Law: compartment_1*(parameter_63*species_33*species_32-parameter_64*species_34)
parameter_159 = 0.01Reaction: => species_99; species_98, species_98, Rate Law: compartment_1*parameter_159*species_98
parameter_128 = 9.0E-4; parameter_127 = 0.9854Reaction: species_75 => species_76; species_75, species_76, Rate Law: compartment_1*(parameter_127*species_75^2-parameter_128*species_76)
parameter_83 = 0.0015; parameter_84 = 0.0045Reaction: species_47 => species_32 + species_35; species_47, species_32, species_35, Rate Law: compartment_1*(parameter_83*species_47-parameter_84*species_32*species_35)
parameter_236 = 0.1; parameter_235 = 0.02Reaction: species_26 + species_95 => s122; species_26, species_95, s122, Rate Law: parameter_235*species_26*species_95-parameter_236*s122
parameter_82 = 0.021; parameter_81 = 0.3Reaction: species_46 => species_47 + species_48; species_46, species_47, species_48, Rate Law: compartment_1*(parameter_81*species_46-parameter_82*species_47*species_48)
parameter_231 = 0.001; parameter_232 = 400.0Reaction: => species_30; species_92, species_92, Rate Law: c3*parameter_231*species_92/(parameter_232+species_92)
parameter_139 = 0.005; parameter_140 = 0.5Reaction: species_82 + species_85 => species_86; species_82, species_85, species_86, Rate Law: compartment_1*(parameter_139*species_82*species_85-parameter_140*species_86)
parameter_89 = 0.01; parameter_90 = 0.55Reaction: species_32 + species_48 => species_36; species_32, species_48, species_36, Rate Law: compartment_1*(parameter_89*species_32*species_48-parameter_90*species_36)
parameter_148 = 0.003Reaction: species_90 => species_84 + species_20; species_90, Rate Law: compartment_1*parameter_148*species_90
parameter_158 = 0.001Reaction: species_97 => species_98; species_97, Rate Law: parameter_158*species_97
parameter_49 = 0.2; parameter_50 = 2.0E-7Reaction: species_29 => species_26 + species_28; species_29, species_26, species_28, Rate Law: c3*(parameter_49*species_29-parameter_50*species_26*species_28)
parameter_122 = 0.5; parameter_121 = 0.005Reaction: species_61 + species_64 => species_66; species_61, species_64, species_66, Rate Law: compartment_1*(parameter_121*species_61*species_64-parameter_122*species_66)
parameter_138 = 0.4Reaction: species_83 => species_82 + species_85; species_83, Rate Law: compartment_1*parameter_138*species_83
parameter_44 = 0.2; parameter_43 = 0.001Reaction: species_24 + species_26 => species_27; species_24, species_26, species_27, Rate Law: c3*(parameter_43*species_24*species_26-parameter_44*species_27)
parameter_146 = 0.001; parameter_147 = 0.2Reaction: species_85 + species_20 => species_90; species_85, species_20, species_90, Rate Law: compartment_1*(parameter_146*species_85*species_20-parameter_147*species_90)
parameter_35 = 0.001; parameter_36 = 0.2Reaction: species_14 + species_20 => species_22; species_14, species_20, species_22, Rate Law: compartment_1*(parameter_35*species_14*species_20-parameter_36*species_22)
parameter_242 = 0.0015Reaction: s135 => species_20 + species_12 + species_84; s135, Rate Law: compartment_1*parameter_242*s135
parameter_133 = 0.04; parameter_134 = 0.2Reaction: species_80 => species_81; species_80, species_81, Rate Law: compartment_1*(parameter_133*species_80^2-parameter_134*species_81)
parameter_143 = 0.001; parameter_144 = 0.2Reaction: species_82 + species_108 => species_88; species_82, species_108, species_88, Rate Law: compartment_1*(parameter_143*species_82*species_108-parameter_144*species_88)
parameter_58 = 5.0E-4Reaction: species_31 => ; species_31, Rate Law: compartment_1*parameter_58*species_31
parameter_162 = 5.0E-4Reaction: species_98 => ; species_98, Rate Law: compartment_1*parameter_162*species_98
parameter_169 = 0.001; parameter_170 = 0.2Reaction: species_92 + species_24 => species_102; species_102, species_24, species_92, Rate Law: c2*(parameter_169*species_24*species_92-parameter_170*species_102)
parameter_245 = 0.0025Reaction: s126 => species_95 + species_28 + species_24; s126, Rate Law: parameter_245*s126
parameter_223 = 3.999994653Reaction: s118 => species_12 + species_82; s118, Rate Law: compartment_1*parameter_223*s118
parameter_135 = 0.005Reaction: species_81 => species_82; species_81, Rate Law: compartment_1*parameter_135*species_81
parameter_137 = 0.8; parameter_136 = 0.008Reaction: species_82 + species_84 => species_83; species_82, species_84, species_83, Rate Law: compartment_1*(parameter_136*species_82*species_84-parameter_137*species_83)
parameter_165 = 0.2; parameter_164 = 0.001Reaction: species_87 + species_20 => species_101; species_87, species_20, species_101, Rate Law: compartment_1*(parameter_164*species_87*species_20-parameter_165*species_101)
parameter_54 = 0.001Reaction: species_30 => species_31; species_30, Rate Law: parameter_54*species_30
parameter_56 = 5.0; parameter_57 = 0.1Reaction: species_9 + species_19 => species_15; species_9, species_19, species_15, Rate Law: compartment_1*(parameter_56*species_9*species_19-parameter_57*species_15)
parameter_163 = 5.0E-4Reaction: species_99 => ; species_99, Rate Law: compartment_1*parameter_163*species_99
parameter_32 = 0.001; parameter_33 = 0.2Reaction: species_12 + species_20 => species_21; species_12, species_20, species_21, Rate Law: compartment_1*(parameter_32*species_12*species_20-parameter_33*species_21)
parameter_125 = 20000.0; parameter_124 = 0.2335Reaction: species_74 => species_75; species_63, species_63, species_74, Rate Law: compartment_1*parameter_124*species_63*species_74/(species_74+parameter_125)
parameter_119 = 0.6; parameter_118 = 0.014Reaction: species_63 + species_64 => species_65; species_63, species_64, species_65, Rate Law: compartment_1*(parameter_118*species_63*species_64-parameter_119*species_65)
parameter_111 = 0.058Reaction: species_58 => species_57 + species_51; species_58, Rate Law: compartment_1*parameter_111*species_58
parameter_179 = 5.0E-4Reaction: species_105 => species_99 + species_106; species_105, Rate Law: compartment_1*parameter_179*species_105

States:

NameDescription
species 100[Tyrosine-protein kinase JAK2; Interferon gamma; Interferon gamma receptor 1; Suppressor of cytokine signaling 1; SBO:0000286; phosphorylated]
species 98[Suppressor of cytokine signaling 1; SBO:0000278]
species 20[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit]
species 91[Signal transducer and activator of transcription 1-alpha/beta; Signal transducer and activator of transcription 1-alpha/beta; phosphorylated]
species 47[Growth factor receptor-bound protein 2; Son of sevenless homolog 1]
species 66[Mitogen-activated protein kinase 1; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; phosphorylated]
species 21[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit; Signal transducer and activator of transcription 3; phosphorylated]
species 57[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
species 15[Interleukin-6 receptor subunit alpha; Interleukin-6; Tyrosine-protein kinase JAK1; Interleukin-6 receptor subunit beta; Suppressor of cytokine signaling 3; SBO:0000286]
species 83[Interferon gamma; Tyrosine-protein kinase JAK2; Interferon gamma receptor 1; Signal transducer and activator of transcription 1-alpha/beta; SBO:0000286; phosphorylated]
species 33[Interleukin-6; Interleukin-6 receptor subunit alpha; Interleukin-6 receptor subunit beta; Tyrosine-protein kinase JAK1; Tyrosine-protein phosphatase non-receptor type 11; SBO:0000286; phosphorylated]
species 64[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
species 24[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
species 78[Interferon gamma receptor 1; Tyrosine-protein kinase JAK2]
species 58[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; Dual specificity mitogen-activated protein kinase kinase 1; phosphorylated]
species 48[Tyrosine-protein phosphatase non-receptor type 11; phosphorylated]
species 76[CCAAT/enhancer-binding protein beta; active]
s126[Signal transducer and activator of transcription 1-alpha/beta; Signal transducer and activator of transcription 3; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; phosphorylated]
species 99[Suppressor of cytokine signaling 1]
species 101[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit; Signal transducer and activator of transcription 1-alpha/beta; SBO:0000286; phosphorylated]
species 65[Mitogen-activated protein kinase 1; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; phosphorylated]
species 50[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit; RAF proto-oncogene serine/threonine-protein kinase]
species 27[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; Signal transducer and activator of transcription 3; phosphorylated]
species 63[Mitogen-activated protein kinase 1]
s135[Signal transducer and activator of transcription 3; Signal transducer and activator of transcription 1-alpha/beta; Serine/threonine-protein phosphatase PP1-alpha catalytic subunit; phosphorylated]
species 31[Suppressor of cytokine signaling 3; SBO:0000278]
species 51[Dual specificity mitogen-activated protein kinase kinase 1]
species 104[Interferon gamma receptor 1; Interferon gamma; Tyrosine-protein kinase JAK2; Signal transducer and activator of transcription 1-alpha/beta; Suppressor of cytokine signaling 1; SBO:0000286; phosphorylated]
species 28[Signal transducer and activator of transcription 3]
species 75[CCAAT/enhancer-binding protein beta; active]
species 84[Signal transducer and activator of transcription 1-alpha/beta]
species 29[Signal transducer and activator of transcription 3; Signal transducer and activator of transcription 3; phosphorylated]
species 32[Growth factor receptor-bound protein 2]
species 30[Suppressor of cytokine signaling 3; SBO:0000278]
species 49[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit]
species 74[CCAAT/enhancer-binding protein beta]
species 81[Interferon gamma; Tyrosine-protein kinase JAK2; Interferon gamma receptor 1; SBO:0000286]
species 14[SBO:0000608; phosphorylated; Signal transducer and activator of transcription 3]
species 82[Interferon gamma; Tyrosine-protein kinase JAK2; Interferon gamma receptor 1; SBO:0000286; phosphorylated]
species 80[Tyrosine-protein kinase JAK2; Interferon gamma receptor 1; Interferon gamma]
species 46[Tyrosine-protein phosphatase non-receptor type 11; Son of sevenless homolog 1; Growth factor receptor-bound protein 2]
species 26[Signal transducer and activator of transcription 3; phosphorylated]
species 90[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit; Signal transducer and activator of transcription 1-alpha/beta; phosphorylated]

Qi2013 - IL-6 and IFN crosstalk model (non-competitive): BIOMD0000000543v0.0.1

Qi2013 - IL-6 and IFN crosstalk model (non-competitive)This model [[BIOMD0000000543]](http://www.ebi.ac.uk/biomodels-ma…

Details

BACKGROUND: Interferon-gamma (IFN-gamma) and interleukin-6 (IL-6) are multifunctional cytokines that regulate immune responses, cell proliferation, and tumour development and progression, which frequently have functionally opposing roles. The cellular responses to both cytokines are activated via the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. During the past 10 years, the crosstalk mechanism between the IFN-gamma and IL-6 pathways has been studied widely and several biological hypotheses have been proposed, but the kinetics and detailed crosstalk mechanism remain unclear. RESULTS: Using established mathematical models and new experimental observations of the crosstalk between the IFN-gamma and IL-6 pathways, we constructed a new crosstalk model that considers three possible crosstalk levels: (1) the competition between STAT1 and STAT3 for common receptor docking sites; (2) the mutual negative regulation between SOCS1 and SOCS3; and (3) the negative regulatory effects of the formation of STAT1/3 heterodimers. A number of simulations were tested to explore the consequences of cross-regulation between the two pathways. The simulation results agreed well with the experimental data, thereby demonstrating the effectiveness and correctness of the model. CONCLUSION: In this study, we developed a crosstalk model of the IFN-gamma and IL-6 pathways to theoretically investigate their cross-regulation mechanism. The simulation experiments showed the importance of the three crosstalk levels between the two pathways. In particular, the unbalanced competition between STAT1 and STAT3 for IFNR and gp130 led to preferential activation of IFN-gamma and IL-6, while at the same time the formation of STAT1/3 heterodimers enhanced preferential signal transduction by sequestering a fraction of the activated STATs. The model provided a good explanation of the experimental observations and provided insights that may inform further research to facilitate a better understanding of the cross-regulation mechanism between the two pathways. link: http://identifiers.org/pubmed/23384097

Parameters:

NameDescription
parameter_68 = 1.3; parameter_67 = 0.015Reaction: species_38 + species_37 => species_39; species_38, species_37, species_39, Rate Law: compartment_1*(parameter_67*species_38*species_37-parameter_68*species_39)
parameter_48 = 0.005Reaction: species_25 => species_24 + species_29; species_25, Rate Law: c3*parameter_48*species_25
parameter_153 = 0.2; parameter_152 = 0.001Reaction: species_95 + species_24 => species_94; species_24, species_94, species_95, Rate Law: c2*(parameter_152*species_24*species_95-parameter_153*species_94)
parameter_233 = 0.02; parameter_234 = 0.1Reaction: species_12 + species_85 => s120; species_12, species_85, s120, Rate Law: compartment_1*(parameter_233*species_12*species_85-parameter_234*s120)
parameter_123 = 0.3Reaction: species_66 => species_64 + species_59; species_66, Rate Law: compartment_1*parameter_123*species_66
parameter_120 = 0.27Reaction: species_65 => species_64 + species_61; species_65, Rate Law: compartment_1*parameter_120*species_65
parameter_166 = 0.003Reaction: species_101 => species_91 + species_20; species_101, Rate Law: compartment_1*parameter_166*species_101
parameter_69 = 0.5; parameter_70 = 1.0E-4Reaction: species_39 => species_40 + species_37; species_39, species_40, species_37, Rate Law: compartment_1*(parameter_69*species_39-parameter_70*species_40*species_37)
parameter_51 = 0.05Reaction: species_28 => species_11; species_28, Rate Law: parameter_51*species_28
parameter_155 = 0.005Reaction: species_94 => species_96 + species_24; species_94, Rate Law: c2*parameter_155*species_94
parameter_150 = 0.2; parameter_149 = 2.0E-7Reaction: species_84 + species_85 => species_91; species_84, species_85, species_91, Rate Law: compartment_1*(parameter_149*species_84*species_85-parameter_150*species_91)
parameter_65 = 0.01; parameter_66 = 0.0214Reaction: species_35 + species_34 => species_37; species_35, species_34, species_37, Rate Law: compartment_1*(parameter_65*species_35*species_34-parameter_66*species_37)
parameter_241 = 0.2; parameter_240 = 0.001Reaction: s122 + species_24 => s126; s122, species_24, s126, Rate Law: parameter_240*s122*species_24-parameter_241*s126
parameter_238 = 0.001; parameter_239 = 0.2Reaction: species_20 + s120 => s135; species_20, s120, s135, Rate Law: compartment_1*(parameter_238*species_20*s120-parameter_239*s135)
parameter_126 = 0.0388Reaction: species_75 => species_74; species_75, Rate Law: compartment_1*parameter_126*species_75
parameter_22 = 2.0; parameter_21 = 0.002Reaction: species_10 + species_84 => species_17; species_10, species_84, species_17, Rate Law: parameter_21*species_10*species_84-parameter_22*species_17
parameter_79 = 0.47; parameter_80 = 2.45E-4Reaction: species_37 => species_46 + species_9; species_37, species_46, species_9, Rate Law: compartment_1*(parameter_79*species_37-parameter_80*species_46*species_9)
parameter_1 = 0.1; parameter_2 = 0.05Reaction: species_2 + species_1 => species_3; species_2, species_1, species_3, Rate Law: parameter_1*species_2*species_1-parameter_2*species_3
parameter_23 = 0.008; parameter_24 = 0.8Reaction: species_67 + species_11 => species_17; species_67, species_11, species_17, Rate Law: parameter_23*species_67*species_11-parameter_24*species_17
parameter_131 = 0.02; parameter_132 = 0.02Reaction: species_79 + species_78 => species_80; species_79, species_78, species_80, Rate Law: parameter_131*species_79*species_78-parameter_132*species_80
parameter_99 = 1.0Reaction: species_50 => species_41 + species_49; species_50, Rate Law: compartment_1*parameter_99*species_50
parameter_243 = 0.0015Reaction: s135 => species_85 + species_11 + species_20; s135, Rate Law: compartment_1*parameter_243*s135
parameter_72 = 0.0053; parameter_71 = 0.001Reaction: species_40 + species_41 => species_42; species_40, species_41, species_42, Rate Law: compartment_1*(parameter_71*species_40*species_41-parameter_72*species_42)
parameter_8 = 0.8; parameter_7 = 0.008Reaction: s118 + species_84 => species_68; s118, species_84, species_68, Rate Law: compartment_1*(parameter_7*s118*species_84-parameter_8*species_68)
parameter_244 = 0.0025Reaction: s126 => species_26 + species_24 + species_96; s126, Rate Law: parameter_244*s126
parameter_14 = 0.008; parameter_15 = 0.8Reaction: species_9 + species_11 => species_10; species_9, species_11, species_10, Rate Law: compartment_1*(parameter_14*species_9*species_11-parameter_15*species_10)
parameter_96 = 0.0429; parameter_95 = 0.03Reaction: species_33 + species_47 => species_37; species_33, species_47, species_37, Rate Law: compartment_1*(parameter_95*species_33*species_47-parameter_96*species_37)
parameter_74 = 7.0E-4; parameter_73 = 1.0Reaction: species_42 => species_43 + species_44; species_42, species_43, species_44, Rate Law: compartment_1*(parameter_73*species_42-parameter_74*species_43*species_44)
parameter_229 = 0.005; parameter_230 = 0.5Reaction: species_85 + species_9 => s139; species_85, species_9, s139, Rate Law: compartment_1*(parameter_229*species_85*species_9-parameter_230*s139)
parameter_53 = 400.0; parameter_52 = 0.01Reaction: => species_30; species_23, species_23, Rate Law: c3*parameter_52*species_23/(parameter_53+species_23)
parameter_168 = 0.5; parameter_167 = 0.005Reaction: species_95 => species_92; species_95, species_92, Rate Law: c2*(parameter_167*species_95^2-parameter_168*species_92)
parameter_221 = 0.002; parameter_222 = 2.0Reaction: species_82 + species_11 => s118; species_82, species_11, s118, Rate Law: compartment_1*(parameter_221*species_82*species_11-parameter_222*s118)
parameter_159 = 0.01Reaction: => species_99; species_98, species_98, Rate Law: compartment_1*parameter_159*species_98
parameter_37 = 0.003Reaction: species_22 => species_18 + species_20; species_22, Rate Law: compartment_1*parameter_37*species_22
parameter_25 = 0.2Reaction: species_17 => species_10 + species_85; species_17, Rate Law: parameter_25*species_17
parameter_223 = 0.2Reaction: s118 => species_12 + species_82; s118, Rate Law: compartment_1*parameter_223*s118
parameter_83 = 0.0015; parameter_84 = 0.0045Reaction: species_47 => species_32 + species_35; species_47, species_32, species_35, Rate Law: compartment_1*(parameter_83*species_47-parameter_84*species_32*species_35)
parameter_236 = 0.1; parameter_235 = 0.02Reaction: species_26 + species_95 => s122; species_26, species_95, s122, Rate Law: parameter_235*species_26*species_95-parameter_236*s122
parameter_10 = 2.0; parameter_9 = 0.002Reaction: species_83 + species_11 => species_68; species_83, species_11, species_68, Rate Law: compartment_1*(parameter_9*species_83*species_11-parameter_10*species_68)
parameter_38 = 2.0E-7; parameter_39 = 0.2Reaction: species_11 + species_12 => species_18; species_11, species_12, species_18, Rate Law: compartment_1*(parameter_38*species_11*species_12-parameter_39*species_18)
parameter_237 = 0.005Reaction: s120 => s122; s120, Rate Law: compartment_1*parameter_237*s120
parameter_139 = 0.005; parameter_140 = 0.5Reaction: species_82 + species_85 => species_86; species_82, species_85, species_86, Rate Law: compartment_1*(parameter_139*species_82*species_85-parameter_140*species_86)
parameter_89 = 0.01; parameter_90 = 0.55Reaction: species_32 + species_48 => species_36; species_32, species_48, species_36, Rate Law: compartment_1*(parameter_89*species_32*species_48-parameter_90*species_36)
parameter_26 = 0.4Reaction: species_17 => species_67 + species_12; species_17, Rate Law: parameter_26*species_17
parameter_158 = 0.001Reaction: species_97 => species_98; species_97, Rate Law: parameter_158*species_97
parameter_49 = 0.2; parameter_50 = 2.0E-7Reaction: species_29 => species_26 + species_28; species_29, species_26, species_28, Rate Law: c3*(parameter_49*species_29-parameter_50*species_26*species_28)
parameter_19 = 0.4Reaction: species_68 => s118 + species_85; species_68, Rate Law: compartment_1*parameter_19*species_68
parameter_122 = 0.5; parameter_121 = 0.005Reaction: species_61 + species_64 => species_66; species_61, species_64, species_66, Rate Law: compartment_1*(parameter_121*species_61*species_64-parameter_122*species_66)
parameter_138 = 0.4Reaction: species_83 => species_82 + species_85; species_83, Rate Law: compartment_1*parameter_138*species_83
parameter_44 = 0.2; parameter_43 = 0.001Reaction: species_24 + species_26 => species_27; species_24, species_26, species_27, Rate Law: c3*(parameter_43*species_24*species_26-parameter_44*species_27)
parameter_151 = 0.005Reaction: species_87 => species_92; species_87, Rate Law: parameter_151*species_87
parameter_35 = 0.001; parameter_36 = 0.2Reaction: species_14 + species_20 => species_22; species_14, species_20, species_22, Rate Law: compartment_1*(parameter_35*species_14*species_20-parameter_36*species_22)
parameter_20 = 0.2Reaction: species_68 => species_83 + species_12; species_68, Rate Law: compartment_1*parameter_20*species_68
parameter_34 = 0.003Reaction: species_21 => species_11 + species_20; species_21, Rate Law: compartment_1*parameter_34*species_21
parameter_78 = 2.2E-4; parameter_77 = 0.023Reaction: species_45 => species_37 + species_38; species_45, species_37, species_38, Rate Law: compartment_1*(parameter_77*species_45-parameter_78*species_37*species_38)
parameter_242 = 0.0015Reaction: s135 => species_20 + species_12 + species_84; s135, Rate Law: compartment_1*parameter_242*s135
parameter_224 = 0.005; parameter_225 = 0.5Reaction: species_12 + species_82 => s119; species_12, species_82, s119, Rate Law: compartment_1*(parameter_224*species_12*species_82-parameter_225*s119)
parameter_162 = 5.0E-4Reaction: species_98 => ; species_98, Rate Law: compartment_1*parameter_162*species_98
parameter_245 = 0.0025Reaction: s126 => species_95 + species_28 + species_24; s126, Rate Law: parameter_245*s126
parameter_169 = 0.001; parameter_170 = 0.2Reaction: species_92 + species_24 => species_102; species_102, species_24, species_92, Rate Law: c2*(parameter_169*species_24*species_92-parameter_170*species_102)
parameter_76 = 0.4; parameter_75 = 0.0079Reaction: species_37 + species_43 => species_45; species_37, species_43, species_45, Rate Law: compartment_1*(parameter_75*species_37*species_43-parameter_76*species_45)
parameter_165 = 0.2; parameter_164 = 0.001Reaction: species_87 + species_20 => species_101; species_87, species_20, species_101, Rate Law: compartment_1*(parameter_164*species_87*species_20-parameter_165*species_101)
parameter_142 = 0.1; parameter_141 = 0.02Reaction: species_85 => species_87; species_85, species_87, Rate Law: compartment_1*(parameter_141*species_85^2-parameter_142*species_87)
parameter_56 = 5.0; parameter_57 = 0.1Reaction: species_9 + species_19 => species_15; species_9, species_19, species_15, Rate Law: compartment_1*(parameter_56*species_9*species_19-parameter_57*species_15)
parameter_45 = 0.005Reaction: species_27 => species_28 + species_24; species_27, Rate Law: c3*parameter_45*species_27
parameter_228 = 0.2Reaction: s138 => species_9 + species_85; s138, Rate Law: compartment_1*parameter_228*s138
parameter_125 = 20000.0; parameter_124 = 0.2335Reaction: species_74 => species_75; species_63, species_63, species_74, Rate Law: compartment_1*parameter_124*species_63*species_74/(species_74+parameter_125)
parameter_94 = 0.064; parameter_93 = 0.03Reaction: species_35 + species_36 => species_46; species_35, species_36, species_46, Rate Law: compartment_1*(parameter_93*species_35*species_36-parameter_94*species_46)

States:

NameDescription
species 67[Interleukin-6; Interleukin-6 receptor subunit alpha; Tyrosine-protein kinase JAK1; Interleukin-6 receptor subunit beta; SBO:0000286; Signal transducer and activator of transcription 1-alpha/beta]
species 27[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; Signal transducer and activator of transcription 3; phosphorylated]
species 36[Growth factor receptor-bound protein 2; Tyrosine-protein phosphatase non-receptor type 11]
species 98[Suppressor of cytokine signaling 1; SBO:0000278]
species 1[Interleukin-6]
species 20[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit]
species 28[Signal transducer and activator of transcription 3]
s120[Signal transducer and activator of transcription 3; Signal transducer and activator of transcription 1-alpha/beta; SBO:0000607; phosphorylated]
s122[Signal transducer and activator of transcription 3; Signal transducer and activator of transcription 1-alpha/beta; SBO:0000607; phosphorylated]
species 75[CCAAT/enhancer-binding protein beta; active]
species 91[Signal transducer and activator of transcription 1-alpha/beta; Signal transducer and activator of transcription 1-alpha/beta; phosphorylated]
species 79[Interferon gamma]
species 92[SBO:0000608; phosphorylated; Signal transducer and activator of transcription 1-alpha/beta]
species 39[Interleukin-6 receptor subunit alpha; Interleukin-6; Tyrosine-protein kinase JAK1; Interleukin-6 receptor subunit beta; SBO:0000286; Son of sevenless homolog 1; Tyrosine-protein phosphatase non-receptor type 11; Growth factor receptor-bound protein 2; GTPase HRas]
species 68[Interferon gamma receptor 1; Interferon gamma; Tyrosine-protein kinase JAK2; SBO:0000286; phosphorylated; Signal transducer and activator of transcription 1-alpha/beta; Signal transducer and activator of transcription 3]
species 66[Mitogen-activated protein kinase 1; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; phosphorylated]
species 21[Serine/threonine-protein phosphatase PP1-alpha catalytic subunit; Signal transducer and activator of transcription 3; phosphorylated]
species 32[Growth factor receptor-bound protein 2]
species 29[Signal transducer and activator of transcription 3; Signal transducer and activator of transcription 3; phosphorylated]
species 30[Suppressor of cytokine signaling 3; SBO:0000278]
species 17[Interleukin-6; Interleukin-6 receptor subunit beta; Interleukin-6 receptor subunit alpha; Tyrosine-protein kinase JAK1; SBO:0000286; Signal transducer and activator of transcription 1-alpha/beta; Signal transducer and activator of transcription 3]
species 12[Signal transducer and activator of transcription 3; phosphorylated]
species 15[Interleukin-6 receptor subunit alpha; Interleukin-6; Tyrosine-protein kinase JAK1; Interleukin-6 receptor subunit beta; SBO:0000286; Suppressor of cytokine signaling 3]
species 94[Signal transducer and activator of transcription 1-alpha/beta; phosphorylated; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
s118[Interferon gamma; Interferon gamma receptor 1; Tyrosine-protein kinase JAK2; SBO:0000286; Signal transducer and activator of transcription 3]
s119[Interferon gamma receptor 1; Interferon gamma; Tyrosine-protein kinase JAK2; SBO:0000286; phosphorylated; Signal transducer and activator of transcription 3]
species 37[Interleukin-6 receptor subunit alpha; Interleukin-6; Interleukin-6 receptor subunit beta; Tyrosine-protein kinase JAK1; SBO:0000286; Son of sevenless homolog 1; Tyrosine-protein phosphatase non-receptor type 11; Growth factor receptor-bound protein 2]
species 38[GTPase HRas; inactive]
species 42[RAF proto-oncogene serine/threonine-protein kinase; GTPase HRas]
species 74[CCAAT/enhancer-binding protein beta]
species 64[Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
species 11[Signal transducer and activator of transcription 3]
species 85[Signal transducer and activator of transcription 1-alpha/beta; phosphorylated]
species 95[Signal transducer and activator of transcription 1-alpha/beta; phosphorylated]
species 43[GTPase HRas; phosphorylated; active]
species 22[Signal transducer and activator of transcription 3; Serine/threonine-protein phosphatase PP1-alpha catalytic subunit; SBO:0000608; phosphorylated]
species 82[Interferon gamma; Tyrosine-protein kinase JAK2; Interferon gamma receptor 1; SBO:0000286; phosphorylated]
s126[Signal transducer and activator of transcription 1-alpha/beta; Signal transducer and activator of transcription 3; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; phosphorylated]
species 41[RAF proto-oncogene serine/threonine-protein kinase]
species 99[Suppressor of cytokine signaling 1]
species 26[Signal transducer and activator of transcription 3; phosphorylated]
species 40[GTPase HRas; active]

Qiao2004_ThrombinGeneration: MODEL1108260015v0.0.1

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

Details

The paper described a limited part of the coagulation pathway, and in particular the inhibitory effects of activated protein C in the context of thrombin production. This is a computational modeling study with various assumption made of kinetic rates laws and their summation. The level of complexity and assumed parameters makes conclusions uncertain. However, an interesting outcome is that kinetic reaction rates may show oscillation behavior under particular, high levels of protein C feedback inhibition. The model would defy quantitative practical use, but could have predictive value as a qualitative descriptor of coagulation. link: http://identifiers.org/pubmed/15121060

Qiao2007_MAPK_Signaling_Bistable: MODEL6185511733v0.0.1

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

Details

Physicochemical models of signaling pathways are characterized by high levels of structural and parametric uncertainty, reflecting both incomplete knowledge about signal transduction and the intrinsic variability of cellular processes. As a result, these models try to predict the dynamics of systems with tens or even hundreds of free parameters. At this level of uncertainty, model analysis should emphasize statistics of systems-level properties, rather than the detailed structure of solutions or boundaries separating different dynamic regimes. Based on the combination of random parameter search and continuation algorithms, we developed a methodology for the statistical analysis of mechanistic signaling models. In applying it to the well-studied MAPK cascade model, we discovered a large region of oscillations and explained their emergence from single-stage bistability. The surprising abundance of strongly nonlinear (oscillatory and bistable) input/output maps revealed by our analysis may be one of the reasons why the MAPK cascade in vivo is embedded in more complex regulatory structures. We argue that this type of analysis should accompany nonlinear multiparameter studies of stationary as well as transient features in network dynamics. link: http://identifiers.org/pubmed/17907797

Qiao2007_MAPK_Signaling_Oscillatory: MODEL6185746832v0.0.1

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

Details

Physicochemical models of signaling pathways are characterized by high levels of structural and parametric uncertainty, reflecting both incomplete knowledge about signal transduction and the intrinsic variability of cellular processes. As a result, these models try to predict the dynamics of systems with tens or even hundreds of free parameters. At this level of uncertainty, model analysis should emphasize statistics of systems-level properties, rather than the detailed structure of solutions or boundaries separating different dynamic regimes. Based on the combination of random parameter search and continuation algorithms, we developed a methodology for the statistical analysis of mechanistic signaling models. In applying it to the well-studied MAPK cascade model, we discovered a large region of oscillations and explained their emergence from single-stage bistability. The surprising abundance of strongly nonlinear (oscillatory and bistable) input/output maps revealed by our analysis may be one of the reasons why the MAPK cascade in vivo is embedded in more complex regulatory structures. We argue that this type of analysis should accompany nonlinear multiparameter studies of stationary as well as transient features in network dynamics. link: http://identifiers.org/pubmed/17907797