1
|
Xu J, Smith L. Curating models from BioModels: Developing a workflow for creating OMEX files. PLoS One 2024; 19:e0314875. [PMID: 39636894 PMCID: PMC11620473 DOI: 10.1371/journal.pone.0314875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
The reproducibility of computational biology models can be greatly facilitated by widely adopted standards and public repositories. We examined 50 models from the BioModels Database and attempted to validate the original curation and correct some of them if necessary. For each model, we reproduced these published results using Tellurium. Once reproduced we manually created a new set of files, with the model information stored by the Systems Biology Markup Language (SBML), and simulation instructions stored by the Simulation Experiment Description Markup Language (SED-ML), and everything included in an Open Modeling EXchange (OMEX) file, which could be used with a variety of simulators to reproduce the same results. On the one hand, the reproducibility procedure of 50 models developed a manual workflow that we would use to build an automatic platform to help users more easily curate and verify models in the future. On the other hand, these exercises allowed us to find the limitations and possible enhancement of the current curation and tooling to verify and curate models.
Collapse
Affiliation(s)
- Jin Xu
- Department of Bioengineering, University of Washington, Seattle, WA, United States of America
| | - Lucian Smith
- Department of Bioengineering, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
2
|
Nadal A, Cardesa A, Agaimy A, Almangush A, Franchi A, Hellquist H, Leivo I, Zidar N, Ferlito A. Massive parallel sequencing of head and neck conventional squamous cell carcinomas: A comprehensive review. Virchows Arch 2024; 485:965-976. [PMID: 39613893 DOI: 10.1007/s00428-024-03987-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and is a cause of significant mortality and morbidity. The epidemiology of this cancer varies worldwide due to either genetic differences in populations or differences in carcinogen exposure. The application of massive parallel sequencing-based techniques in HNSCC should provide a helpful understanding of the genetic alterations that eventually lead to HNSCC development and progression, and ideally, could be used for personalized therapy. In this review, the reader will find an overview of the mutational profile of conventional HNSCC according to published results on massive parallel sequencing data that confirm the pivotal role of TP53 and the frequent involvement of CDKN2A and PIK3CA. The reader will also find a more detailed description of the genes, such as NOTCH1 and FBXW7, that were not identified in HNSCCs before the development of these techniques, the differences that can be site-specific, such as the different mutational signatures that indicate specific carcinogens for various subsites of the head and neck, and finally, the actionability of these findings that should allow more personalized therapy for patients.
Collapse
Affiliation(s)
- Alfons Nadal
- Pathology Department, Department of Clinical Fundamentals, Universitat de Barcelona, IDIBAPS, Clínic Barcelona, Barcelona, Spain.
| | | | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany
| | - Alhadi Almangush
- Department of Pathology, University of Helsinki, Helsinki, Finland
- Institute of Biomedicine, Pathology, University of Turku, Turku, Finland
| | - Alessandro Franchi
- Section of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Henrik Hellquist
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Campus de Gambelas, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal
| | - Ilmo Leivo
- Institute of Biomedicine, Pathology, University of Turku, Turku University Central Hospital, 20521, Turku, Finland
| | - Nina Zidar
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Alfio Ferlito
- International Head and Neck Scientific Group, Padua, Italy
| |
Collapse
|
3
|
Xu J, Smith L. Curating models from BioModels: Developing a workflow for creating OMEX files. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585236. [PMID: 38559029 PMCID: PMC10979985 DOI: 10.1101/2024.03.15.585236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The reproducibility of computational biology models can be greatly facilitated by widely adopted standards and public repositories. We examined 50 models from the BioModels Database and attempted to validate the original curation and correct some of them if necessary. For each model, we reproduced these published results using Tellurium. Once reproduced we manually created a new set of files, with the model information stored by the Systems Biology Markup Language (SBML), and simulation instructions stored by the Simulation Experiment Description Markup Language (SED-ML), and everything included in an Open Modeling EXchange (OMEX) file, which could be used with a variety of simulators to reproduce the same results. On the one hand, the reproducibility procedure of 50 models developed a manual workflow that we would use to build an automatic platform to help users more easily curate and verify models in the future. On the other hand, these exercises allowed us to find the limitations and possible enhancement of the current curation and tooling to verify and curate models.
Collapse
Affiliation(s)
- Jin Xu
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Lucian Smith
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
4
|
Vieira-Lara MA, Bakker BM. The paradox of fatty-acid β-oxidation in muscle insulin resistance: Metabolic control and muscle heterogeneity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167172. [PMID: 38631409 DOI: 10.1016/j.bbadis.2024.167172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
The skeletal muscle is a metabolically heterogeneous tissue that plays a key role in maintaining whole-body glucose homeostasis. It is well known that muscle insulin resistance (IR) precedes the development of type 2 diabetes. There is a consensus that the accumulation of specific lipid species in the tissue can drive IR. However, the role of the mitochondrial fatty-acid β-oxidation in IR and, consequently, in the control of glucose uptake remains paradoxical: interventions that either inhibit or activate fatty-acid β-oxidation have been shown to prevent IR. We here discuss the current theories and evidence for the interplay between β-oxidation and glucose uptake in IR. To address the underlying intricacies, we (1) dive into the control of glucose uptake fluxes into muscle tissues using the framework of Metabolic Control Analysis, and (2) disentangle concepts of flux and catalytic capacities taking into account skeletal muscle heterogeneity. Finally, we speculate about hitherto unexplored mechanisms that could bring contrasting evidence together. Elucidating how β-oxidation is connected to muscle IR and the underlying role of muscle heterogeneity enhances disease understanding and paves the way for new treatments for type 2 diabetes.
Collapse
Affiliation(s)
- Marcel A Vieira-Lara
- Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Barbara M Bakker
- Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
5
|
Giudice G, Chen H, Koutsandreas T, Petsalaki E. phuEGO: A Network-Based Method to Reconstruct Active Signaling Pathways From Phosphoproteomics Datasets. Mol Cell Proteomics 2024; 23:100771. [PMID: 38642805 PMCID: PMC11134849 DOI: 10.1016/j.mcpro.2024.100771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024] Open
Abstract
Signaling networks are critical for virtually all cell functions. Our current knowledge of cell signaling has been summarized in signaling pathway databases, which, while useful, are highly biased toward well-studied processes, and do not capture context specific network wiring or pathway cross-talk. Mass spectrometry-based phosphoproteomics data can provide a more unbiased view of active cell signaling processes in a given context, however, it suffers from low signal-to-noise ratio and poor reproducibility across experiments. While progress in methods to extract active signaling signatures from such data has been made, there are still limitations with respect to balancing bias and interpretability. Here we present phuEGO, which combines up-to-three-layer network propagation with ego network decomposition to provide small networks comprising active functional signaling modules. PhuEGO boosts the signal-to-noise ratio from global phosphoproteomics datasets, enriches the resulting networks for functional phosphosites and allows the improved comparison and integration across datasets. We applied phuEGO to five phosphoproteomics data sets from cell lines collected upon infection with SARS CoV2. PhuEGO was better able to identify common active functions across datasets and to point to a subnetwork enriched for known COVID-19 targets. Overall, phuEGO provides a flexible tool to the community for the improved functional interpretation of global phosphoproteomics datasets.
Collapse
Affiliation(s)
- Girolamo Giudice
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridgeshire, United Kingdom
| | - Haoqi Chen
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridgeshire, United Kingdom
| | - Thodoris Koutsandreas
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridgeshire, United Kingdom
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridgeshire, United Kingdom.
| |
Collapse
|
6
|
Knöchel J, Kloft C, Huisinga W. Index analysis: An approach to understand signal transduction with application to the EGFR signalling pathway. PLoS Comput Biol 2024; 20:e1011777. [PMID: 38315738 PMCID: PMC10868873 DOI: 10.1371/journal.pcbi.1011777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/15/2024] [Accepted: 12/21/2023] [Indexed: 02/07/2024] Open
Abstract
In systems biology and pharmacology, large-scale kinetic models are used to study the dynamic response of a system to a specific input or stimulus. While in many applications, a deeper understanding of the input-response behaviour is highly desirable, it is often hindered by the large number of molecular species and the complexity of the interactions. An approach that identifies key molecular species for a given input-response relationship and characterises dynamic properties of states is therefore highly desirable. We introduce the concept of index analysis; it is based on different time- and state-dependent quantities (indices) to identify important dynamic characteristics of molecular species. All indices are defined for a specific pair of input and response variables as well as for a specific magnitude of the input. In application to a large-scale kinetic model of the EGFR signalling cascade, we identified different phases of signal transduction, the peculiar role of Phosphatase3 during signal activation and Ras recycling during signal onset. In addition, we discuss the challenges and pitfalls of interpreting the relevance of molecular species based on knock-out simulation studies, and provide an alternative view on conflicting results on the importance of parallel EGFR downstream pathways. Beyond the applications in model interpretation, index analysis is envisioned to be a valuable tool in model reduction.
Collapse
Affiliation(s)
- Jane Knöchel
- Institute of Mathematics, Universität Potsdam, Potsdam, Germany
- Graduate Research Training Program PharMetrX: Pharmacometrics & Computational Disease Modeling, Freie Universität Berlin and Universität Potsdam, Berlin/Potsdam, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | | |
Collapse
|
7
|
Westerhoff HV. On paradoxes between optimal growth, metabolic control analysis, and flux balance analysis. Biosystems 2023; 233:104998. [PMID: 37591451 DOI: 10.1016/j.biosystems.2023.104998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023]
Abstract
In Microbiology it is often assumed that growth rate is maximal. This may be taken to suggest that the dependence of the growth rate on every enzyme activity is at the top of an inverse-parabolic function, i.e. that all flux control coefficients should equal zero. This might seem to imply that the sum of these flux control coefficients equals zero. According to the summation law of Metabolic Control Analysis (MCA) the sum of flux control coefficients should equal 1 however. And in Flux Balance Analysis (FBA) catabolism is often limited by a hard bound, causing catabolism to fully control the fluxes, again in apparent contrast with a flux control coefficient of zero. Here we resolve these paradoxes (apparent contradictions) in an analysis that uses the 'Edinburgh pathway', the 'Amsterdam pathway', as well as a generic metabolic network providing the building blocks or Gibbs energy for microbial growth. We review and show that (i) optimization depends on so-called enzyme control coefficients rather than the 'catalytic control coefficients' of MCA's summation law, (ii) when optimization occurs at fixed total protein, the former differ from the latter to the extent that they may all become equal to zero in the optimum state, (iii) in more realistic scenarios of optimization where catalytically inert biomass is compensating or maintenance metabolism is taken into consideration, the optimum enzyme concentrations should not be expected to equal those that maximize the specific growth rate, (iv) optimization may be in terms of yield rather than specific growth rate, which resolves the paradox because the sum of catalytic control coefficients on yield equals 0, (v) FBA effectively maximizes growth yield, and for yield the summation law states 0 rather than 1, thereby removing the paradox, (vi) furthermore, FBA then comes more often to a 'hard optimum' defined by a maximum catabolic flux and a catabolic-enzyme control coefficient of 1. The trade-off between maintenance metabolism and growth is highlighted as worthy of further analysis.
Collapse
Affiliation(s)
- Hans V Westerhoff
- Department of Molecular Cell Biology, Vrije Universiteit Amsterdam, A-Life, De Boelelaan 1085, 1081 HV, Amsterdam, the Netherlands; Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands; School of Biological Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom; Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, Stellenbosch, 7600, South Africa.
| |
Collapse
|
8
|
Bruggeman FJ, Remeijer M, Droste M, Salinas L, Wortel M, Planqué R, Sauro HM, Teusink B, Westerhoff HV. Whole-cell metabolic control analysis. Biosystems 2023; 234:105067. [PMID: 39492480 DOI: 10.1016/j.biosystems.2023.105067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/05/2024]
Abstract
Since its conception some fifty years ago, metabolic control analysis (MCA) aims to understand how cells control their metabolism by adjusting the activity of their enzymes. Here we extend its scope to a whole-cell context. We consider metabolism in the evolutionary context of growth-rate maximisation by optimisation of protein concentrations. This framework allows for the prediction of flux control coefficients from proteomics data or stoichiometric modelling. Since genes compete for finite biosynthetic resources, we treat all protein concentrations as interdependent. We show that elementary flux modes (EFMs) emerge naturally as the optimal metabolic networks in the whole-cell context and we derive their control properties. In the evolutionary optimum, the number of expressed EFMs is determined by the number of protein-concentration constraints that limit growth rate. We use published glucose-limited chemostat data of S. cerevisiae to illustrate that it uses only two EFMs prior to the onset of fermentation and that it uses four EFMs during fermentation. We discuss published enzyme-titration data to show that S. cerevisiae and E. coli indeed can express proteins at growth-rate maximising concentrations. Accordingly, we extend MCA to elementary flux modes operating at an optimal state. We find that the expression of growth-unassociated proteins changes results from classical metabolic control analysis. Finally, we show how flux control coefficients can be estimated from proteomics and ribosome-profiling data. We analyse published proteomics data of E. coli to provide a whole-cell perspective of the control of metabolic enzymes on growth rate. We hope that this paper stimulates a renewed interest in metabolic control analysis, so that it can serve again the purpose it once had: to identify general principles that emerge from the biochemistry of the cell and are conserved across biological species.
Collapse
Affiliation(s)
- Frank J Bruggeman
- Systems Biology Lab, A-LIFE, AIMMS, VU University, Amsterdam, Netherlands.
| | - Maaike Remeijer
- Systems Biology Lab, A-LIFE, AIMMS, VU University, Amsterdam, Netherlands
| | - Maarten Droste
- Systems Biology Lab, A-LIFE, AIMMS, VU University, Amsterdam, Netherlands; Department of Mathematics, VU University, Amsterdam, Netherlands
| | - Luis Salinas
- Systems Biology Lab, A-LIFE, AIMMS, VU University, Amsterdam, Netherlands
| | - Meike Wortel
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Robert Planqué
- Department of Mathematics, VU University, Amsterdam, Netherlands
| | - Herbert M Sauro
- Department of Bioengineering, University of Washington, Seattle, WA, 98195-5061, USA
| | - Bas Teusink
- Systems Biology Lab, A-LIFE, AIMMS, VU University, Amsterdam, Netherlands
| | - Hans V Westerhoff
- Systems Biology Lab, A-LIFE, AIMMS, VU University, Amsterdam, Netherlands
| |
Collapse
|
9
|
Sarmiento-Ortega VE, Moroni-González D, Diaz A, Brambila E, Treviño S. ROS and ERK Pathway Mechanistic Approach on Hepatic Insulin Resistance After Chronic Oral Exposure to Cadmium NOAEL Dose. Biol Trace Elem Res 2023; 201:3903-3918. [PMID: 36348173 DOI: 10.1007/s12011-022-03471-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
Cadmium is a critical toxic agent in occupational and non-occupational settings and acute and chronic environmental exposure situations that have recently been associated with metabolic disease development. Until now, the no observed adverse effect level (NOAEL) of cadmium has not been studied regarding insulin resistance development. Therefore, we aimed to monitor whether chronic oral exposure to cadmium NOAEL dose induces insulin resistance in Wistar rats and investigate if oxidative stress and/or inflammation are related. Male Wistar rats were separated into control (standard normocalorie diet + water free of cadmium) and cadmium groups (standard normocalorie diet + drinking water with 15 ppm CdCl2). At 15, 30, and 60 days, oral glucose tolerance, insulin response, and insulin resistance were analyzed using mathematical models. In the liver glycogen, triglyceride, pro- and anti-inflammatory cytokines, cadmium, zinc, metallothioneins, and redox balance were quantified. Immunoreactivity analysis of proteins involved in metabolic and mitogenic insulin signaling was performed. The results showed that a cadmium NOAEL dose after 15 days of exposure causes ROS and mitogenic arm of insulin signaling to increase while hepatic glycogen diminishes. At 30 days, Cd accumulation accentuated ROS production, hepatic triglyceride overaccumulation, and mitogenic signals that develop insulin resistance. Finally, inflammation and lipid peroxidation appear after 60 days of Cd exposure, while lipids and carbohydrate homeostasis deteriorate. In conclusion, environmental exposure to cadmium NAOEL dose causes hepatic Cd accumulation and ROS overproduction that chronically declines the antioxidant defense, deteriorates metabolic homeostasis associated with the mitogenic pathway of insulin signaling, and induces insulin resistance.
Collapse
Affiliation(s)
- Victor Enrique Sarmiento-Ortega
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, Chemistry Department, Meritorious Autonomous University of Puebla, 14 South, FCQ1, Ciudad Universitaria, C.P. 72560, Puebla, Mexico
| | - Diana Moroni-González
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, Chemistry Department, Meritorious Autonomous University of Puebla, 14 South, FCQ1, Ciudad Universitaria, C.P. 72560, Puebla, Mexico
| | - Alfonso Diaz
- Department of Pharmacy, Faculty of Chemistry Science, Meritorious Autonomous University of Puebla, 22 South, FCQ9, Ciudad Universitaria, C.P. 72560, Puebla, Mexico
| | - Eduardo Brambila
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, Chemistry Department, Meritorious Autonomous University of Puebla, 14 South, FCQ1, Ciudad Universitaria, C.P. 72560, Puebla, Mexico
| | - Samuel Treviño
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, Chemistry Department, Meritorious Autonomous University of Puebla, 14 South, FCQ1, Ciudad Universitaria, C.P. 72560, Puebla, Mexico.
| |
Collapse
|
10
|
Sarmiento-Ortega VE, Moroni-González D, Diaz A, García-González MÁ, Brambila E, Treviño S. Hepatic Insulin Resistance Model in the Male Wistar Rat Using Exogenous Insulin Glargine Administration. Metabolites 2023; 13:572. [PMID: 37110230 PMCID: PMC10144445 DOI: 10.3390/metabo13040572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/04/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Metabolic diseases are a worldwide health problem. Insulin resistance (IR) is their distinctive hallmark. For their study, animal models that provide reliable information are necessary, permitting the analysis of the cluster of abnormalities that conform to it, its progression, and time-dependent molecular modifications. We aimed to develop an IR model by exogenous insulin administration. The effective dose of insulin glargine to generate hyperinsulinemia but without hypoglycemia was established. Then, two groups (control and insulin) of male Wistar rats of 100 g weight were formed. The selected dose (4 U/kg) was administered for 15, 30, 45, and 60 days. Zoometry, a glucose tolerance test, insulin response, IR, and the serum lipid profile were assessed. We evaluated insulin signaling, glycogenesis and lipogenesis, redox balance, and inflammation in the liver. Results showed an impairment of glucose tolerance, dyslipidemia, hyperinsulinemia, and peripheral and time-dependent selective IR. At the hepatic level, insulin signaling was impaired, resulting in reduced hepatic glycogen levels and triglyceride accumulation, an increase in the ROS level with MAPK-ERK1/2 response, and mild pro-oxidative microenvironmental sustained by MT, GSH, and GR activity. Hepatic IR coincides with additions in MAPK-p38, NF-κB, and zoometric changes. In conclusion, daily insulin glargine administration generated a progressive IR model. At the hepatic level, the IR was combined with oxidative conditions but without inflammation.
Collapse
Affiliation(s)
- Victor Enrique Sarmiento-Ortega
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Meritorious Autonomous University of Puebla, 14 Sur. FCQ1, Ciudad Universitaria, Puebla City 72560, Mexico
| | - Diana Moroni-González
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Meritorious Autonomous University of Puebla, 14 Sur. FCQ1, Ciudad Universitaria, Puebla City 72560, Mexico
| | - Alfonso Diaz
- Department of Pharmacy, Faculty of Chemistry Science, Meritorious Autonomous University of Puebla, 22 South, FCQ9, Ciudad Universitaria, Puebla City 72560, Mexico
| | - Miguel Ángel García-González
- Laboratory of Clinical Pharmacy, Faculty of Chemistry Science, Meritorious Autonomous University of Puebla, 22 South, FCQ10, Ciudad Universitaria, Puebla City 72560, Mexico
| | - Eduardo Brambila
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Meritorious Autonomous University of Puebla, 14 Sur. FCQ1, Ciudad Universitaria, Puebla City 72560, Mexico
| | - Samuel Treviño
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Meritorious Autonomous University of Puebla, 14 Sur. FCQ1, Ciudad Universitaria, Puebla City 72560, Mexico
| |
Collapse
|
11
|
Kariya Y, Honma M, Tokuda K, Konagaya A, Suzuki H. Utility of constraints reflecting system stability on analyses for biological models. PLoS Comput Biol 2022; 18:e1010441. [PMID: 36084151 PMCID: PMC9491612 DOI: 10.1371/journal.pcbi.1010441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 09/21/2022] [Accepted: 07/26/2022] [Indexed: 12/03/2022] Open
Abstract
Simulating complex biological models consisting of multiple ordinary differential equations can aid in the prediction of the pharmacological/biological responses; however, they are often hampered by the availability of reliable kinetic parameters. In the present study, we aimed to discover the properties of behaviors without determining an optimal combination of kinetic parameter values (parameter set). The key idea was to collect as many parameter sets as possible. Given that many systems are biologically stable and resilient (BSR), we focused on the dynamics around the steady state and formulated objective functions for BSR by partial linear approximation of the focused region. Using the objective functions and modified global cluster Newton method, we developed an algorithm for a thorough exploration of the allowable parameter space for biological systems (TEAPS). We first applied TEAPS to the NF-κB signaling model. This system shows a damped oscillation after stimulation and seems to fit the BSR constraint. By applying TEAPS, we found several directions in parameter space which stringently determines the BSR property. In such directions, the experimentally fitted parameter values were included in the range of the obtained parameter sets. The arachidonic acid metabolic pathway model was used as a model related to pharmacological responses. The pharmacological effects of nonsteroidal anti-inflammatory drugs were simulated using the parameter sets obtained by TEAPS. The structural properties of the system were partly extracted by analyzing the distribution of the obtained parameter sets. In addition, the simulations showed inter-drug differences in prostacyclin to thromboxane A2 ratio such that aspirin treatment tends to increase the ratio, while rofecoxib treatment tends to decrease it. These trends are comparable to the clinical observations. These results on real biological models suggest that the parameter sets satisfying the BSR condition can help in finding biologically plausible parameter sets and understanding the properties of biological systems. We propose a new method to analyze the properties of biological dynamic models, which we named TEAPS (Thorough Exploration of Allowable Parameter Space). TEAPS can thoroughly determine combinations of parameter values for ordinary differential equations with which an initial state in a certain range converges to a particular fixed point. This stable and resilient behavior is a characteristic shared with many biological systems, including metabolic systems and intracellular signaling systems. Therefore, this thorough search outlined the possible parameter space as biological systems for target models, which helps to understand the system constraints when the target systems behave dynamically. The obtained parameter space can be used as an initial space for parameter tuning. For models that include a large number of parameters, the parameter space to be searched in the parameter tuning process is too large; therefore, narrowing down the space by TEAPS potentially contributes to the analysis of the dynamics of complicated biological models. Thus, our approach can partly overcome the current problem in parameter tuning and can advance the computational dynamic analyses of biological systems.
Collapse
Affiliation(s)
- Yoshiaki Kariya
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masashi Honma
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| | - Keita Tokuda
- Department of Computer Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akihiko Konagaya
- Molecular Robotics Research Institute, Limited, Kyowa Create Dai-ichi, Minato-ku, Tokyo, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
12
|
Scalable reaction network modeling with automatic validation of consistency in Event-B. Sci Rep 2022; 12:1287. [PMID: 35079072 PMCID: PMC8789811 DOI: 10.1038/s41598-022-05308-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/08/2021] [Indexed: 11/27/2022] Open
Abstract
Constructing a large biological model is a difficult, error-prone process. Small errors in writing a part of the model cascade to the system level and their sources are difficult to trace back. In this paper we extend a recent approach based on Event-B, a state-based formal method with refinement as its central ingredient, allowing us to validate for model consistency step-by-step in an automated way. We demonstrate this approach on a model of the heat shock response in eukaryotes and its scalability on a model of the \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\mathsf {ErbB}$$\end{document}ErbB signaling pathway. All consistency properties of the model were proved automatically with computer support.
Collapse
|
13
|
Jiang Y, Xin N, Xiong Y, Guo Y, Yuan Y, Zhang Q, Gong P. αCGRP Regulates Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells Through ERK1/2 and p38 MAPK Signaling Pathways. Cell Transplant 2022; 31:9636897221107636. [PMID: 35758252 PMCID: PMC9247368 DOI: 10.1177/09636897221107636] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
As a typical neuropeptide richly distributed in central and peripheral nervous
systems, α-calcitonin-gene-related peptide (αCGRP) has recently been found to
play a crucial role in bone development and metabolism, but the mechanisms
involved are not fully uncovered. Here, this study aimed to investigate the
effects and underlying molecular mechanisms of αCGRP in regulating the
osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Using
microarray technology, gene ontology (GO) and kyoto encyclopedia of genes and
genomes (KEGG) analyses revealed that osteogenic properties of BMSCs were
facilitated and mitogen-activated protein kinase (MAPK) signaling pathway was
upregulated by αCGRP in this process. Through western blot assay, we proved that
αCGRP led to an increased phosphorylation level of extracellular
signal-regulated kinases 1 and 2 (ERK1/2) and p38 MAPK signaling cascades in a
time-dependent manner. And αCGRP could promote differentiative capacity of
BMSCs, showing upregulated mRNA and protein expression level of alkaline
phosphatase (Alp), collagen type 1 (Col-1), osteopontin (Opn), and runt-related
transcription factor 2 (Runx2), as well as increased ALP activity and calcified
nodules. The addition of ERK1/2 or p38 MAPK inhibitor—U0126 or SB203580,
resulted in an impaired osteogenic differentiation of BMSCs. Besides,
inactivation of this signal transduction had negative impacts on proliferative
activity and apoptotic process of αCGRP-mediated BMSCs. Our findings
demonstrated that MAPK signaling pathway, at least in part, was responsible for
the enhanced BMSCs’ osteogenesis induced by αCGRP, which might offer us
promising strategies for bone-related disorders.
Collapse
Affiliation(s)
- Yixuan Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Na Xin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanjun Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Jinjiang Out-Patient Section, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ying Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Rahman I, Athar MT, Islam M. Type 2 Diabetes, Obesity, and Cancer Share Some Common and Critical Pathways. Front Oncol 2021; 10:600824. [PMID: 33552973 PMCID: PMC7855858 DOI: 10.3389/fonc.2020.600824] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetes and cancer are among the most frequent and complex diseases. Epidemiological evidence showed that the patients suffering from diabetes are significantly at higher risk for a number of cancer types. There are a number of evidence that support the hypothesis that these diseases are interlinked, and obesity may aggravate the risk(s) of type 2 diabetes and cancer. Multi-level unwanted alterations such as (epi-)genetic alterations, changes at the transcriptional level, and altered signaling pathways (receptor, cytoplasmic, and nuclear level) are the major source which promotes a number of complex diseases and such heterogeneous level of complexities are considered as the major barrier in the development of therapeutic agents. With so many known challenges, it is critical to understand the relationships and the commonly shared causes between type 2 diabetes and cancer, which is difficult to unravel and understand. Furthermore, the real complexity arises from contended corroborations that specific drug(s) (individually or in combination) during the treatment of type 2 diabetes may increase or decrease the cancer risk or affect cancer prognosis. In this review article, we have presented the recent and most updated evidence from the studies where the origin, biological background, the correlation between them have been presented or proved. Furthermore, we have summarized the methodological challenges and tasks that are frequently encountered. We have also outlined the physiological links between type 2 diabetes and cancers. Finally, we have presented and summarized the outline of the hallmarks for both these diseases, diabetes and cancer.
Collapse
Affiliation(s)
- Ishrat Rahman
- Department of Basic Dental Sciences, College of Dentistry, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Md Tanwir Athar
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Mozaffarul Islam
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Jiang SL, Fang DA, Xu DP. Transcriptome changes of Takifugu obscurus liver after acute exposure to phenanthrene. Physiol Genomics 2021; 53:116-124. [PMID: 33459152 DOI: 10.1152/physiolgenomics.00100.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Phenanthrene (Phe) is a model compound in polycyclic aromatic hydrocarbon (PAH) research. Reportedly, Phe treatment induced oxidative stress and histological disorders to Takifugu obscurus liver. In this study, to further explore the molecular responses of T. obscurus liver to Phe exposure, transcriptome sequencing was applied to compare mRNA transcription profiles between Phe treatment and the control. Compared with the control, 1,581 and 1,428 genes were significantly upregulated and downregulated in Phe treatment, respectively. Further analysis revealed that Phe treatment mainly upregulated genes in Ras-MAPK and PI3K-akt signaling pathways, which represented insulin resistance and further activated the FOXO signaling pathway. The triacylglycerol biosynthesis was promoted but the gluconeogenesis process was inhibited in response to Phe treatment, demonstrating that Phe exposure disturbed the sugar and lipid metabolism. Moreover, Phe treatment upregulated the Apelin-APJ and ErbB signaling pathways, promoting angiogenesis in T. obscurus liver. Insulin resistance, promoted triacylglycerol biosynthesis, and angiogenesis might explain the molecular mechanisms underlying carcinogenic toxicity of Phe. Overall, this study provides new insights to understand the environmental risk of Phe to fishes.
Collapse
Affiliation(s)
- Shu-Lun Jiang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Di-An Fang
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| | - Dong-Po Xu
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, China
| |
Collapse
|
16
|
Treviño S, Diaz A. Vanadium and insulin: Partners in metabolic regulation. J Inorg Biochem 2020; 208:111094. [PMID: 32438270 DOI: 10.1016/j.jinorgbio.2020.111094] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022]
Abstract
Since the 1970s, the biological role of vanadium compounds has been discussed as insulin-mimetic or insulin-enhancer agents. The action of vanadium compounds has been investigated to determine how they influence the insulin signaling pathway. Khan and coworkers proposed key proteins for the insulin pathway study, introducing the concept "critical nodes". In this review, we also considered critical kinases and phosphatases that participate in this pathway, which will permit a better comprehension of a critical node, where vanadium can act: a) insulin receptor, insulin receptor substrates, and protein tyrosine phosphatases; b) phosphatidylinositol 3'-kinase, 3-phosphoinositide-dependent protein kinase and mammalian target of rapamycin complex, protein kinase B, and phosphatase and tensin homolog; and c) insulin receptor substrates and mitogen-activated protein kinases, each node having specific negative modulators. Additionally, leptin signaling was considered because together with insulin, it modulates glucose and lipid homeostasis. Even in recent literature, the possibility of vanadium acting against metabolic diseases or cancer is confirmed although the mechanisms of action are not well understood because these critical nodes have not been systematically investigated. Through this review, we establish that vanadium compounds mainly act as phosphatase inhibitors and hypothesize on their capacity to affect kinases, which are critical to other hormones that also act on common parts of the insulin pathway.
Collapse
Affiliation(s)
- Samuel Treviño
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, University Autonomous of Puebla, 14 South. FCQ1, University City, Puebla, C.P. 72560, Mexico.
| | - Alfonso Diaz
- Department of Pharmacy, Faculty of Chemistry Science, University Autonomous of Puebla, 22 South, FCQ9, University City, Puebla, C.P. 72560, Mexico.
| |
Collapse
|
17
|
It's about time: Analysing simplifying assumptions for modelling multi-step pathways in systems biology. PLoS Comput Biol 2020; 16:e1007982. [PMID: 32598362 PMCID: PMC7351226 DOI: 10.1371/journal.pcbi.1007982] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 07/10/2020] [Accepted: 05/27/2020] [Indexed: 11/19/2022] Open
Abstract
Thoughtful use of simplifying assumptions is crucial to make systems biology models tractable while still representative of the underlying biology. A useful simplification can elucidate the core dynamics of a system. A poorly chosen assumption can, however, either render a model too complicated for making conclusions or it can prevent an otherwise accurate model from describing experimentally observed dynamics. Here, we perform a computational investigation of sequential multi-step pathway models that contain fewer pathway steps than the system they are designed to emulate. We demonstrate when such models will fail to reproduce data and how detrimental truncation of a pathway leads to detectable signatures in model dynamics and its optimised parameters. An alternative assumption is suggested for simplifying such pathways. Rather than assuming a truncated number of pathway steps, we propose to use the assumption that the rates of information propagation along the pathway is homogeneous and, instead, letting the length of the pathway be a free parameter. We first focus on linear pathways that are sequential and have first-order kinetics, and we show how this assumption results in a three-parameter model that consistently outperforms its truncated rival and a delay differential equation alternative in recapitulating observed dynamics. We then show how the proposed assumption allows for similarly terse and effective models of non-linear pathways. Our results provide a foundation for well-informed decision making during model simplifications.
Collapse
|
18
|
Miningou N, Blackwell KT. The road to ERK activation: Do neurons take alternate routes? Cell Signal 2020; 68:109541. [PMID: 31945453 PMCID: PMC7127974 DOI: 10.1016/j.cellsig.2020.109541] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/11/2020] [Accepted: 01/12/2020] [Indexed: 01/29/2023]
Abstract
The ERK cascade is a central signaling pathway that regulates a wide variety of cellular processes including proliferation, differentiation, learning and memory, development, and synaptic plasticity. A wide range of inputs travel from the membrane through different signaling pathway routes to reach activation of one set of output kinases, ERK1&2. The classical ERK activation pathway beings with growth factor activation of receptor tyrosine kinases. Numerous G-protein coupled receptors and ionotropic receptors also lead to ERK through increases in the second messengers calcium and cAMP. Though both types of pathways are present in diverse cell types, a key difference is that most stimuli to neurons, e.g. synaptic inputs, are transient, on the order of milliseconds to seconds, whereas many stimuli acting on non-neural tissue, e.g. growth factors, are longer duration. The ability to consolidate these inputs to regulate the activation of ERK in response to diverse signals raises the question of which factors influence the difference in ERK activation pathways. This review presents both experimental studies and computational models aimed at understanding the control of ERK activation and whether there are fundamental differences between neurons and other cells. Our main conclusion is that differences between cell types are quite subtle, often related to differences in expression pattern and quantity of some molecules such as Raf isoforms. In addition, the spatial location of ERK is critical, with regulation by scaffolding proteins producing differences due to colocalization of upstream molecules that may differ between neurons and other cells.
Collapse
Affiliation(s)
- Nadiatou Miningou
- Department of Chemistry and Biochemistry, George Mason University, Fairfax, VA 22030, United States of America
| | - Kim T Blackwell
- Interdisciplinary Program in Neuroscience and Bioengineering Department, George Mason University, Fairfax, VA 22030, United States of America.
| |
Collapse
|
19
|
Cowan AE, Mendes P, Blinov ML. ModelBricks-modules for reproducible modeling improving model annotation and provenance. NPJ Syst Biol Appl 2019; 5:37. [PMID: 31602314 PMCID: PMC6783478 DOI: 10.1038/s41540-019-0114-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/28/2019] [Indexed: 01/27/2023] Open
Abstract
Most computational models in biology are built and intended for "single-use"; the lack of appropriate annotation creates models where the assumptions are unknown, and model elements are not uniquely identified. Simply recreating a simulation result from a publication can be daunting; expanding models to new and more complex situations is a herculean task. As a result, new models are almost always created anew, repeating literature searches for kinetic parameters, initial conditions and modeling specifics. It is akin to building a brick house starting with a pile of clay. Here we discuss a concept for building annotated, reusable models, by starting with small well-annotated modules we call ModelBricks. Curated ModelBricks, accessible through an open database, could be used to construct new models that will inherit ModelBricks annotations and thus be easier to understand and reuse. Key features of ModelBricks include reliance on a commonly used standard language (SBML), rule-based specification describing species as a collection of uniquely identifiable molecules, association with model specific numerical parameters, and more common annotations. Physical bricks can vary substantively; likewise, to be useful the structure of ModelBricks must be highly flexible-it should encapsulate mechanisms from single reactions to multiple reactions in a complex process. Ultimately, a modeler would be able to construct large models by using multiple ModelBricks, preserving annotations and provenance of model elements, resulting in a highly annotated model. We envision the library of ModelBricks to rapidly grow from community contributions. Persistent citable references will incentivize model creators to contribute new ModelBricks.
Collapse
Affiliation(s)
- Ann E. Cowan
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT USA
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT USA
| | - Pedro Mendes
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT USA
- Center for Quantitative Medicine, UConn Health, Farmington, CT USA
- Department of Cell Biology, UConn Health, Farmington, CT USA
| | - Michael L. Blinov
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT USA
| |
Collapse
|
20
|
Identifiability from a Few Species for a Class of Biochemical Reaction Networks. Bull Math Biol 2019; 81:2133-2175. [PMID: 30945101 DOI: 10.1007/s11538-019-00594-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/04/2019] [Indexed: 10/27/2022]
Abstract
Under mass-action kinetics, biochemical reaction networks give rise to polynomial autonomous dynamical systems whose parameters are often difficult to estimate. We deal in this paper with the problem of identifying the kinetic parameters of a class of biochemical networks which are abundant, such as multisite phosphorylation systems and phosphorylation cascades (for example, MAPK cascades). For any system of this class, we explicitly exhibit a single species for each connected component of the associated digraph such that the successive total derivatives of its concentration allow us to identify all the parameters occurring in the component. The number of derivatives needed is bounded essentially by the length of the corresponding connected component of the digraph. Moreover, in the particular case of the cascades, we show that the parameters can be identified from a bounded number of successive derivatives of the last product of the last layer. This theoretical result induces also a heuristic interpolation-based identifiability procedure to recover the values of the rate constants from exact measurements.
Collapse
|
21
|
Lam I, Pickering CM, Mac Gabhann F. Context-dependent regulation of receptor tyrosine kinases: Insights from systems biology approaches. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2019; 11:e1437. [PMID: 30255986 PMCID: PMC6537588 DOI: 10.1002/wsbm.1437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 06/07/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022]
Abstract
Receptor tyrosine kinases (RTKs) are cell membrane proteins that provide cells with the ability to sense proteins in their environments. Many RTKs are essential to development and organ growth. Derangement of RTKs-by mutation or by overexpression-is central to several developmental and adult disorders including cancer, short stature, and vascular pathologies. The mechanism of action of RTKs is complex and is regulated by contextual components, including the existence of multiple competing ligands and receptors in many families, the intracellular location of the RTK, the dynamic and cell-specific coexpression of other RTKs, and the commonality of downstream signaling pathways. This means that both the state of the cell and the microenvironment outside the cell play a role, which makes sense given the pivotal location of RTKs as the nexus linking the extracellular milieu to intracellular signaling and modification of cell behavior. In this review, we describe these different contextual components through the lens of systems biology, in which both computational modeling and experimental "omics" approaches have been used to better understand RTK networks. The complexity of these networks is such that using these systems biology approaches is necessary to get a handle on the mechanisms of pathology and the design of therapeutics targeting RTKs. In particular, we describe in detail three concrete examples (involving ErbB3, VEGFR2, and AXL) that illustrate how systems approaches can reveal key mechanistic and therapeutic insights. This article is categorized under: Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Mechanistic Models Translational, Genomic, and Systems Medicine > Therapeutic Methods.
Collapse
Affiliation(s)
- Inez Lam
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Christina M Pickering
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Feilim Mac Gabhann
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
22
|
Iancu B, Sanwal U, Gratie C, Petre I. Refinement-based modeling of the ErbB signaling pathway. Comput Biol Med 2019; 106:91-96. [PMID: 30708221 DOI: 10.1016/j.compbiomed.2019.01.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/18/2019] [Accepted: 01/19/2019] [Indexed: 11/26/2022]
Abstract
The construction of large scale biological models is a laborious task, which is often addressed by adopting iterative routines for model augmentation, adding certain details to an initial high level abstraction of the biological phenomenon of interest. Refitting a model at every step of its development is time consuming and computationally intensive. The concept of model refinement brings about an effective alternative by providing adequate parameter values that ensure the preservation of its quantitative fit at every refinement step. We demonstrate this approach by constructing the largest-ever refinement-based biomodel, consisting of 421 species and 928 reactions. We start from an already fit, relatively small literature model whose consistency we check formally. We then construct the final model through an algorithmic step-by-step refinement procedure that ensures the preservation of the model's fit.
Collapse
Affiliation(s)
- Bogdan Iancu
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, Finland; Department of Computer Science, Åbo Akademi University, Finland
| | - Usman Sanwal
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, Finland; Department of Computer Science, Åbo Akademi University, Finland
| | - Cristian Gratie
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, Finland; Department of Computer Science, Åbo Akademi University, Finland
| | - Ion Petre
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, Finland; Department of Mathematics and Statistics, University of Turku, Finland; National Institute for Research and Development in Biological Sciences, Romania.
| |
Collapse
|
23
|
Arkun Y, Yasemi M. Dynamics and control of the ERK signaling pathway: Sensitivity, bistability, and oscillations. PLoS One 2018; 13:e0195513. [PMID: 29630631 PMCID: PMC5891012 DOI: 10.1371/journal.pone.0195513] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/22/2018] [Indexed: 02/06/2023] Open
Abstract
Cell signaling is the process by which extracellular information is transmitted into the cell to perform useful biological functions. The ERK (extracellular-signal-regulated kinase) signaling controls several cellular processes such as cell growth, proliferation, differentiation and apoptosis. The ERK signaling pathway considered in this work starts with an extracellular stimulus and ends with activated (double phosphorylated) ERK which gets translocated into the nucleus. We model and analyze this complex pathway by decomposing it into three functional subsystems. The first subsystem spans the initial part of the pathway from the extracellular growth factor to the formation of the SOS complex, ShC-Grb2-SOS. The second subsystem includes the activation of Ras which is mediated by the SOS complex. This is followed by the MAPK subsystem (or the Raf-MEK-ERK pathway) which produces the double phosphorylated ERK upon being activated by Ras. Although separate models exist in the literature at the subsystems level, a comprehensive model for the complete system including the important regulatory feedback loops is missing. Our dynamic model combines the existing subsystem models and studies their steady-state and dynamic interactions under feedback. We establish conditions under which bistability and oscillations exist for this important pathway. In particular, we show how the negative and positive feedback loops affect the dynamic characteristics that determine the cellular outcome.
Collapse
Affiliation(s)
- Yaman Arkun
- Department of Chemical and Biological Engineering, Koc University, Rumeli Feneri Yolu, Sariyer, Istanbul, Turkey
- * E-mail:
| | - Mohammadreza Yasemi
- Department of Chemical and Biological Engineering, Koc University, Rumeli Feneri Yolu, Sariyer, Istanbul, Turkey
| |
Collapse
|
24
|
Irmawati A, Jasmin N, Sidarningsih. The effect of moderate exercise on the elevation of Bax/Bcl-2 ratio in oral squamous epithelial cells induced by benzopyrene. Vet World 2018; 11:177-180. [PMID: 29657400 PMCID: PMC5891871 DOI: 10.14202/vetworld.2018.177-180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/08/2018] [Indexed: 11/29/2022] Open
Abstract
Aim: The aim of this study was to analyze the effect of moderate exercise on the elevation of Bax/Bcl-2 ratio. Materials and Methods: Eighteen Mus musculus strain Swiss Webster (Balb/c) were divided into three groups (n=6). K1 and K2 had contact with water 3 times/week for 12 weeks, while the members of the K3 group swam 3 times/week for 12 weeks while carrying load weighed 3% of their body weight. After 5 weeks, they were induced with 0.04 ml oleum olivarum (K1), 0.08 mg benzopyrene/0.04 ml oleum olivarum (K2, K3) 3 times/week for 4 weeks. Immunohistochemistry assays were used to determine the ratio of Bax/Bcl-2 expression. The results were analyzed using an independent t-test. Result: The Bax/Bcl-2 ratio increased significantly in K3 compared to K2 (p=0.00). Conclusion: Moderate exercise could increase the Bax/Bcl-2 ratio in oral squamous epithelial cells induced by benzopyrene.
Collapse
Affiliation(s)
- Anis Irmawati
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Nadira Jasmin
- Undergraduate student of Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Sidarningsih
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
25
|
Learning to read and write in evolution: from static pseudoenzymes and pseudosignalers to dynamic gear shifters. Biochem Soc Trans 2017; 45:635-652. [PMID: 28620026 DOI: 10.1042/bst20160281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 11/17/2022]
Abstract
We present a systems biology view on pseudoenzymes that acknowledges that genes are not selfish: the genome is. With network function as the selectable unit, there has been an evolutionary bonus for recombination of functions of and within proteins. Many proteins house a functionality by which they 'read' the cell's state, and one by which they 'write' and thereby change that state. Should the writer domain lose its cognate function, a 'pseudoenzyme' or 'pseudosignaler' arises. GlnK involved in Escherichia coli ammonia assimilation may well be a pseudosignaler, associating 'reading' the nitrogen state of the cell to 'writing' the ammonium uptake activity. We identify functional pseudosignalers in the cyclin-dependent kinase complexes regulating cell-cycle progression. For the mitogen-activated protein kinase pathway, we illustrate how a 'dead' pseudosignaler could produce potentially selectable functionalities. Four billion years ago, bioenergetics may have shuffled 'electron-writers', producing various networks that all served the same function of anaerobic ATP synthesis and carbon assimilation from hydrogen and carbon dioxide, but at different ATP/acetate ratios. This would have enabled organisms to deal with variable challenges of energy need and substrate supply. The same principle might enable 'gear-shifting' in real time, by dynamically generating different pseudo-redox enzymes, reshuffling their coenzymes, and rerouting network fluxes. Non-stationary pH gradients in thermal vents together with similar such shuffling mechanisms may have produced a first selectable proton-motivated pyrophosphate synthase and subsequent ATP synthase. A combination of functionalities into enzymes, signalers, and the pseudo-versions thereof may offer fitness in terms of plasticity, both in real time and in evolution.
Collapse
|
26
|
Kim DH, Kim DK, Zhou K, Park S, Kwon Y, Jeong MG, Lee NK, Ryu SH. Single particle tracking-based reaction progress kinetic analysis reveals a series of molecular mechanisms of cetuximab-induced EGFR processes in a single living cell. Chem Sci 2017; 8:4823-4832. [PMID: 28959404 PMCID: PMC5602156 DOI: 10.1039/c7sc01159h] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/21/2017] [Indexed: 01/18/2023] Open
Abstract
Cellular processes occur through the orchestration of multi-step molecular reactions. Reaction progress kinetic analysis (RPKA) can provide the mechanistic details to elucidate the multi-step molecular reactions. However, current tools have limited ability to simultaneously monitor dynamic variations in multiple complex states at the single molecule level to apply RPKA in living cells. In this research, a single particle tracking-based reaction progress kinetic analysis (sptRPKA) was developed to simultaneously determine the kinetics of multiple states of protein complexes in the membrane of a single living cell. The subpopulation ratios of different states were quantitatively (and statistically) reliably extracted from the diffusion coefficient distribution rapidly acquired by single particle tracking at constant and high density over a long period of time using super-resolution microscopy. Using sptRPKA, a series of molecular mechanisms of epidermal growth factor receptor (EGFR) cellular processing induced by cetuximab were investigated. By comprehensively measuring the rate constants and cooperativity of the molecular reactions involving four EGFR complex states, a previously unknown intermediate state was identified that represents the rate limiting step responsible for the selectivity of cetuximab-induced EGFR endocytosis to cancer cells.
Collapse
Affiliation(s)
- Do-Hyeon Kim
- Department of Life Sciences , Pohang University of Science and Technology , Pohang , 790-784 , Republic of Korea .
| | - Dong-Kyun Kim
- School of Interdisciplinary Bioscience and Bioengineering , Pohang University of Science and Technology , Pohang , 790-784 , Republic of Korea
| | - Kai Zhou
- Department of Life Sciences , Pohang University of Science and Technology , Pohang , 790-784 , Republic of Korea .
| | - Soyeon Park
- Department of Life Sciences , Pohang University of Science and Technology , Pohang , 790-784 , Republic of Korea .
| | - Yonghoon Kwon
- Department of Life Sciences , Pohang University of Science and Technology , Pohang , 790-784 , Republic of Korea .
| | - Min Gyu Jeong
- Integrative Biosciences and Biotechnology , Pohang University of Science and Technology , Pohang , 790-784 , Republic of Korea
| | - Nam Ki Lee
- School of Interdisciplinary Bioscience and Bioengineering , Pohang University of Science and Technology , Pohang , 790-784 , Republic of Korea.,Department of Chemistry , Seoul National University , Seoul , 08826 , Republic of Korea .
| | - Sung Ho Ryu
- Department of Life Sciences , Pohang University of Science and Technology , Pohang , 790-784 , Republic of Korea . .,School of Interdisciplinary Bioscience and Bioengineering , Pohang University of Science and Technology , Pohang , 790-784 , Republic of Korea
| |
Collapse
|
27
|
Álvaro-Bartolomé M, Salort G, García-Sevilla JA. Disruption of brain MEK-ERK sequential phosphorylation and activation during midazolam-induced hypnosis in mice: Roles of GABA A receptor, MEK1 inactivation, and phosphatase MKP-3. Prog Neuropsychopharmacol Biol Psychiatry 2017; 75:84-93. [PMID: 28111292 DOI: 10.1016/j.pnpbp.2017.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 01/08/2023]
Abstract
Midazolam is a positive allosteric modulator at GABAA receptor that induces a short hypnosis and neuroplasticity, in which the sequential phosphorylation of MEK1/2 and ERK1/2 was shown to play a role. This study investigated the parallel activation of p-MEK and p-ERK and regulatory mechanisms induced by midazolam through the stimulation of GABAA receptors in the mouse brain. During the time course of midazolam (60mg/kg)-induced sleep in mice (lasting for about 2h) p-Ser217/221 MEK1/2 was increased (+146% to +258%) whereas, unexpectedly, p-Tyr204/Thr202 ERK1/2 was found decreased (-16% to -38%), revealing uncoupling of MEK to ERK signals in various brain regions. Midazolam-induced p-MEK1/2 upregulation was prevented by pretreatment (30min) with flumazenil (10mg/kg), indicating the involvement of GABAA receptors. Also unexpectedly, midazolam-induced p-ERK1/2 downregulation was not prevented by flumazenil (10 or 30mg/kg). Notably, during midazolam-induced sleep the content of inactivated p-Thr286 MEK1, which can dampen ERK1/2 activation, was increased (+33% to +149%) through a mechanism sensitive to flumazenil (10mg/kg). Midazolam also increased MKP-3 (+13% to +73%) content and this upregulation was prevented by flumazenil (10mg/kg); an effect suggesting ERK inactivation because MKP-3 is the phosphatase selective for ERK1/2 dephosphorylation. The results indicate that during midazolam-induced sleep in mice there is an uncoupling of p-MEK (increased) to p-ERK (decreased) signals. p-ERK1/2 downregulation (not involving GABAA receptors) is the result of increased inactivated MEK1 and phosphatase MKP-3 (both effects involving GABAA receptors). These findings are relevant for the neurobiology and clinical use of benzodiazepines.
Collapse
Affiliation(s)
- María Álvaro-Bartolomé
- Laboratory of Neuropharmacology, IUNICS-IdISPa, University of the Balearic Islands (UIB), Palma de Mallorca, Spain; Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - Glòria Salort
- Laboratory of Neuropharmacology, IUNICS-IdISPa, University of the Balearic Islands (UIB), Palma de Mallorca, Spain; Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - Jesús A García-Sevilla
- Laboratory of Neuropharmacology, IUNICS-IdISPa, University of the Balearic Islands (UIB), Palma de Mallorca, Spain; Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain.
| |
Collapse
|
28
|
Purutçuoğlu V, Ayyıldız E, Wit E. Comparison of two inference approaches in Gaussian graphical models. TURKISH JOURNAL OF BIOCHEMISTRY 2017. [DOI: 10.1515/tjb-2016-0298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractIntroduction:The Gaussian Graphical Model (GGM) is one of the well-known probabilistic models which is based on the conditional independency of nodes in the biological system. Here, we compare the estimates of the GGM parameters by the graphical lasso (glasso) method and the threshold gradient descent (TGD) algorithm.Methods:We evaluate the performance of both techniques via certain measures such as specificity, F-measure and AUC (area under the curve). The analyses are conducted by Monte Carlo runs under different dimensional systems.Results:The results indicate that the TGD algorithm is more accurate than the glasso method in all selected criteria, whereas, it is more computationally demanding than this method too.Discussion and conclusion:Therefore, in high dimensional systems, we recommend glasso for its computational efficiency in spite of its loss in accuracy and we believe than the computational cost of the TGD algorithm can be improved by suggesting alternative steps in inference of the network.
Collapse
|
29
|
Huang L, Jiang Y, Chen Y. Predicting Drug Combination Index and Simulating the Network-Regulation Dynamics by Mathematical Modeling of Drug-Targeted EGFR-ERK Signaling Pathway. Sci Rep 2017; 7:40752. [PMID: 28102344 PMCID: PMC5244366 DOI: 10.1038/srep40752] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/06/2016] [Indexed: 02/05/2023] Open
Abstract
Synergistic drug combinations enable enhanced therapeutics. Their discovery typically involves the measurement and assessment of drug combination index (CI), which can be facilitated by the development and applications of in-silico CI predictive tools. In this work, we developed and tested the ability of a mathematical model of drug-targeted EGFR-ERK pathway in predicting CIs and in analyzing multiple synergistic drug combinations against observations. Our mathematical model was validated against the literature reported signaling, drug response dynamics, and EGFR-MEK drug combination effect. The predicted CIs and combination therapeutic effects of the EGFR-BRaf, BRaf-MEK, FTI-MEK, and FTI-BRaf inhibitor combinations showed consistent synergism. Our results suggest that existing pathway models may be potentially extended for developing drug-targeted pathway models to predict drug combination CI values, isobolograms, and drug-response surfaces as well as to analyze the dynamics of individual and combinations of drugs. With our model, the efficacy of potential drug combinations can be predicted. Our method complements the developed in-silico methods (e.g. the chemogenomic profile and the statistically-inferenced network models) by predicting drug combination effects from the perspectives of pathway dynamics using experimental or validated molecular kinetic constants, thereby facilitating the collective prediction of drug combination effects in diverse ranges of disease systems.
Collapse
Affiliation(s)
- Lu Huang
- The Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen, 518055, P.R. China
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
- Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, 117543 Singapore
| | - Yuyang Jiang
- The Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen, 518055, P.R. China
| | - Yuzong Chen
- Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, 117543 Singapore
- State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Modeling Cellular Noise Underlying Heterogeneous Cell Responses in the Epidermal Growth Factor Signaling Pathway. PLoS Comput Biol 2016; 12:e1005222. [PMID: 27902699 PMCID: PMC5130170 DOI: 10.1371/journal.pcbi.1005222] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 10/25/2016] [Indexed: 12/03/2022] Open
Abstract
Cellular heterogeneity, which plays an essential role in biological phenomena, such as drug resistance and migration, is considered to arise from intrinsic (i.e., reaction kinetics) and extrinsic (i.e., protein variability) noise in the cell. However, the mechanistic effects of these types of noise to determine the heterogeneity of signal responses have not been elucidated. Here, we report that the output of epidermal growth factor (EGF) signaling activity is modulated by cellular noise, particularly by extrinsic noise of particular signaling components in the pathway. We developed a mathematical model of the EGF signaling pathway incorporating regulation between extracellular signal-regulated kinase (ERK) and nuclear pore complex (NPC), which is necessary for switch-like activation of the nuclear ERK response. As the threshold of switch-like behavior is more sensitive to perturbations than the graded response, the effect of biological noise is potentially critical for cell fate decision. Our simulation analysis indicated that extrinsic noise, but not intrinsic noise, contributes to cell-to-cell heterogeneity of nuclear ERK. In addition, we accurately estimated variations in abundance of the signal proteins between individual cells by direct comparison of experimental data with simulation results using Apparent Measurement Error (AME). AME was constant regardless of whether the protein levels varied in a correlated manner, while covariation among proteins influenced cell-to-cell heterogeneity of nuclear ERK, suppressing the variation. Simulations using the estimated protein abundances showed that each protein species has different effects on cell-to-cell variation in the nuclear ERK response. In particular, variability of EGF receptor, Ras, Raf, and MEK strongly influenced cellular heterogeneity, while others did not. Overall, our results indicated that cellular heterogeneity in response to EGF is strongly driven by extrinsic noise, and that such heterogeneity results from variability of particular protein species that function as sensitive nodes, which may contribute to the pathogenesis of human diseases. Individual cell behaviors are controlled by a variety of noise, such as fluctuations in biochemical reactions, protein variability, molecular diffusion, transcriptional noise, cell-to-cell contact, temperature, and pH. Such cellular noise often interferes with signal responses from external stimuli, and such heterogeneity functions in induction of drug resistance, survival, and migration of cells. Thus, heterogeneous cellular responses have positive and negative roles. However, the regulatory mechanisms that produce cellular heterogeneity are unclear. By mathematical modeling and simulations, we investigated how heterogeneous signaling responses are evoked in the EGF signaling pathway and influence the switch-like activation of nuclear ERK. This study demonstrated that cellular heterogeneity of the EGF signaling response is evoked by cell-to-cell variation of particular signaling proteins, such as EGFR, Ras, Raf, and MEK, which act as sensitive nodes in the pathway. These results suggest that signaling responses in individual cells can be predicted from the levels of proteins of sensitive nodes. This study also suggested that proteins of sensitive nodes may serve as cell survival mechanisms.
Collapse
|
31
|
Gawthrop PJ, Crampin EJ. Modular bond-graph modelling and analysis of biomolecular systems. IET Syst Biol 2016; 10:187-201. [PMID: 27762233 PMCID: PMC8687434 DOI: 10.1049/iet-syb.2015.0083] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 01/05/2016] [Accepted: 01/18/2016] [Indexed: 12/28/2022] Open
Abstract
Bond graphs can be used to build thermodynamically-compliant hierarchical models of biomolecular systems. As bond graphs have been widely used to model, analyse and synthesise engineering systems, this study suggests that they can play the same rôle in the modelling, analysis and synthesis of biomolecular systems. The particular structure of bond graphs arising from biomolecular systems is established and used to elucidate the relation between thermodynamically closed and open systems. Block diagram representations of the dynamics implied by these bond graphs are used to reveal implicit feedback structures and are linearised to allow the application of control-theoretical methods. Two concepts of modularity are examined: computational modularity where physical correctness is retained and behavioural modularity where module behaviour (such as ultrasensitivity) is retained. As well as providing computational modularity, bond graphs provide a natural formulation of behavioural modularity and reveal the sources of retroactivity. A bond graph approach to reducing retroactivity, and thus inter-module interaction, is shown to require a power supply such as that provided by the ATP ⇌ ADP + Pi reaction. The mitogen-activated protein kinase cascade (Raf-MEK-ERK pathway) is used as an illustrative example.
Collapse
Affiliation(s)
- Peter J Gawthrop
- Centre for Systems Genomics, University of Melbourne, Victoria 3010, Australia.
| | - Edmund J Crampin
- ARC Centre of Excellence in Convergent Bio-Nano Science, Melbourne School of Engineering, University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
32
|
Nayak S, Siddiqui JK, Varner JD. Modelling and analysis of an ensemble of eukaryotic translation initiation models. IET Syst Biol 2016; 5:2. [PMID: 21261397 DOI: 10.1049/iet-syb.2009.0065] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Programmed protein synthesis plays an important role in the cell cycle. Deregulated translation has been observed in several cancers. In this study, the authors constructed an ensemble of mathematical models describing the integration of growth factor signals with translation initiation. Using these models, the authors estimated critical structural features of the translation architecture. Sensitivity and robustness analysis with and without growth factors suggested that a balance between competing regulatory programmes governed translation initiation. Proteins such as Akt and mTor promoted initiation by integrating growth factor signals with the assembly of the 80S initiation complex. However, negative regulators such as PTEN and 4EBP1 restrained initiation in the absence of stimulation. Other proteins such as eIF4E were also found to be structurally critical as deletion of amplification of these components resulted in a network incapable of nominal operation. These findings could help understand the molecular basis of translation deregulation observed in cancer and perhaps lead to new anti-cancer therapeutic strategies. [Includes supplementary material].
Collapse
Affiliation(s)
- S Nayak
- Cornell University, School of Chemical and Biomolecular Engineering, Ithaca, USA
| | | | | |
Collapse
|
33
|
Dynamic Modeling and Analysis of the Cross-Talk between Insulin/AKT and MAPK/ERK Signaling Pathways. PLoS One 2016; 11:e0149684. [PMID: 26930065 PMCID: PMC4773096 DOI: 10.1371/journal.pone.0149684] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/02/2016] [Indexed: 12/26/2022] Open
Abstract
Feedback loops play a key role in the regulation of the complex interactions in signal transduction networks. By studying the network of interactions among the biomolecules present in signaling pathways at the systems level, it is possible to understand how the biological functions are regulated and how the diseases emerge from their deregulations. This paper identifies the key feedback loops involved in the cross-talk among the insulin-AKT and MAPK/ERK signaling pathways. We developed a mathematical model that can be used to study the steady-state and dynamic behavior of the interactions among these two important signaling pathways. Modeling analysis and simulation case studies identify the key interaction parameters and the feedback loops that determine the normal and disease phenotypes.
Collapse
|
34
|
Cursons J, Gao J, Hurley DG, Print CG, Dunbar PR, Jacobs MD, Crampin EJ. Regulation of ERK-MAPK signaling in human epidermis. BMC SYSTEMS BIOLOGY 2015. [PMID: 26209520 PMCID: PMC4514964 DOI: 10.1186/s12918-015-0187-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background The skin is largely comprised of keratinocytes within the interfollicular epidermis. Over approximately two weeks these cells differentiate and traverse the thickness of the skin. The stage of differentiation is therefore reflected in the positions of cells within the tissue, providing a convenient axis along which to study the signaling events that occur in situ during keratinocyte terminal differentiation, over this extended two-week timescale. The canonical ERK-MAPK signaling cascade (Raf-1, MEK-1/2 and ERK-1/2) has been implicated in controlling diverse cellular behaviors, including proliferation and differentiation. While the molecular interactions involved in signal transduction through this cascade have been well characterized in cell culture experiments, our understanding of how this sequence of events unfolds to determine cell fate within a homeostatic tissue environment has not been fully characterized. Methods We measured the abundance of total and phosphorylated ERK-MAPK signaling proteins within interfollicular keratinocytes in transverse cross-sections of human epidermis using immunofluorescence microscopy. To investigate these data we developed a mathematical model of the signaling cascade using a normalized-Hill differential equation formalism. Results These data show coordinated variation in the abundance of phosphorylated ERK-MAPK components across the epidermis. Statistical analysis of these data shows that associations between phosphorylated ERK-MAPK components which correspond to canonical molecular interactions are dependent upon spatial position within the epidermis. The model demonstrates that the spatial profile of activation for ERK-MAPK signaling components across the epidermis may be maintained in a cell-autonomous fashion by an underlying spatial gradient in calcium signaling. Conclusions Our data demonstrate an extended phospho-protein profile of ERK-MAPK signaling cascade components across the epidermis in situ, and statistical associations in these data indicate canonical ERK-MAPK interactions underlie this spatial profile of ERK-MAPK activation. Using mathematical modelling we have demonstrated that spatially varying calcium signaling components across the epidermis may be sufficient to maintain the spatial profile of ERK-MAPK signaling cascade components in a cell-autonomous manner. These findings may have significant implications for the wide range of cancer drugs which therapeutically target ERK-MAPK signaling components. Electronic supplementary material The online version of this article (doi:10.1186/s12918-015-0187-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joseph Cursons
- Systems Biology Laboratory, Melbourne School of Engineering, University of Melbourne, Melbourne, Australia. .,NICTA Victoria Research Lab, Melbourne, Australia. .,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Melbourne School of Engineering, University of Melbourne, Melbourne, Australia. .,Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand. .,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand.
| | - Jerry Gao
- Systems Biology Laboratory, Melbourne School of Engineering, University of Melbourne, Melbourne, Australia.
| | - Daniel G Hurley
- Systems Biology Laboratory, Melbourne School of Engineering, University of Melbourne, Melbourne, Australia. .,NICTA Victoria Research Lab, Melbourne, Australia. .,Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand. .,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand. .,Bioinformatics Institute, University of Auckland, Auckland, New Zealand. .,Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Cristin G Print
- Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand. .,Bioinformatics Institute, University of Auckland, Auckland, New Zealand. .,Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - P Rod Dunbar
- Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand. .,School of Biological Sciences, University of Auckland, Auckland, New Zealand.
| | - Marc D Jacobs
- Department of Biology, New Zealand International College, ACG New Zealand, Auckland, New Zealand.
| | - Edmund J Crampin
- Systems Biology Laboratory, Melbourne School of Engineering, University of Melbourne, Melbourne, Australia. .,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Melbourne School of Engineering, University of Melbourne, Melbourne, Australia. .,Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand. .,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand. .,School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia. .,School of Medicine, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
35
|
Westerhoff HV, Nakayama S, Mondeel TDGA, Barberis M. Systems Pharmacology: An opinion on how to turn the impossible into grand challenges. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 15:23-31. [PMID: 26464087 DOI: 10.1016/j.ddtec.2015.06.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/23/2015] [Accepted: 06/25/2015] [Indexed: 11/20/2022]
Abstract
A pharmacology that hits single disease-causing molecules with a single drug passively distributing to the target tissue, was almost ready. Such a pharmacology is not (going to be) effective however: a great many diseases are systems biology diseases; complex networks of some hundred thousand types of molecule, determine the functions that constitute human health, through nonlinear interactions. Malfunctions are caused by a variety of molecular failures at the same time; rarely the same variety in different individuals; in complex constellations of OR and AND logics. Few molecules cause disease single-handedly and few drugs will cure the disease all by themselves when dosed for a limited amount of time. We here discuss the implications that this discovery of the network nature of disease should have for pharmacology. We suggest ways in which pharmacokinetics, pharmacodynamics, but also systems biology and genomics may have to change so as better to deal with systems-biology diseases.
Collapse
Affiliation(s)
- Hans V Westerhoff
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands; Molecular Cell Physiology, Amsterdam Institute for Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands; Manchester Centre for Integrative Systems Biology, The University of Manchester, UK.
| | - Shintaro Nakayama
- Molecular Cell Physiology, Amsterdam Institute for Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands
| | - Thierry D G A Mondeel
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
| |
Collapse
|
36
|
Stimulus-dependent differences in signalling regulate epithelial-mesenchymal plasticity and change the effects of drugs in breast cancer cell lines. Cell Commun Signal 2015; 13:26. [PMID: 25975820 PMCID: PMC4432969 DOI: 10.1186/s12964-015-0106-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/22/2015] [Indexed: 12/18/2022] Open
Abstract
Introduction The normal process of epithelial mesenchymal transition (EMT) is subverted by carcinoma cells to facilitate metastatic spread. Cancer cells rarely undergo a full conversion to the mesenchymal phenotype, and instead adopt positions along the epithelial-mesenchymal axis, a propensity we refer to as epithelial mesenchymal plasticity (EMP). EMP is associated with increased risk of metastasis in breast cancer and consequent poor prognosis. Drivers towards the mesenchymal state in malignant cells include growth factor stimulation or exposure to hypoxic conditions. Methods We have examined EMP in two cell line models of breast cancer: the PMC42 system (PMC42-ET and PMC42-LA sublines) and MDA-MB-468 cells. Transition to a mesenchymal phenotype was induced across all three cell lines using epidermal growth factor (EGF) stimulation, and in MDA-MB-468 cells by hypoxia. We used RNA sequencing to identify gene expression changes that occur as cells transition to a more-mesenchymal phenotype, and identified the cell signalling pathways regulated across these experimental systems. We then used inhibitors to modulate signalling through these pathways, verifying the conclusions of our transcriptomic analysis. Results We found that EGF and hypoxia both drive MDA-MB-468 cells to phenotypically similar mesenchymal states. Comparing the transcriptional response to EGF and hypoxia, we have identified differences in the cellular signalling pathways that mediate, and are influenced by, EMT. Significant differences were observed for a number of important cellular signalling components previously implicated in EMT, such as HBEGF and VEGFA. We have shown that EGF- and hypoxia-induced transitions respond differently to treatment with chemical inhibitors (presented individually and in combinations) in these breast cancer cells. Unexpectedly, MDA-MB-468 cells grown under hypoxic growth conditions became even more mesenchymal following exposure to certain kinase inhibitors that prevent growth-factor induced EMT, including the mTOR inhibitor everolimus and the AKT1/2/3 inhibitor AZD5363. Conclusions While resulting in a common phenotype, EGF and hypoxia induced subtly different signalling systems in breast cancer cells. Our findings have important implications for the use of kinase inhibitor-based therapeutic interventions in breast cancers, where these heterogeneous signalling landscapes will influence the therapeutic response. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0106-x) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
Hierarchical feedback modules and reaction hubs in cell signaling networks. PLoS One 2015; 10:e0125886. [PMID: 25951347 PMCID: PMC4424001 DOI: 10.1371/journal.pone.0125886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 03/24/2015] [Indexed: 11/20/2022] Open
Abstract
Despite much effort, identification of modular structures and study of their organizing and functional roles remain a formidable challenge in molecular systems biology, which, however, is essential in reaching a systematic understanding of large-scale cell regulation networks and hence gaining capacity of exerting effective interference to cell activity. Combining graph theoretic methods with available dynamics information, we successfully retrieved multiple feedback modules of three important signaling networks. These feedbacks are structurally arranged in a hierarchical way and dynamically produce layered temporal profiles of output signals. We found that global and local feedbacks act in very different ways and on distinct features of the information flow conveyed by signal transduction but work highly coordinately to implement specific biological functions. The redundancy embodied with multiple signal-relaying channels and feedback controls bestow great robustness and the reaction hubs seated at junctions of different paths announce their paramount importance through exquisite parameter management. The current investigation reveals intriguing general features of the organization of cell signaling networks and their relevance to biological function, which may find interesting applications in analysis, design and control of bio-networks.
Collapse
|
38
|
Pérez Millán M, Turjanski AG. MAPK's networks and their capacity for multistationarity due to toric steady states. Math Biosci 2015; 262:125-37. [PMID: 25640872 DOI: 10.1016/j.mbs.2014.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 11/26/2014] [Accepted: 12/03/2014] [Indexed: 11/19/2022]
Abstract
Mitogen-activated protein kinase (MAPK) signaling pathways play an essential role in the transduction of environmental stimuli to the nucleus, thereby regulating a variety of cellular processes, including cell proliferation, differentiation and programmed cell death. The components of the MAPK extracellular activated protein kinase (ERK) cascade represent attractive targets for cancer therapy as their aberrant activation is a frequent event among highly prevalent human cancers. MAPK networks are a model for computational simulation, mostly using ordinary and partial differential equations. Key results showed that these networks can have switch-like behavior, bistability and oscillations. In this work, we consider three representative ERK networks, one with a negative feedback loop, which present a binomial steady state ideal under mass-action kinetics. We therefore apply the theoretical result present in to find a set of rate constants that allow two significantly different stable steady states in the same stoichiometric compatibility class for each network. Our approach makes it possible to study certain aspects of the system, such as multistationarity, without relying on simulation, since we do not assume a priori any constant but the topology of the network. As the performed analysis is general it could be applied to many other important biochemical networks.
Collapse
Affiliation(s)
- Mercedes Pérez Millán
- Dto. de Matemática, FCEN, Universidad de Buenos Aires, Ciudad Universitaria, Pab. I, C1428EGA Buenos Aires, Argentina; Dto. de Ciencias Exactas, CBC, Universidad de Buenos Aires, Ramos Mejía 841, C1405CAE Buenos Aires, Argentina.
| | - Adrián G Turjanski
- Dto. de Química Biológica, FCEN, Universidad de Buenos Aires, Ciudad Universitaria, Pab. II, C1428EGA Buenos Aires, Argentina.
| |
Collapse
|
39
|
The pseudophosphatase MK-STYX induces neurite-like outgrowths in PC12 cells. PLoS One 2014; 9:e114535. [PMID: 25479605 PMCID: PMC4257672 DOI: 10.1371/journal.pone.0114535] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/10/2014] [Indexed: 01/17/2023] Open
Abstract
The rat pheochromocytoma PC12 cell line is a widely used system to study neuronal differentiation for which sustained activation of the extracellular signaling related kinase (ERK) pathway is required. Here, we investigate the function of MK-STYX [MAPK (mitogen-activated protein kinase) phosphoserine/threonine/tyrosine-binding protein] in neuronal differentiation. MK-STYX is a member of the MAPK phosphatase (MKP) family, which is generally responsible for dephosphorylating the ERKs. However, MK-STYX lacks catalytic activity due to the absence of the nucleophilic cysteine in the active site signature motif HC(X5)R that is essential for phosphatase activity. Despite being catalytically inactive, MK-STYX has been shown to play a role in important cellular pathways, including stress responses. Here we show that PC12 cells endogenously express MK-STYX. In addition, MK-STYX, but not its catalytically active mutant, induced neurite-like outgrowths in PC12 cells. Furthermore, MK-STYX dramatically increased the number of cells with neurite extensions in response to nerve growth factor (NGF), whereas the catalytically active mutant did not. MK-STYX continued to induce neurites in the presence of a MEK (MAP kinase kinase) inhibitor suggesting that MK-STYX does not act through the Ras-ERK/MAPK pathway but is involved in another pathway whose inactivation leads to neuronal differentiation. RhoA activity assays indicated that MK-STYX induced extensions through the Rho signaling pathway. MK-STYX decreased RhoA activation, whereas RhoA activation increased when MK-STYX was down-regulated. Furthermore, MK-STYX affected downstream players of RhoA such as the actin binding protein cofilin. The presence of MK-STYX decreased the phosphorylation of cofilin in non NGF stimulated cells, but increased its phosphorylation in NGF stimulated cells, whereas knocking down MK-STYX caused an opposite effect. Taken together our data suggest that MK-STYX may be a regulator of RhoA signaling, and implicate this pseudophosphatase as a regulator of neuronal differentiation.
Collapse
|
40
|
Kosmidis EK, Moschou V, Ziogas G, Boukovinas I, Albani M, Laskaris NA. Functional aspects of the EGF-induced MAP kinase cascade: a complex self-organizing system approach. PLoS One 2014; 9:e111612. [PMID: 25372488 PMCID: PMC4221048 DOI: 10.1371/journal.pone.0111612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 09/28/2014] [Indexed: 11/19/2022] Open
Abstract
The EGF-induced MAP kinase cascade is one of the most important and best characterized networks in intracellular signalling. It has a vital role in the development and maturation of living organisms. However, when deregulated, it is involved in the onset of a number of diseases. Based on a computational model describing a "surface" and an "internalized" parallel route, we use systems biology techniques to characterize aspects of the network's functional organization. We examine the re-organization of protein groups from low to high external stimulation, define functional groups of proteins within the network, determine the parameter best encoding for input intensity and predict the effect of protein removal to the system's output response. Extensive functional re-organization of proteins is observed in the lower end of stimulus concentrations. As we move to higher concentrations the variability is less pronounced. 6 functional groups have emerged from a consensus clustering approach, reflecting different dynamical aspects of the network. Mutual information investigation revealed that the maximum activation rate of the two output proteins best encodes for stimulus intensity. Removal of each protein of the network resulted in a range of graded effects, from complete silencing to intense activation. Our results provide a new "vista" of the EGF-induced MAP kinase cascade, from the perspective of complex self-organizing systems. Functional grouping of the proteins reveals an organizational scheme contrasting the current understanding of modular topology. The six identified groups may provide the means to experimentally follow the dynamics of this complex network. Also, the vulnerability analysis approach may be used for the development of novel therapeutic targets in the context of personalized medicine.
Collapse
Affiliation(s)
- Efstratios K. Kosmidis
- Laboratory of Physiology, Department of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
- * E-mail:
| | - Vasiliki Moschou
- Laboratory of Physiology, Department of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Georgios Ziogas
- AIIA Laboratory, Department of Informatics, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | | | - Maria Albani
- Laboratory of Physiology, Department of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Nikolaos A. Laskaris
- AIIA Laboratory, Department of Informatics, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| |
Collapse
|
41
|
Oyarzún DA, Bramhall JL, López-Caamal F, Richards FM, Jodrell DI, Krippendorff BF. The EGFR demonstrates linear signal transmission. Integr Biol (Camb) 2014; 6:736-42. [PMID: 24934872 DOI: 10.1039/c4ib00062e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
Cells sense information encoded in extracellular ligand concentrations and process it using intracellular signalling cascades. Using mathematical modelling and high-throughput imaging of individual cells, we studied how a transient extracellular growth factor signal is sensed by the epidermal growth factor receptor system, processed by downstream signalling, and transmitted to the nucleus. We found that transient epidermal growth factor signals are linearly translated into an activated epidermal growth factor receptor integrated over time. This allows us to generate a simplified model of receptor signaling where the receptor acts as a perfect sensor of extracellular information, while the nonlinear input-output relationship of EGF-EGFR triggered signalling is a consequence of the downstream MAPK cascade alone.
Collapse
|
42
|
"RAF" neighborhood: protein-protein interaction in the Raf/Mek/Erk pathway. FEBS Lett 2014; 588:2398-406. [PMID: 24937142 PMCID: PMC4099524 DOI: 10.1016/j.febslet.2014.06.025] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 06/05/2014] [Accepted: 06/06/2014] [Indexed: 12/19/2022]
Abstract
The Raf/Mek/Erk signaling pathway, activated downstream of Ras primarily to promote proliferation, represents the best studied of the evolutionary conserved MAPK cascades. The investigation of the pathway has continued unabated since its discovery roughly 30 years ago. In the last decade, however, the identification of unexpected in vivo functions of pathway components, as well as the discovery of Raf mutations in human cancer, the ensuing quest for inhibitors, and the efforts to understand their mechanism of action, have boosted interest tremendously. From this large body of work, protein-protein interaction has emerged as a recurrent, crucial theme. This review focuses on the role of protein complexes in the regulation of the Raf/Mek/Erk pathway and in its cross-talk with other signaling cascades. Mapping these interactions and finding a way of exploiting them for therapeutic purposes is one of the challenges of future molecule-targeted therapy.
Collapse
|
43
|
Gaykalova DA, Mambo E, Choudhary A, Houghton J, Buddavarapu K, Sanford T, Darden W, Adai A, Hadd A, Latham G, Danilova LV, Bishop J, Li RJ, Westra WH, Hennessey P, Koch WM, Ochs MF, Califano JA, Sun W. Novel insight into mutational landscape of head and neck squamous cell carcinoma. PLoS One 2014; 9:e93102. [PMID: 24667986 PMCID: PMC3965530 DOI: 10.1371/journal.pone.0093102] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 02/28/2014] [Indexed: 12/30/2022] Open
Abstract
Development of head and neck squamous cell carcinoma (HNSCC) is characterized by accumulation of mutations in several oncogenes and tumor suppressor genes. We have formerly described the mutation pattern of HNSCC and described NOTCH signaling pathway alterations. Given the complexity of the HNSCC, here we extend the previous study to understand the overall HNSCC mutation context and to discover additional genetic alterations. We performed high depth targeted exon sequencing of 51 highly actionable cancer-related genes with a high frequency of mutation across many cancer types, including head and neck. DNA from primary tumor tissues and matched normal tissues was analyzed for 37 HNSCC patients. We identified 26 non-synonymous or stop-gained mutations targeting 11 of 51 selected genes. These genes were mutated in 17 out of 37 (46%) studied HNSCC patients. Smokers harbored 3.2-fold more mutations than non-smokers. Importantly, TP53 was mutated in 30%, NOTCH1 in 8% and FGFR3 in 5% of HNSCC. HPV negative patients harbored 4-fold more TP53 mutations than HPV positive patients. These data confirm prior reports of the HNSCC mutational profile. Additionally, we detected mutations in two new genes, CEBPA and FES, which have not been previously reported in HNSCC. These data extend the spectrum of HNSCC mutations and define novel mutation targets in HNSCC carcinogenesis, especially for smokers and HNSCC without HPV infection.
Collapse
Affiliation(s)
- Daria A. Gaykalova
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | | | | | | | | | | | - Will Darden
- Asuragen Inc., Austin, Texas, United States of America
| | - Alex Adai
- Asuragen Inc., Austin, Texas, United States of America
| | - Andrew Hadd
- Asuragen Inc., Austin, Texas, United States of America
| | - Gary Latham
- Asuragen Inc., Austin, Texas, United States of America
| | - Ludmila V. Danilova
- Department of Oncology and Health Science Informatics, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Justin Bishop
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Ryan J. Li
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - William H. Westra
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Patrick Hennessey
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Wayne M. Koch
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Michael F. Ochs
- Department of Oncology and Health Science Informatics, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
| | - Joseph A. Califano
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- Milton J. Dance Head and Neck Center, Greater Baltimore Medical Center, Baltimore, Maryland, United States of America
- * E-mail: (WS); (JAC)
| | - Wenyue Sun
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States of America
- * E-mail: (WS); (JAC)
| |
Collapse
|
44
|
Hagen DR, White JK, Tidor B. Convergence in parameters and predictions using computational experimental design. Interface Focus 2014; 3:20130008. [PMID: 24511374 PMCID: PMC3915829 DOI: 10.1098/rsfs.2013.0008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Typically, biological models fitted to experimental data suffer from significant parameter uncertainty, which can lead to inaccurate or uncertain predictions. One school of thought holds that accurate estimation of the true parameters of a biological system is inherently problematic. Recent work, however, suggests that optimal experimental design techniques can select sets of experiments whose members probe complementary aspects of a biochemical network that together can account for its full behaviour. Here, we implemented an experimental design approach for selecting sets of experiments that constrain parameter uncertainty. We demonstrated with a model of the epidermal growth factor–nerve growth factor pathway that, after synthetically performing a handful of optimal experiments, the uncertainty in all 48 parameters converged below 10 per cent. Furthermore, the fitted parameters converged to their true values with a small error consistent with the residual uncertainty. When untested experimental conditions were simulated with the fitted models, the predicted species concentrations converged to their true values with errors that were consistent with the residual uncertainty. This paper suggests that accurate parameter estimation is achievable with complementary experiments specifically designed for the task, and that the resulting parametrized models are capable of accurate predictions.
Collapse
Affiliation(s)
- David R Hagen
- Department of Biological Engineering , Massachusetts Institute of Technology , Cambridge, MA 02139 , USA ; Computer Science and Artificial Intelligence Laboratory , Massachusetts Institute of Technology , Cambridge, MA 02139 , USA
| | - Jacob K White
- Department of Electrical Engineering and Computer Science , Massachusetts Institute of Technology , Cambridge, MA 02139 , USA
| | - Bruce Tidor
- Department of Biological Engineering , Massachusetts Institute of Technology , Cambridge, MA 02139 , USA ; Computer Science and Artificial Intelligence Laboratory , Massachusetts Institute of Technology , Cambridge, MA 02139 , USA ; Department of Electrical Engineering and Computer Science , Massachusetts Institute of Technology , Cambridge, MA 02139 , USA
| |
Collapse
|
45
|
Mobashir M, Madhusudhan T, Isermann B, Beyer T, Schraven B. Negative interactions and feedback regulations are required for transient cellular response. Sci Rep 2014; 4:3718. [PMID: 24430195 PMCID: PMC3893651 DOI: 10.1038/srep03718] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022] Open
Abstract
Signal transduction is a process required to conduct information from a receptor to the nucleus. This process is vital for the control of cellular function and fate. The dynamics of signaling activation and inhibition determine processes such as apoptosis, proliferation, and differentiation. Thus, it is important to understand the factors modulating transient and sustained response. To address this question, by applying mathematical approach we have studied the factors which can alter the activation nature of downstream signaling molecules. The factors which we have investigated are loops (feed forward and feedback loops), cross-talk of signal transduction pathways, and the change in the concentration of the signaling molecules. Based on our results we conclude that among these factors feedback loop and the cross-talks which directly inhibit the target protein dominantly controls the transient cellular response.
Collapse
Affiliation(s)
- Mohammad Mobashir
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Thati Madhusudhan
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Tilo Beyer
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Burkhart Schraven
- 1] Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, 39120, Magdeburg, Germany [2] Department of Immune Control, Helmholtz Centre for Infectious Disease (HZI), Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
46
|
Marín-Hernández A, López-Ramírez SY, Gallardo-Pérez JC, Rodríguez-Enríquez S, Moreno-Sánchez R, Saavedra E. Systems Biology Approaches to Cancer Energy Metabolism. SYSTEMS BIOLOGY OF METABOLIC AND SIGNALING NETWORKS 2014. [DOI: 10.1007/978-3-642-38505-6_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
47
|
Koch Y, Wolf T, Sorger PK, Eils R, Brors B. Decision-tree based model analysis for efficient identification of parameter relations leading to different signaling states. PLoS One 2013; 8:e82593. [PMID: 24367526 PMCID: PMC3867358 DOI: 10.1371/journal.pone.0082593] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 11/03/2013] [Indexed: 11/18/2022] Open
Abstract
In systems biology, a mathematical description of signal transduction processes is used to gain a more detailed mechanistic understanding of cellular signaling networks. Such models typically depend on a number of parameters that have different influence on the model behavior. Local sensitivity analysis is able to identify parameters that have the largest effect on signaling strength. Bifurcation analysis shows on which parameters a qualitative model response depends. Most methods for model analysis are intrinsically univariate. They typically cannot consider combinations of parameters since the search space for such analysis would be too large. This limitation is important since activation of a signaling pathway often relies on multiple rather than on single factors. Here, we present a novel method for model analysis that overcomes this limitation. As input to a model defined by a system of ordinary differential equations, we consider parameters for initial chemical species concentrations. The model is used to simulate the system response, which is then classified into pre-defined classes (e.g., active or not active). This is combined with a scan of the parameter space. Parameter sets leading to a certain system response are subjected to a decision tree algorithm, which learns conditions that lead to this response. We compare our method to two alternative multivariate approaches to model analysis: analytical solution for steady states combined with a parameter scan, and direct Lyapunov exponent (DLE) analysis. We use three previously published models including a model for EGF receptor internalization and two apoptosis models to demonstrate the power of our approach. Our method reproduces critical parameter relations previously obtained by both steady-state and DLE analysis while being more generally applicable and substantially less computationally expensive. The method can be used as a general tool to predict multivariate control strategies for pathway activation and to suggest strategies for drug intervention.
Collapse
Affiliation(s)
- Yvonne Koch
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg, Germany
| | - Thomas Wolf
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg, Germany
- Institute of General Pathology, Heidelberg University Medical School, University of Heidelberg, Im Neuenheimer Feld 224, Heidelberg, Germany
| | - Peter K. Sorger
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, Massachusetts, United States of America
| | - Roland Eils
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg, Germany
- Department for Bioinformatics and Functional Genomics, Institute for Pharmacy and Molecular Biotechnology, and Bioquant Center, University of Heidelberg, Im Neuenheimer Feld 267, Heidelberg, Germany
| | - Benedikt Brors
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg, Germany
| |
Collapse
|
48
|
Josset L, Tisoncik-Go J, Katze MG. Moving H5N1 studies into the era of systems biology. Virus Res 2013; 178:151-67. [PMID: 23499671 PMCID: PMC3834220 DOI: 10.1016/j.virusres.2013.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 02/24/2013] [Indexed: 12/20/2022]
Abstract
The dynamics of H5N1 influenza virus pathogenesis are multifaceted and can be seen as an emergent property that cannot be comprehended without looking at the system as a whole. In past years, most of the high-throughput studies on H5N1-host interactions have focused on the host transcriptomic response, at the cellular or the lung tissue level. These studies pointed out that the dynamics and magnitude of the innate immune response and immune cell infiltration is critical to H5N1 pathogenesis. However, viral-host interactions are multidimensional and advances in technologies are creating new possibilities to systematically measure additional levels of 'omic data (e.g. proteomic, metabolomic, and RNA profiling) at each temporal and spatial scale (from the single cell to the organism) of the host response. Natural host genetic variation represents another dimension of the host response that determines pathogenesis. Systems biology models of H5N1 disease aim at understanding and predicting pathogenesis through integration of these different dimensions by using intensive computational modeling. In this review, we describe the importance of 'omic studies for providing a more comprehensive view of infection and mathematical models that are being developed to integrate these data. This review provides a roadmap for what needs to be done in the future and what computational strategies should be used to build a global model of H5N1 pathogenesis. It is time for systems biology of H5N1 pathogenesis to take center stage as the field moves toward a more comprehensive view of virus-host interactions.
Collapse
Affiliation(s)
- Laurence Josset
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA 98195, United States
| | | | | |
Collapse
|
49
|
van Heeswijk WC, Westerhoff HV, Boogerd FC. Nitrogen assimilation in Escherichia coli: putting molecular data into a systems perspective. Microbiol Mol Biol Rev 2013; 77:628-95. [PMID: 24296575 PMCID: PMC3973380 DOI: 10.1128/mmbr.00025-13] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
We present a comprehensive overview of the hierarchical network of intracellular processes revolving around central nitrogen metabolism in Escherichia coli. The hierarchy intertwines transport, metabolism, signaling leading to posttranslational modification, and transcription. The protein components of the network include an ammonium transporter (AmtB), a glutamine transporter (GlnHPQ), two ammonium assimilation pathways (glutamine synthetase [GS]-glutamate synthase [glutamine 2-oxoglutarate amidotransferase {GOGAT}] and glutamate dehydrogenase [GDH]), the two bifunctional enzymes adenylyl transferase/adenylyl-removing enzyme (ATase) and uridylyl transferase/uridylyl-removing enzyme (UTase), the two trimeric signal transduction proteins (GlnB and GlnK), the two-component regulatory system composed of the histidine protein kinase nitrogen regulator II (NRII) and the response nitrogen regulator I (NRI), three global transcriptional regulators called nitrogen assimilation control (Nac) protein, leucine-responsive regulatory protein (Lrp), and cyclic AMP (cAMP) receptor protein (Crp), the glutaminases, and the nitrogen-phosphotransferase system. First, the structural and molecular knowledge on these proteins is reviewed. Thereafter, the activities of the components as they engage together in transport, metabolism, signal transduction, and transcription and their regulation are discussed. Next, old and new molecular data and physiological data are put into a common perspective on integral cellular functioning, especially with the aim of resolving counterintuitive or paradoxical processes featured in nitrogen assimilation. Finally, we articulate what still remains to be discovered and what general lessons can be learned from the vast amounts of data that are available now.
Collapse
|
50
|
Kariya Y, Honma M, Suzuki H. Systems-based understanding of pharmacological responses with combinations of multidisciplinary methodologies. Biopharm Drug Dispos 2013; 34:489-507. [DOI: 10.1002/bdd.1865] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 10/06/2013] [Indexed: 12/25/2022]
Affiliation(s)
- Yoshiaki Kariya
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine; The University of Tokyo; 113-8655 Tokyo Japan
| | - Masashi Honma
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine; The University of Tokyo; 113-8655 Tokyo Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine; The University of Tokyo; 113-8655 Tokyo Japan
| |
Collapse
|