1
|
Viswan NA, Tribut A, Gasparyan M, Radulescu O, Bhalla US. Mathematical basis and toolchain for hierarchical optimization of biochemical networks. PLoS Comput Biol 2024; 20:e1012624. [PMID: 39621764 PMCID: PMC11637339 DOI: 10.1371/journal.pcbi.1012624] [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: 08/06/2024] [Revised: 12/12/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
Biological signalling systems are complex, and efforts to build mechanistic models must confront a huge parameter space, indirect and sparse data, and frequently encounter multiscale and multiphysics phenomena. We present HOSS, a framework for Hierarchical Optimization of Systems Simulations, to address such problems. HOSS operates by breaking down extensive systems models into individual pathway blocks organized in a nested hierarchy. At the first level, dependencies are solely on signalling inputs, and subsequent levels rely only on the preceding ones. We demonstrate that each independent pathway in every level can be efficiently optimized. Once optimized, its parameters are held constant while the pathway serves as input for succeeding levels. We develop an algorithmic approach to identify the necessary nested hierarchies for the application of HOSS in any given biochemical network. Furthermore, we devise two parallelizable variants that generate numerous model instances using stochastic scrambling of parameters during initial and intermediate stages of optimization. Our results indicate that these variants produce superior models and offer an estimate of solution degeneracy. Additionally, we showcase the effectiveness of the optimization methods for both abstracted, event-based simulations and ODE-based models.
Collapse
Affiliation(s)
- Nisha Ann Viswan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
- The University of Trans-Disciplinary Health Sciences and Technology, Bangalore, India
| | - Alexandre Tribut
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS and INSERM, Montpellier, France
- Ecole Centrale de Nantes, Nantes, France
| | - Manvel Gasparyan
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS and INSERM, Montpellier, France
| | - Ovidiu Radulescu
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS and INSERM, Montpellier, France
| | - Upinder S. Bhalla
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| |
Collapse
|
2
|
Lakrisenko P, Pathirana D, Weindl D, Hasenauer J. Benchmarking methods for computing local sensitivities in ordinary differential equation models at dynamic and steady states. PLoS One 2024; 19:e0312148. [PMID: 39441813 PMCID: PMC11498742 DOI: 10.1371/journal.pone.0312148] [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: 06/11/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Estimating parameters of dynamic models from experimental data is a challenging, and often computationally-demanding task. It requires a large number of model simulations and objective function gradient computations, if gradient-based optimization is used. In many cases, steady-state computation is a part of model simulation, either due to steady-state data or an assumption that the system is at steady state at the initial time point. Various methods are available for steady-state and gradient computation. Yet, the most efficient pair of methods (one for steady states, one for gradients) for a particular model is often not clear. In order to facilitate the selection of methods, we explore six method pairs for computing the steady state and sensitivities at steady state using six real-world problems. The method pairs involve numerical integration or Newton's method to compute the steady-state, and-for both forward and adjoint sensitivity analysis-numerical integration or a tailored method to compute the sensitivities at steady-state. Our evaluation shows that all method pairs provide accurate steady-state and gradient values, and that the two method pairs that combine numerical integration for the steady-state with a tailored method for the sensitivities at steady-state were the most robust, and amongst the most computationally-efficient. We also observed that while Newton's method for steady-state computation yields a substantial speedup compared to numerical integration, it may lead to a large number of simulation failures. Overall, our study provides a concise overview across current methods for computing sensitivities at steady state. While our study shows that there is no universally-best method pair, it also provides guidance to modelers in choosing the right methods for a problem at hand.
Collapse
Affiliation(s)
- Polina Lakrisenko
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- School of Life Sciences, Technische Universität München, Freising, Germany
| | - Dilan Pathirana
- Faculty of Mathematics and Natural Sciences, and the Life and Medical Sciences Institute (LIMES), Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Daniel Weindl
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Faculty of Mathematics and Natural Sciences, and the Life and Medical Sciences Institute (LIMES), Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Jan Hasenauer
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Faculty of Mathematics and Natural Sciences, and the Life and Medical Sciences Institute (LIMES), Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
3
|
Armistead J, Höpfl S, Goldhausen P, Müller-Hartmann A, Fahle E, Hatzold J, Franzen R, Brodesser S, Radde NE, Hammerschmidt M. A sphingolipid rheostat controls apoptosis versus apical cell extrusion as alternative tumour-suppressive mechanisms. Cell Death Dis 2024; 15:746. [PMID: 39397024 PMCID: PMC11471799 DOI: 10.1038/s41419-024-07134-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024]
Abstract
Evasion of cell death is a hallmark of cancer, and consequently the induction of cell death is a common strategy in cancer treatment. However, the molecular mechanisms regulating different types of cell death are poorly understood. We have formerly shown that in the epidermis of hypomorphic zebrafish hai1a mutant embryos, pre-neoplastic transformations of keratinocytes caused by unrestrained activity of the type II transmembrane serine protease Matriptase-1 heal spontaneously. This healing is driven by Matriptase-dependent increased sphingosine kinase (SphK) activity and sphingosine-1-phosphate (S1P)-mediated keratinocyte loss via apical cell extrusion. In contrast, amorphic hai1afr26 mutants with even higher Matriptase-1 and SphK activity die within a few days. Here we show that this lethality is not due to epidermal carcinogenesis, but to aberrant tp53-independent apoptosis of keratinocytes caused by increased levels of pro-apoptotic C16 ceramides, sphingolipid counterparts to S1P within the sphingolipid rheostat, which severely compromises the epidermal barrier. Mathematical modelling of sphingolipid rheostat homeostasis, combined with in vivo manipulations of components of the rheostat or the ceramide de novo synthesis pathway, indicate that this unexpected overproduction of ceramides is caused by a negative feedback loop sensing ceramide levels and controlling ceramide replenishment via de novo synthesis. Therefore, despite their initial decrease due to increased conversion to S1P, ceramides eventually reach cell death-inducing levels, making transformed pre-neoplastic keratinocytes die even before they are extruded, thereby abrogating the normally barrier-preserving mode of apical live cell extrusion. Our results offer an in vivo perspective of the dynamics of sphingolipid homeostasis and its relevance for epithelial cell survival versus cell death, linking apical cell extrusion and apoptosis. Implications for human carcinomas and their treatments are discussed.
Collapse
Affiliation(s)
- Joy Armistead
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| | - Sebastian Höpfl
- Institute for Stochastics and Applications, University of Stuttgart, Stuttgart, Germany
| | - Pierre Goldhausen
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany
| | | | - Evelin Fahle
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany
| | - Julia Hatzold
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany
| | - Rainer Franzen
- Max-Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Susanne Brodesser
- Lipidomics/Metabolomics Facility, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Nicole E Radde
- Institute for Stochastics and Applications, University of Stuttgart, Stuttgart, Germany
| | - Matthias Hammerschmidt
- Institute of Zoology / Developmental Biology, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
4
|
Schmiester L, Brasó-Maristany F, González-Farré B, Pascual T, Gavilá J, Tekpli X, Geisler J, Kristensen VN, Frigessi A, Prat A, Köhn-Luque A. Computational Model Predicts Patient Outcomes in Luminal B Breast Cancer Treated with Endocrine Therapy and CDK4/6 Inhibition. Clin Cancer Res 2024; 30:3779-3787. [PMID: 38922642 DOI: 10.1158/1078-0432.ccr-24-0244] [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: 01/28/2024] [Revised: 04/12/2024] [Accepted: 06/24/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE Development of a computational biomarker to predict, prior to treatment, the response to CDK4/6 inhibition (CDK4/6i) in combination with endocrine therapy in patients with breast cancer. EXPERIMENTAL DESIGN A mechanistic mathematical model that accounts for protein signaling and drug mechanisms of action was developed and trained on extensive, publicly available data from breast cancer cell lines. The model was built to provide a patient-specific response score based on the expression of six genes (CCND1, CCNE1, ESR1, RB1, MYC, and CDKN1A). The model was validated in five independent cohorts of 148 patients in total with early-stage or advanced breast cancer treated with endocrine therapy and CDK4/6i. Response was measured either by evaluating Ki67 levels and PAM50 risk of relapse (ROR) after neoadjuvant treatment or by evaluating progression-free survival (PFS). RESULTS The model showed significant association with patient's outcomes in all five cohorts. The model predicted high Ki67 [area under the curve; AUC (95% confidence interval, CI) of 0.80 (0.64-0.92), 0.81 (0.60-1.00) and 0.80 (0.65-0.93)] and high PAM50 ROR [AUC of 0.78 (0.64-0.89)]. This observation was not obtained in patients treated with chemotherapy. In the other cohorts, patient stratification based on the model prediction was significantly associated with PFS [hazard ratio (HR) = 2.92 (95% CI, 1.08-7.86), P = 0.034 and HR = 2.16 (1.02 4.55), P = 0.043]. CONCLUSIONS A mathematical modeling approach accurately predicts patient outcome following CDK4/6i plus endocrine therapy that marks a step toward more personalized treatments in patients with Luminal B breast cancer.
Collapse
Affiliation(s)
- Leonard Schmiester
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Blanca González-Farré
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Tomás Pascual
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Joaquín Gavilá
- SOLTI Cancer Research Group, Barcelona, Spain
- Department of Medical Oncology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Xavier Tekpli
- Department of Medical Genetics, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Jürgen Geisler
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Akershus University Hospital, Oslo, Norway
| | - Vessela N Kristensen
- Department of Medical Genetics, Oslo University Hospital, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Arnoldo Frigessi
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Alvaro Köhn-Luque
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
5
|
Duez Q, van de Wiel J, van Sluijs B, Ghosh S, Baltussen MG, Derks MTGM, Roithová J, Huck WTS. Quantitative Online Monitoring of an Immobilized Enzymatic Network by Ion Mobility-Mass Spectrometry. J Am Chem Soc 2024; 146:20778-20787. [PMID: 39013149 PMCID: PMC11295183 DOI: 10.1021/jacs.4c04218] [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: 03/26/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024]
Abstract
The forward design of in vitro enzymatic reaction networks (ERNs) requires a detailed analysis of network kinetics and potentially hidden interactions between the substrates and enzymes. Although flow chemistry allows for a systematic exploration of how the networks adapt to continuously changing conditions, the analysis of the reaction products is often a bottleneck. Here, we report on the interface between a continuous stirred-tank reactor, in which an immobilized enzymatic network made of 12 enzymes is compartmentalized, and an ion mobility-mass spectrometer. Feeding uniformly 13C-labeled inputs to the enzymatic network generates all isotopically labeled reaction intermediates and products, which are individually detected by ion mobility-mass spectrometry (IMS-MS) based on their mass-to-charge ratios and inverse ion mobilities. The metabolic flux can be continuously and quantitatively monitored by diluting the ERN output with nonlabeled standards of known concentrations. The real-time quantitative data obtained by IMS-MS are then harnessed to train a model of network kinetics, which proves sufficiently predictive to control the ERN output after a single optimally designed experiment. The high resolution of the time-course data provided by this approach is an important stepping stone to design and control sizable and intricate ERNs.
Collapse
Affiliation(s)
| | | | - Bob van Sluijs
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 AJ, The Netherlands
| | - Souvik Ghosh
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 AJ, The Netherlands
| | - Mathieu G. Baltussen
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 AJ, The Netherlands
| | - Max T. G. M. Derks
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 AJ, The Netherlands
| | - Jana Roithová
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 AJ, The Netherlands
| | - Wilhelm T. S. Huck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 AJ, The Netherlands
| |
Collapse
|
6
|
Dorešić D, Grein S, Hasenauer J. Efficient parameter estimation for ODE models of cellular processes using semi-quantitative data. Bioinformatics 2024; 40:i558-i566. [PMID: 38940161 PMCID: PMC11211815 DOI: 10.1093/bioinformatics/btae210] [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] [Indexed: 06/29/2024] Open
Abstract
MOTIVATION Quantitative dynamical models facilitate the understanding of biological processes and the prediction of their dynamics. The parameters of these models are commonly estimated from experimental data. Yet, experimental data generated from different techniques do not provide direct information about the state of the system but a nonlinear (monotonic) transformation of it. For such semi-quantitative data, when this transformation is unknown, it is not apparent how the model simulations and the experimental data can be compared. RESULTS We propose a versatile spline-based approach for the integration of a broad spectrum of semi-quantitative data into parameter estimation. We derive analytical formulas for the gradients of the hierarchical objective function and show that this substantially increases the estimation efficiency. Subsequently, we demonstrate that the method allows for the reliable discovery of unknown measurement transformations. Furthermore, we show that this approach can significantly improve the parameter inference based on semi-quantitative data in comparison to available methods. AVAILABILITY AND IMPLEMENTATION Modelers can easily apply our method by using our implementation in the open-source Python Parameter EStimation TOolbox (pyPESTO) available at https://github.com/ICB-DCM/pyPESTO.
Collapse
Affiliation(s)
- Domagoj Dorešić
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Stephan Grein
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
| | - Jan Hasenauer
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Center for Mathematics, Technische Universität München, 85748 Garching, Germany
| |
Collapse
|
7
|
Kiss AE, Venkatasubramani AV, Pathirana D, Krause S, Sparr A, Hasenauer J, Imhof A, Müller M, Becker P. Processivity and specificity of histone acetylation by the male-specific lethal complex. Nucleic Acids Res 2024; 52:4889-4905. [PMID: 38407474 PMCID: PMC11109948 DOI: 10.1093/nar/gkae123] [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: 12/04/2023] [Revised: 01/29/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024] Open
Abstract
Acetylation of lysine 16 of histone H4 (H4K16ac) stands out among the histone modifications, because it decompacts the chromatin fiber. The metazoan acetyltransferase MOF (KAT8) regulates transcription through H4K16 acetylation. Antibody-based studies had yielded inconclusive results about the selectivity of MOF to acetylate the H4 N-terminus. We used targeted mass spectrometry to examine the activity of MOF in the male-specific lethal core (4-MSL) complex on nucleosome array substrates. This complex is part of the Dosage Compensation Complex (DCC) that activates X-chromosomal genes in male Drosophila. During short reaction times, MOF acetylated H4K16 efficiently and with excellent selectivity. Upon longer incubation, the enzyme progressively acetylated lysines 12, 8 and 5, leading to a mixture of oligo-acetylated H4. Mathematical modeling suggests that MOF recognizes and acetylates H4K16 with high selectivity, but remains substrate-bound and continues to acetylate more N-terminal H4 lysines in a processive manner. The 4-MSL complex lacks non-coding roX RNA, a critical component of the DCC. Remarkably, addition of RNA to the reaction non-specifically suppressed H4 oligo-acetylation in favor of specific H4K16 acetylation. Because RNA destabilizes the MSL-nucleosome interaction in vitro we speculate that RNA accelerates enzyme-substrate turn-over in vivo, thus limiting the processivity of MOF, thereby increasing specific H4K16 acetylation.
Collapse
Affiliation(s)
- Anna E Kiss
- Biomedical Center, Molecular Biology Division, Ludwig-Maximilians-University of Munich, Planegg-Martinsried, Germany
| | - Anuroop V Venkatasubramani
- Biomedical Center, Molecular Biology Division, Ludwig-Maximilians-University of Munich, Planegg-Martinsried, Germany
| | - Dilan Pathirana
- Life and Medical Sciences (LIMES) Institute, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Silke Krause
- Biomedical Center, Molecular Biology Division, Ludwig-Maximilians-University of Munich, Planegg-Martinsried, Germany
| | - Aline Campos Sparr
- Biomedical Center, Molecular Biology Division, Ludwig-Maximilians-University of Munich, Planegg-Martinsried, Germany
| | - Jan Hasenauer
- Life and Medical Sciences (LIMES) Institute, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Axel Imhof
- Biomedical Center, Molecular Biology Division, Ludwig-Maximilians-University of Munich, Planegg-Martinsried, Germany
| | - Marisa Müller
- Biomedical Center, Molecular Biology Division, Ludwig-Maximilians-University of Munich, Planegg-Martinsried, Germany
| | - Peter B Becker
- Biomedical Center, Molecular Biology Division, Ludwig-Maximilians-University of Munich, Planegg-Martinsried, Germany
| |
Collapse
|
8
|
Höpfl S, Albadry M, Dahmen U, Herrmann KH, Kindler EM, König M, Reichenbach JR, Tautenhahn HM, Wei W, Zhao WT, Radde NE. Bayesian modelling of time series data (BayModTS)-a FAIR workflow to process sparse and highly variable data. Bioinformatics 2024; 40:btae312. [PMID: 38741151 PMCID: PMC11128094 DOI: 10.1093/bioinformatics/btae312] [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: 01/29/2024] [Revised: 04/11/2024] [Accepted: 05/13/2024] [Indexed: 05/16/2024] Open
Abstract
MOTIVATION Systems biology aims to better understand living systems through mathematical modelling of experimental and clinical data. A pervasive challenge in quantitative dynamical modelling is the integration of time series measurements, which often have high variability and low sampling resolution. Approaches are required to utilize such information while consistently handling uncertainties. RESULTS We present BayModTS (Bayesian modelling of time series data), a new FAIR (findable, accessible, interoperable, and reusable) workflow for processing and analysing sparse and highly variable time series data. BayModTS consistently transfers uncertainties from data to model predictions, including process knowledge via parameterized models. Further, credible differences in the dynamics of different conditions can be identified by filtering noise. To demonstrate the power and versatility of BayModTS, we applied it to three hepatic datasets gathered from three different species and with different measurement techniques: (i) blood perfusion measurements by magnetic resonance imaging in rat livers after portal vein ligation, (ii) pharmacokinetic time series of different drugs in normal and steatotic mice, and (iii) CT-based volumetric assessment of human liver remnants after clinical liver resection. AVAILABILITY AND IMPLEMENTATION The BayModTS codebase is available on GitHub at https://github.com/Systems-Theory-in-Systems-Biology/BayModTS. The repository contains a Python script for the executable BayModTS workflow and a widely applicable SBML (systems biology markup language) model for retarded transient functions. In addition, all examples from the paper are included in the repository. Data and code of the application examples are stored on DaRUS: https://doi.org/10.18419/darus-3876. The raw MRI ROI voxel data were uploaded to DaRUS: https://doi.org/10.18419/darus-3878. The steatosis metabolite data are published on FairdomHub: 10.15490/fairdomhub.1.study.1070.1.
Collapse
Affiliation(s)
- Sebastian Höpfl
- Institute for Stochastics and Applications, University of Stuttgart, 70569 Stuttgart, Germany
| | - Mohamed Albadry
- Experimental Transplantation Surgery, Department of General, Vascular and Visceral Surgery, University Hospital Jena, 07745 Jena, Germany
- Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkom, Menoufia, Egypt
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Vascular and Visceral Surgery, University Hospital Jena, 07745 Jena, Germany
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, University Hospital Jena, 07743 Jena, Germany
| | - Eva Marie Kindler
- Clinic for General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany
| | - Matthias König
- Institute for Biology, Faculty of Life Sciences, Humboldt-University Berlin, 10115 Berlin, Germany
| | - Jürgen Rainer Reichenbach
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, University Hospital Jena, 07743 Jena, Germany
| | - Hans-Michael Tautenhahn
- Clinic for Visceral, Transplantation, Thoracic and Vascular Surgery, Leipzig University Hospital, 04103 Leipzig, Germany
| | - Weiwei Wei
- Experimental Transplantation Surgery, Department of General, Vascular and Visceral Surgery, University Hospital Jena, 07745 Jena, Germany
| | - Wan-Ting Zhao
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, University Hospital Jena, 07743 Jena, Germany
| | - Nicole Erika Radde
- Institute for Stochastics and Applications, University of Stuttgart, 70569 Stuttgart, Germany
| |
Collapse
|
9
|
Merkt S, Ali S, Gudina EK, Adissu W, Gize A, Muenchhoff M, Graf A, Krebs S, Elsbernd K, Kisch R, Betizazu SS, Fantahun B, Bekele D, Rubio-Acero R, Gashaw M, Girma E, Yilma D, Zeynudin A, Paunovic I, Hoelscher M, Blum H, Hasenauer J, Kroidl A, Wieser A. Long-term monitoring of SARS-CoV-2 seroprevalence and variants in Ethiopia provides prediction for immunity and cross-immunity. Nat Commun 2024; 15:3463. [PMID: 38658564 PMCID: PMC11043357 DOI: 10.1038/s41467-024-47556-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 04/03/2024] [Indexed: 04/26/2024] Open
Abstract
Under-reporting of COVID-19 and the limited information about circulating SARS-CoV-2 variants remain major challenges for many African countries. We analyzed SARS-CoV-2 infection dynamics in Addis Ababa and Jimma, Ethiopia, focusing on reinfection, immunity, and vaccination effects. We conducted an antibody serology study spanning August 2020 to July 2022 with five rounds of data collection across a population of 4723, sequenced PCR-test positive samples, used available test positivity rates, and constructed two mathematical models integrating this data. A multivariant model explores variant dynamics identifying wildtype, alpha, delta, and omicron BA.4/5 as key variants in the study population, and cross-immunity between variants, revealing risk reductions between 24% and 69%. An antibody-level model predicts slow decay leading to sustained high antibody levels. Retrospectively, increased early vaccination might have substantially reduced infections during the delta and omicron waves in the considered group of individuals, though further vaccination now seems less impactful.
Collapse
Affiliation(s)
- Simon Merkt
- Life and Medical Sciences (LIMES), University of Bonn, Bonn, Germany
| | - Solomon Ali
- Saint Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Esayas Kebede Gudina
- Jimma University Clinical Trial Unit, Jimma University Institute of Health, Jimma, Ethiopia
| | - Wondimagegn Adissu
- Jimma University Clinical Trial Unit, Jimma University Institute of Health, Jimma, Ethiopia
| | - Addisu Gize
- Saint Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
- CIH LMU Center for International Health, LMU Munich, Munich, Germany
| | - Maximilian Muenchhoff
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Alexander Graf
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich, Germany
| | - Kira Elsbernd
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Rebecca Kisch
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Bereket Fantahun
- Saint Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Delayehu Bekele
- Saint Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Raquel Rubio-Acero
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mulatu Gashaw
- Jimma University Clinical Trial Unit, Jimma University Institute of Health, Jimma, Ethiopia
| | - Eyob Girma
- Jimma University Clinical Trial Unit, Jimma University Institute of Health, Jimma, Ethiopia
| | - Daniel Yilma
- Jimma University Clinical Trial Unit, Jimma University Institute of Health, Jimma, Ethiopia
| | - Ahmed Zeynudin
- Jimma University Clinical Trial Unit, Jimma University Institute of Health, Jimma, Ethiopia
| | - Ivana Paunovic
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Immunology, Infection and Pandemic Research IIP, Fraunhofer ITMP, Munich, Germany
| | - Michael Hoelscher
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Immunology, Infection and Pandemic Research IIP, Fraunhofer ITMP, Munich, Germany
- Unit Global Health, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich, Germany
| | - Jan Hasenauer
- Life and Medical Sciences (LIMES), University of Bonn, Bonn, Germany.
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany.
- Center for Mathematics, Technische Universität München, Garching, Germany.
| | - Arne Kroidl
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany.
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Andreas Wieser
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany.
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
- Immunology, Infection and Pandemic Research IIP, Fraunhofer ITMP, Munich, Germany.
- Faculty of Medicine, Max Von Pettenkofer Institute, LMU Munich, Munich, Germany.
| |
Collapse
|
10
|
Burbano de Lara S, Kemmer S, Biermayer I, Feiler S, Vlasov A, D'Alessandro LA, Helm B, Mölders C, Dieter Y, Ghallab A, Hengstler JG, Körner C, Matz-Soja M, Götz C, Damm G, Hoffmann K, Seehofer D, Berg T, Schilling M, Timmer J, Klingmüller U. Basal MET phosphorylation is an indicator of hepatocyte dysregulation in liver disease. Mol Syst Biol 2024; 20:187-216. [PMID: 38216754 PMCID: PMC10912216 DOI: 10.1038/s44320-023-00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 01/14/2024] Open
Abstract
Chronic liver diseases are worldwide on the rise. Due to the rapidly increasing incidence, in particular in Western countries, metabolic dysfunction-associated steatotic liver disease (MASLD) is gaining importance as the disease can develop into hepatocellular carcinoma. Lipid accumulation in hepatocytes has been identified as the characteristic structural change in MASLD development, but molecular mechanisms responsible for disease progression remained unresolved. Here, we uncover in primary hepatocytes from a preclinical model fed with a Western diet (WD) an increased basal MET phosphorylation and a strong downregulation of the PI3K-AKT pathway. Dynamic pathway modeling of hepatocyte growth factor (HGF) signal transduction combined with global proteomics identifies that an elevated basal MET phosphorylation rate is the main driver of altered signaling leading to increased proliferation of WD-hepatocytes. Model-adaptation to patient-derived hepatocytes reveal patient-specific variability in basal MET phosphorylation, which correlates with patient outcome after liver surgery. Thus, dysregulated basal MET phosphorylation could be an indicator for the health status of the liver and thereby inform on the risk of a patient to suffer from liver failure after surgery.
Collapse
Affiliation(s)
- Sebastian Burbano de Lara
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
| | - Svenja Kemmer
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
- Institute of Physics, University of Freiburg, Freiburg, Germany
- FDM - Freiburg Center for Data Analysis and Modeling, University of Freiburg, Freiburg, Germany
| | - Ina Biermayer
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
| | - Svenja Feiler
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of General, Visceral and Transplant Surgery, Heidelberg University, Heidelberg, Germany
| | - Artyom Vlasov
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lorenza A D'Alessandro
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Helm
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christina Mölders
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
| | - Yannik Dieter
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ahmed Ghallab
- Systems Toxicology, Leibniz Research Center for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Jan G Hengstler
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
- Systems Toxicology, Leibniz Research Center for Working Environment and Human Factors, Technical University Dortmund, Dortmund, Germany
| | - Christiane Körner
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
- Division of Hepatology, Clinic of Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Madlen Matz-Soja
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
- Division of Hepatology, Clinic of Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Christina Götz
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital Leipzig, Leipzig University, 04103, Leipzig, Germany
| | - Georg Damm
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital Leipzig, Leipzig University, 04103, Leipzig, Germany
| | - Katrin Hoffmann
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
- Department of General, Visceral and Transplant Surgery, Heidelberg University, Heidelberg, Germany
| | - Daniel Seehofer
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital Leipzig, Leipzig University, 04103, Leipzig, Germany
| | - Thomas Berg
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany
- Division of Hepatology, Clinic of Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Marcel Schilling
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jens Timmer
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany.
- Institute of Physics, University of Freiburg, Freiburg, Germany.
- FDM - Freiburg Center for Data Analysis and Modeling, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| | - Ursula Klingmüller
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Liver Systems Medicine against Cancer (LiSyM-Krebs), Heidelberg, Germany.
| |
Collapse
|
11
|
van Sluijs B, Zhou T, Helwig B, Baltussen MG, Nelissen FHT, Heus HA, Huck WTS. Iterative design of training data to control intricate enzymatic reaction networks. Nat Commun 2024; 15:1602. [PMID: 38383500 PMCID: PMC10881569 DOI: 10.1038/s41467-024-45886-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024] Open
Abstract
Kinetic modeling of in vitro enzymatic reaction networks is vital to understand and control the complex behaviors emerging from the nonlinear interactions inside. However, modeling is severely hampered by the lack of training data. Here, we introduce a methodology that combines an active learning-like approach and flow chemistry to efficiently create optimized datasets for a highly interconnected enzymatic reactions network with multiple sub-pathways. The optimal experimental design (OED) algorithm designs a sequence of out-of-equilibrium perturbations to maximize the information about the reaction kinetics, yielding a descriptive model that allows control of the output of the network towards any cost function. We experimentally validate the model by forcing the network to produce different product ratios while maintaining a minimum level of overall conversion efficiency. Our workflow scales with the complexity of the system and enables the optimization of previously unobtainable network outputs.
Collapse
Affiliation(s)
- Bob van Sluijs
- Institute for Molecules and Materials, Radboud University, Nijmegen, AJ, The Netherlands
| | - Tao Zhou
- Institute for Molecules and Materials, Radboud University, Nijmegen, AJ, The Netherlands.
| | - Britta Helwig
- Institute for Molecules and Materials, Radboud University, Nijmegen, AJ, The Netherlands
| | - Mathieu G Baltussen
- Institute for Molecules and Materials, Radboud University, Nijmegen, AJ, The Netherlands
| | - Frank H T Nelissen
- Institute for Molecules and Materials, Radboud University, Nijmegen, AJ, The Netherlands
| | - Hans A Heus
- Institute for Molecules and Materials, Radboud University, Nijmegen, AJ, The Netherlands
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Nijmegen, AJ, The Netherlands.
| |
Collapse
|
12
|
Tummler K, Klipp E. Data integration strategies for whole-cell modeling. FEMS Yeast Res 2024; 24:foae011. [PMID: 38544322 PMCID: PMC11042497 DOI: 10.1093/femsyr/foae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
Data makes the world go round-and high quality data is a prerequisite for precise models, especially for whole-cell models (WCM). Data for WCM must be reusable, contain information about the exact experimental background, and should-in its entirety-cover all relevant processes in the cell. Here, we review basic requirements to data for WCM and strategies how to combine them. As a species-specific resource, we introduce the Yeast Cell Model Data Base (YCMDB) to illustrate requirements and solutions. We discuss recent standards for data as well as for computational models including the modeling process as data to be reported. We outline strategies for constructions of WCM despite their inherent complexity.
Collapse
Affiliation(s)
- Katja Tummler
- Humboldt-Universität zu Berlin, Faculty of Life Sciences, Institute of Biology, Theoretical Biophysics,, Invalidenstr. 42, 10115 Berlin, Germany
| | - Edda Klipp
- Humboldt-Universität zu Berlin, Faculty of Life Sciences, Institute of Biology, Theoretical Biophysics,, Invalidenstr. 42, 10115 Berlin, Germany
| |
Collapse
|
13
|
Lang PF, Penas DR, Banga JR, Weindl D, Novak B. Reusable rule-based cell cycle model explains compartment-resolved dynamics of 16 observables in RPE-1 cells. PLoS Comput Biol 2024; 20:e1011151. [PMID: 38190398 PMCID: PMC10773963 DOI: 10.1371/journal.pcbi.1011151] [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: 05/03/2023] [Accepted: 11/24/2023] [Indexed: 01/10/2024] Open
Abstract
The mammalian cell cycle is regulated by a well-studied but complex biochemical reaction system. Computational models provide a particularly systematic and systemic description of the mechanisms governing mammalian cell cycle control. By combining both state-of-the-art multiplexed experimental methods and powerful computational tools, this work aims at improving on these models along four dimensions: model structure, validation data, validation methodology and model reusability. We developed a comprehensive model structure of the full cell cycle that qualitatively explains the behaviour of human retinal pigment epithelial-1 cells. To estimate the model parameters, time courses of eight cell cycle regulators in two compartments were reconstructed from single cell snapshot measurements. After optimisation with a parallel global optimisation metaheuristic we obtained excellent agreements between simulations and measurements. The PEtab specification of the optimisation problem facilitates reuse of model, data and/or optimisation results. Future perturbation experiments will improve parameter identifiability and allow for testing model predictive power. Such a predictive model may aid in drug discovery for cell cycle-related disorders.
Collapse
Affiliation(s)
- Paul F. Lang
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - David R. Penas
- Computational Biology Lab, MBG-CSIC (Spanish National Research Council), Pontevedra, Spain
| | - Julio R. Banga
- Computational Biology Lab, MBG-CSIC (Spanish National Research Council), Pontevedra, Spain
| | - Daniel Weindl
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Bela Novak
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Viswan NA, Bhalla US. Understanding molecular signaling cascades in neural disease using multi-resolution models. Curr Opin Neurobiol 2023; 83:102808. [PMID: 37972535 DOI: 10.1016/j.conb.2023.102808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/10/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
If the genome defines the program for the operations of a cell, signaling networks execute it. These cascades of chemical, cell-biological, structural, and trafficking events span milliseconds (e.g., synaptic release) to potentially a lifetime (e.g., stabilization of dendritic spines). In principle almost every aspect of neuronal function, particularly at the synapse, depends on signaling. Thus dysfunction of these cascades, whether through mutations, local dysregulation, or infection, leads to disease. The sheer complexity of these pathways is matched by the range of diseases and the diversity of their phenotypes. In this review, we discuss how to build computational models, how these models are essential to tackle this complexity, and the benefits of using families of models at different levels of detail to understand signaling in health and disease.
Collapse
Affiliation(s)
- Nisha Ann Viswan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bellary Road, Bengaluru, 560065, India; The University of Trans-Disciplinary Health Sciences and Technology, Bangalore, India. https://twitter.com/nishanna
| | - Upinder Singh Bhalla
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bellary Road, Bengaluru, 560065, India.
| |
Collapse
|
15
|
Raimúndez E, Fedders M, Hasenauer J. Posterior marginalization accelerates Bayesian inference for dynamical models of biological processes. iScience 2023; 26:108083. [PMID: 37867942 PMCID: PMC10589897 DOI: 10.1016/j.isci.2023.108083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/16/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Bayesian inference is an important method in the life and natural sciences for learning from data. It provides information about parameter and prediction uncertainties. Yet, generating representative samples from the posterior distribution is often computationally challenging. Here, we present an approach that lowers the computational complexity of sample generation for dynamical models with scaling, offset, and noise parameters. The proposed method is based on the marginalization of the posterior distribution. We provide analytical results for a broad class of problems with conjugate priors and show that the method is suitable for a large number of applications. Subsequently, we demonstrate the benefit of the approach for applications from the field of systems biology. We report an improvement up to 50 times in the effective sample size per unit of time. As the scheme is broadly applicable, it will facilitate Bayesian inference in different research fields.
Collapse
Affiliation(s)
- Elba Raimúndez
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Technische Universität München, Center for Mathematics, Garching, Germany
| | - Michael Fedders
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jan Hasenauer
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Technische Universität München, Center for Mathematics, Garching, Germany
- Helmholtz Zentrum München - German Research Center for Environmental Health, Computational Health Center, Neuherberg, Germany
| |
Collapse
|
16
|
Schälte Y, Fröhlich F, Jost PJ, Vanhoefer J, Pathirana D, Stapor P, Lakrisenko P, Wang D, Raimúndez E, Merkt S, Schmiester L, Städter P, Grein S, Dudkin E, Doresic D, Weindl D, Hasenauer J. pyPESTO: a modular and scalable tool for parameter estimation for dynamic models. Bioinformatics 2023; 39:btad711. [PMID: 37995297 PMCID: PMC10689677 DOI: 10.1093/bioinformatics/btad711] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 10/02/2023] [Accepted: 11/22/2023] [Indexed: 11/25/2023] Open
Abstract
SUMMARY Mechanistic models are important tools to describe and understand biological processes. However, they typically rely on unknown parameters, the estimation of which can be challenging for large and complex systems. pyPESTO is a modular framework for systematic parameter estimation, with scalable algorithms for optimization and uncertainty quantification. While tailored to ordinary differential equation problems, pyPESTO is broadly applicable to black-box parameter estimation problems. Besides own implementations, it provides a unified interface to various popular simulation and inference methods. AVAILABILITY AND IMPLEMENTATION pyPESTO is implemented in Python, open-source under a 3-Clause BSD license. Code and documentation are available on GitHub (https://github.com/icb-dcm/pypesto).
Collapse
Affiliation(s)
- Yannik Schälte
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| | - Fabian Fröhlich
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Paul J Jost
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
| | - Jakob Vanhoefer
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
| | - Dilan Pathirana
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
| | - Paul Stapor
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| | - Polina Lakrisenko
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
- School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Dantong Wang
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| | - Elba Raimúndez
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| | - Simon Merkt
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
| | - Leonard Schmiester
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| | - Philipp Städter
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
- Leibniz Institute for Natural Product Research and Infection Biology, 07745 Jena, Germany
| | - Stephan Grein
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
| | - Erika Dudkin
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
| | - Domagoj Doresic
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
| | - Daniel Weindl
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
| | - Jan Hasenauer
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53113 Bonn, Germany
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| |
Collapse
|
17
|
Alamoudi E, Schälte Y, Müller R, Starruß J, Bundgaard N, Graw F, Brusch L, Hasenauer J. FitMultiCell: simulating and parameterizing computational models of multi-scale and multi-cellular processes. Bioinformatics 2023; 39:btad674. [PMID: 37947308 PMCID: PMC10666203 DOI: 10.1093/bioinformatics/btad674] [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: 02/21/2023] [Revised: 10/25/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023] Open
Abstract
MOTIVATION Biological tissues are dynamic and highly organized. Multi-scale models are helpful tools to analyse and understand the processes determining tissue dynamics. These models usually depend on parameters that need to be inferred from experimental data to achieve a quantitative understanding, to predict the response to perturbations, and to evaluate competing hypotheses. However, even advanced inference approaches such as approximate Bayesian computation (ABC) are difficult to apply due to the computational complexity of the simulation of multi-scale models. Thus, there is a need for a scalable pipeline for modeling, simulating, and parameterizing multi-scale models of multi-cellular processes. RESULTS Here, we present FitMultiCell, a computationally efficient and user-friendly open-source pipeline that can handle the full workflow of modeling, simulating, and parameterizing for multi-scale models of multi-cellular processes. The pipeline is modular and integrates the modeling and simulation tool Morpheus and the statistical inference tool pyABC. The easy integration of high-performance infrastructure allows to scale to computationally expensive problems. The introduction of a novel standard for the formulation of parameter inference problems for multi-scale models additionally ensures reproducibility and reusability. By applying the pipeline to multiple biological problems, we demonstrate its broad applicability, which will benefit in particular image-based systems biology. AVAILABILITY AND IMPLEMENTATION FitMultiCell is available open-source at https://gitlab.com/fitmulticell/fit.
Collapse
Affiliation(s)
- Emad Alamoudi
- Life and Medical Sciences Institute, University of Bonn, Bonn 53113, Germany
| | - Yannik Schälte
- Life and Medical Sciences Institute, University of Bonn, Bonn 53113, Germany
- Institute of Computational Biology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg 85764, Germany
- Center for Mathematics, Chair of Mathematical Modeling of Biological Systems, Technische Universität München, Garching 85748, Germany
| | - Robert Müller
- Center of Information Services and High Performance Computing (ZIH), Technische Universität Dresden, Dresden 01062, Germany
| | - Jörn Starruß
- Center of Information Services and High Performance Computing (ZIH), Technische Universität Dresden, Dresden 01062, Germany
| | - Nils Bundgaard
- BioQuant—Center for Quantitative Biology, Heidelberg University, Heidelberg 69120, Germany
| | - Frederik Graw
- BioQuant—Center for Quantitative Biology, Heidelberg University, Heidelberg 69120, Germany
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg 69120, Germany
- Department of Medicine 5, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Lutz Brusch
- Center of Information Services and High Performance Computing (ZIH), Technische Universität Dresden, Dresden 01062, Germany
| | - Jan Hasenauer
- Life and Medical Sciences Institute, University of Bonn, Bonn 53113, Germany
- Institute of Computational Biology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg 85764, Germany
- Center for Mathematics, Chair of Mathematical Modeling of Biological Systems, Technische Universität München, Garching 85748, Germany
| |
Collapse
|
18
|
Wu J, Stewart WCL, Jayaprakash C, Das J. BioNetGMMFit: estimating parameters of a BioNetGen model from time-stamped snapshots of single cells. NPJ Syst Biol Appl 2023; 9:46. [PMID: 37736766 PMCID: PMC10516955 DOI: 10.1038/s41540-023-00299-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 07/31/2023] [Indexed: 09/23/2023] Open
Abstract
Mechanistic models are commonly employed to describe signaling and gene regulatory kinetics in single cells and cell populations. Recent advances in single-cell technologies have produced multidimensional datasets where snapshots of copy numbers (or abundances) of a large number of proteins and mRNA are measured across time in single cells. The availability of such datasets presents an attractive scenario where mechanistic models are validated against experiments, and estimated model parameters enable quantitative predictions of signaling or gene regulatory kinetics. To empower the systems biology community to easily estimate parameters accurately from multidimensional single-cell data, we have merged a widely used rule-based modeling software package BioNetGen, which provides a user-friendly way to code for mechanistic models describing biochemical reactions, and the recently introduced CyGMM, that uses cell-to-cell differences to improve parameter estimation for such networks, into a single software package: BioNetGMMFit. BioNetGMMFit provides parameter estimates of the model, supplied by the user in the BioNetGen markup language (BNGL), which yield the best fit for the observed single-cell, time-stamped data of cellular components. Furthermore, for more precise estimates, our software generates confidence intervals around each model parameter. BioNetGMMFit is capable of fitting datasets of increasing cell population sizes for any mechanistic model specified in the BioNetGen markup language. By streamlining the process of developing mechanistic models for large single-cell datasets, BioNetGMMFit provides an easily-accessible modeling framework designed for scale and the broader biochemical signaling community.
Collapse
Affiliation(s)
- John Wu
- Department of Computer Science, The Ohio State University, 281 W Lane Ave, Columbus, OH, 43210, USA.
- Steve and Cindy Rasmussen Institute for Genomics, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA.
| | | | - Ciriyam Jayaprakash
- Department of Physics, The Ohio State University, 191 W Woodruff Ave, Columbus, OH, 43210, USA
| | - Jayajit Das
- Steve and Cindy Rasmussen Institute for Genomics, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA.
- Departments of Pediatrics, Biomedical Informatics, Pelotonia Institute of Immuno-Oncology, College of Medicine, and Biophysics Program, The Ohio State University, 370 W 9th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
19
|
Cortesi M, Liu D, Yee C, Marsh DJ, Ford CE. A comparative analysis of 2D and 3D experimental data for the identification of the parameters of computational models. Sci Rep 2023; 13:15769. [PMID: 37737283 PMCID: PMC10517149 DOI: 10.1038/s41598-023-42486-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
Computational models are becoming an increasingly valuable tool in biomedical research. Their accuracy and effectiveness, however, rely on the identification of suitable parameters and on appropriate validation of the in-silico framework. Both these steps are highly dependent on the experimental model used as a reference to acquire the data. Selecting the most appropriate experimental framework thus becomes key, together with the analysis of the effect of combining results from different experimental models, a common practice often necessary due to limited data availability. In this work, the same in-silico model of ovarian cancer cell growth and metastasis, was calibrated with datasets acquired from traditional 2D monolayers, 3D cell culture models or a combination of the two. The comparison between the parameters sets obtained in the different conditions, together with the corresponding simulated behaviours, is presented. It provides a framework for the study of the effect of the different experimental models on the development of computational systems. This work also provides a set of general guidelines for the comparative testing and selection of experimental models and protocols to be used for parameter optimization in computational models.
Collapse
Affiliation(s)
- Marilisa Cortesi
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, Australia.
- Laboratory of Cellular and Molecular Engineering, Department of Electrical Electronic and Information Engineering "G. Marconi", Alma Mater Studiorum-University of Bologna, Cesena, Italy.
| | - Dongli Liu
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, Australia
| | - Christine Yee
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Deborah J Marsh
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Caroline E Ford
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, Australia.
| |
Collapse
|
20
|
Deepa Maheshvare M, Raha S, König M, Pal D. A pathway model of glucose-stimulated insulin secretion in the pancreatic β-cell. Front Endocrinol (Lausanne) 2023; 14:1185656. [PMID: 37600713 PMCID: PMC10433753 DOI: 10.3389/fendo.2023.1185656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/08/2023] [Indexed: 08/22/2023] Open
Abstract
The pancreas plays a critical role in maintaining glucose homeostasis through the secretion of hormones from the islets of Langerhans. Glucose-stimulated insulin secretion (GSIS) by the pancreatic β-cell is the main mechanism for reducing elevated plasma glucose. Here we present a systematic modeling workflow for the development of kinetic pathway models using the Systems Biology Markup Language (SBML). Steps include retrieval of information from databases, curation of experimental and clinical data for model calibration and validation, integration of heterogeneous data including absolute and relative measurements, unit normalization, data normalization, and model annotation. An important factor was the reproducibility and exchangeability of the model, which allowed the use of various existing tools. The workflow was applied to construct a novel data-driven kinetic model of GSIS in the pancreatic β-cell based on experimental and clinical data from 39 studies spanning 50 years of pancreatic, islet, and β-cell research in humans, rats, mice, and cell lines. The model consists of detailed glycolysis and phenomenological equations for insulin secretion coupled to cellular energy state, ATP dynamics and (ATP/ADP ratio). Key findings of our work are that in GSIS there is a glucose-dependent increase in almost all intermediates of glycolysis. This increase in glycolytic metabolites is accompanied by an increase in energy metabolites, especially ATP and NADH. One of the few decreasing metabolites is ADP, which, in combination with the increase in ATP, results in a large increase in ATP/ADP ratios in the β-cell with increasing glucose. Insulin secretion is dependent on ATP/ADP, resulting in glucose-stimulated insulin secretion. The observed glucose-dependent increase in glycolytic intermediates and the resulting change in ATP/ADP ratios and insulin secretion is a robust phenomenon observed across data sets, experimental systems and species. Model predictions of the glucose-dependent response of glycolytic intermediates and biphasic insulin secretion are in good agreement with experimental measurements. Our model predicts that factors affecting ATP consumption, ATP formation, hexokinase, phosphofructokinase, and ATP/ADP-dependent insulin secretion have a major effect on GSIS. In conclusion, we have developed and applied a systematic modeling workflow for pathway models that allowed us to gain insight into key mechanisms in GSIS in the pancreatic β-cell.
Collapse
Affiliation(s)
- M. Deepa Maheshvare
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, India
| | - Soumyendu Raha
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, India
| | - Matthias König
- Institute for Biology, Institute for Theoretical Biology, Humboldt-University Berlin, Berlin, Germany
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, India
| |
Collapse
|
21
|
Tatka LT, Smith LP, Hellerstein JL, Sauro HM. Adapting modeling and simulation credibility standards to computational systems biology. J Transl Med 2023; 21:501. [PMID: 37496031 PMCID: PMC10369698 DOI: 10.1186/s12967-023-04290-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023] Open
Abstract
Computational models are increasingly used in high-impact decision making in science, engineering, and medicine. The National Aeronautics and Space Administration (NASA) uses computational models to perform complex experiments that are otherwise prohibitively expensive or require a microgravity environment. Similarly, the Food and Drug Administration (FDA) and European Medicines Agency (EMA) have began accepting models and simulations as forms of evidence for pharmaceutical and medical device approval. It is crucial that computational models meet a standard of credibility when using them in high-stakes decision making. For this reason, institutes including NASA, the FDA, and the EMA have developed standards to promote and assess the credibility of computational models and simulations. However, due to the breadth of models these institutes assess, these credibility standards are mostly qualitative and avoid making specific recommendations. On the other hand, modeling and simulation in systems biology is a narrower domain and several standards are already in place. As systems biology models increase in complexity and influence, the development of a credibility assessment system is crucial. Here we review existing standards in systems biology, credibility standards in other science, engineering, and medical fields, and propose the development of a credibility standard for systems biology models.
Collapse
Affiliation(s)
- Lillian T Tatka
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| | - Lucian P Smith
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | - Herbert M Sauro
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
22
|
Contento L, Castelletti N, Raimúndez E, Le Gleut R, Schälte Y, Stapor P, Hinske LC, Hoelscher M, Wieser A, Radon K, Fuchs C, Hasenauer J. Integrative modelling of reported case numbers and seroprevalence reveals time-dependent test efficiency and infectious contacts. Epidemics 2023; 43:100681. [PMID: 36931114 PMCID: PMC10008049 DOI: 10.1016/j.epidem.2023.100681] [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: 10/06/2021] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Mathematical models have been widely used during the ongoing SARS-CoV-2 pandemic for data interpretation, forecasting, and policy making. However, most models are based on officially reported case numbers, which depend on test availability and test strategies. The time dependence of these factors renders interpretation difficult and might even result in estimation biases. Here, we present a computational modelling framework that allows for the integration of reported case numbers with seroprevalence estimates obtained from representative population cohorts. To account for the time dependence of infection and testing rates, we embed flexible splines in an epidemiological model. The parameters of these splines are estimated, along with the other parameters, from the available data using a Bayesian approach. The application of this approach to the official case numbers reported for Munich (Germany) and the seroprevalence reported by the prospective COVID-19 Cohort Munich (KoCo19) provides first estimates for the time dependence of the under-reporting factor. Furthermore, we estimate how the effectiveness of non-pharmaceutical interventions and of the testing strategy evolves over time. Overall, our results show that the integration of temporally highly resolved and representative data is beneficial for accurate epidemiological analyses.
Collapse
Affiliation(s)
- Lorenzo Contento
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany.
| | - Noemi Castelletti
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Elba Raimúndez
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany; Center for Mathematics, Technische Universität München, Garching, Germany
| | - Ronan Le Gleut
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Core Facility Statistical Consulting, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Yannik Schälte
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Center for Mathematics, Technische Universität München, Garching, Germany
| | - Paul Stapor
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Center for Mathematics, Technische Universität München, Garching, Germany
| | - Ludwig Christian Hinske
- Institut für medizinische Informationsverarbeitung, Biometrie und Epidemiologie, Munich, Germany
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany; Center for International Health (CIH), University Hospital, LMU Munich, Munich, Germany; German Center for Infection Research (DZIF), partner site Munich, Germany
| | - Andreas Wieser
- Division of Infectious Diseases and Tropical Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Infection Research (DZIF), partner site Munich, Germany
| | - Katja Radon
- German Center for Infection Research (DZIF), partner site Munich, Germany; Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany; Comprehensive Pneumology Center (CPC) Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Christiane Fuchs
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Core Facility Statistical Consulting, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Center for Mathematics, Technische Universität München, Garching, Germany; Faculty of Business Administration and Economics, Bielefeld University, Bielefeld, Germany
| | - Jan Hasenauer
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany; Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Center for Mathematics, Technische Universität München, Garching, Germany
| |
Collapse
|
23
|
Egert J, Kreutz C. Realistic simulation of time-course measurements in systems biology. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:10570-10589. [PMID: 37322949 DOI: 10.3934/mbe.2023467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In systems biology, the analysis of complex nonlinear systems faces many methodological challenges. For the evaluation and comparison of the performances of novel and competing computational methods, one major bottleneck is the availability of realistic test problems. We present an approach for performing realistic simulation studies for analyses of time course data as they are typically measured in systems biology. Since the design of experiments in practice depends on the process of interest, our approach considers the size and the dynamics of the mathematical model which is intended to be used for the simulation study. To this end, we used 19 published systems biology models with experimental data and evaluated the relationship between model features (e.g., the size and the dynamics) and features of the measurements such as the number and type of observed quantities, the number and the selection of measurement times, and the magnitude of measurement errors. Based on these typical relationships, our novel approach enables suggestions of realistic simulation study designs in the systems biology context and the realistic generation of simulated data for any dynamic model. The approach is demonstrated on three models in detail and its performance is validated on nine models by comparing ODE integration, parameter optimization, and parameter identifiability. The presented approach enables more realistic and less biased benchmark studies and thereby constitutes an important tool for the development of novel methods for dynamic modeling.
Collapse
Affiliation(s)
- Janine Egert
- Institute of Medical Biometry and Statistics (IMBI), Faculty of Medicine and Medical Center - University of Freiburg, Stefan-Meier-Str. 26, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Clemens Kreutz
- Institute of Medical Biometry and Statistics (IMBI), Faculty of Medicine and Medical Center - University of Freiburg, Stefan-Meier-Str. 26, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies CIBSS, University of Freiburg, 79104 Freiburg, Germany
- Center for Data Analysis and Modelling (FDM), University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
24
|
Mutsuddy A, Erdem C, Huggins JR, Salim M, Cook D, Hobbs N, Feltus FA, Birtwistle MR. Computational speed-up of large-scale, single-cell model simulations via a fully integrated SBML-based format. BIOINFORMATICS ADVANCES 2023; 3:vbad039. [PMID: 37020976 PMCID: PMC10070034 DOI: 10.1093/bioadv/vbad039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/25/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023]
Abstract
Summary Large-scale and whole-cell modeling has multiple challenges, including scalable model building and module communication bottlenecks (e.g. between metabolism, gene expression, signaling, etc.). We previously developed an open-source, scalable format for a large-scale mechanistic model of proliferation and death signaling dynamics, but communication bottlenecks between gene expression and protein biochemistry modules remained. Here, we developed two solutions to communication bottlenecks that speed-up simulation by ∼4-fold for hybrid stochastic-deterministic simulations and by over 100-fold for fully deterministic simulations. Fully deterministic speed-up facilitates model initialization, parameter estimation and sensitivity analysis tasks. Availability and implementation Source code is freely available at https://github.com/birtwistlelab/SPARCED/releases/tag/v1.3.0 implemented in python, and supported on Linux, Windows and MacOS (via Docker).
Collapse
Affiliation(s)
- Arnab Mutsuddy
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Cemal Erdem
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Jonah R Huggins
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
- School of Computing, Clemson University, Clemson, SC, USA
| | | | | | | | - F Alex Feltus
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA
| | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| |
Collapse
|
25
|
Höpfl S, Pleiss J, Radde NE. Bayesian estimation reveals that reproducible models in Systems Biology get more citations. Sci Rep 2023; 13:2695. [PMID: 36792648 PMCID: PMC9931699 DOI: 10.1038/s41598-023-29340-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
The Systems Biology community has taken numerous actions to develop data and modeling standards towards FAIR data and model handling. Nevertheless, the debate about incentives and rewards for individual researchers to make their results reproducible is ongoing. Here, we pose the specific question of whether reproducible models have a higher impact in terms of citations. Therefore, we statistically analyze 328 published models recently classified by Tiwari et al. based on their reproducibility. For hypothesis testing, we use a flexible Bayesian approach that provides complete distributional information for all quantities of interest and can handle outliers. The results show that in the period from 2013, i.e., 10 years after the introduction of SBML, to 2020, the group of reproducible models is significantly more cited than the non-reproducible group. We show that differences in journal impact factors do not explain this effect and that this effect increases with additional standardization of data and error model integration via PEtab. Overall, our statistical analysis demonstrates the long-term merits of reproducible modeling for the individual researcher in terms of citations. Moreover, it provides evidence for the increased use of reproducible models in the scientific community.
Collapse
Affiliation(s)
- Sebastian Höpfl
- Institute for Systems Theory and Automatic Control, University of Stuttgart, Pfaffenwaldring 9, 70569, Stuttgart, Germany
| | - Jürgen Pleiss
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Nicole E Radde
- Institute for Systems Theory and Automatic Control, University of Stuttgart, Pfaffenwaldring 9, 70569, Stuttgart, Germany.
| |
Collapse
|
26
|
Lakrisenko P, Stapor P, Grein S, Paszkowski Ł, Pathirana D, Fröhlich F, Lines GT, Weindl D, Hasenauer J. Efficient computation of adjoint sensitivities at steady-state in ODE models of biochemical reaction networks. PLoS Comput Biol 2023; 19:e1010783. [PMID: 36595539 PMCID: PMC9838866 DOI: 10.1371/journal.pcbi.1010783] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/13/2023] [Accepted: 12/01/2022] [Indexed: 01/04/2023] Open
Abstract
Dynamical models in the form of systems of ordinary differential equations have become a standard tool in systems biology. Many parameters of such models are usually unknown and have to be inferred from experimental data. Gradient-based optimization has proven to be effective for parameter estimation. However, computing gradients becomes increasingly costly for larger models, which are required for capturing the complex interactions of multiple biochemical pathways. Adjoint sensitivity analysis has been pivotal for working with such large models, but methods tailored for steady-state data are currently not available. We propose a new adjoint method for computing gradients, which is applicable if the experimental data include steady-state measurements. The method is based on a reformulation of the backward integration problem to a system of linear algebraic equations. The evaluation of the proposed method using real-world problems shows a speedup of total simulation time by a factor of up to 4.4. Our results demonstrate that the proposed approach can achieve a substantial improvement in computation time, in particular for large-scale models, where computational efficiency is critical.
Collapse
Affiliation(s)
- Polina Lakrisenko
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Center for Mathematics, Technische Universität München, Garching, Germany
| | - Paul Stapor
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Center for Mathematics, Technische Universität München, Garching, Germany
| | - Stephan Grein
- University of Bonn, Life and Medical Sciences Institute, Bonn, Germany
| | | | - Dilan Pathirana
- University of Bonn, Life and Medical Sciences Institute, Bonn, Germany
| | - Fabian Fröhlich
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Daniel Weindl
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Jan Hasenauer
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- University of Bonn, Life and Medical Sciences Institute, Bonn, Germany
| |
Collapse
|
27
|
Mishra S, Wang Z, Volk MJ, Zhao H. Design and application of a kinetic model of lipid metabolism in Saccharomyces cerevisiae. Metab Eng 2023; 75:12-18. [PMID: 36371031 DOI: 10.1016/j.ymben.2022.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 11/10/2022]
Abstract
Lipid biosynthesis plays a vital role in living cells and has been increasingly engineered to overproduce various lipid-based chemicals. However, owing to the tightly constrained and interconnected nature of lipid biosynthesis, both understanding and engineering of lipid metabolism remain challenging, even with the help of mathematical models. Here we report the development of a kinetic metabolic model of lipid metabolism in Saccharomyces cerevisiae that integrates fatty acid biosynthesis, glycerophospholipid metabolism, sphingolipid metabolism, storage lipids, lumped sterol synthesis, and the synthesis and transport of relevant target-chemicals, such as fatty acids and fatty alcohols. The model was trained on lipidomic data of a reference S. cerevisiae strain, single knockout mutants, and lipid overproduction strains reported in literature. The model was used to design mutants for fatty alcohol overproduction and the lipidomic analysis of the resultant mutant strains coupled with model-guided hypothesis led to discovery of a futile cycle in the triacylglycerol biosynthesis pathway. In addition, the model was used to explain successful and unsuccessful mutant designs in metabolic engineering literature. Thus, this kinetic model of lipid metabolism can not only enable the discovery of new phenomenon in lipid metabolism but also the engineering of mutant strains for overproduction of lipids.
Collapse
Affiliation(s)
- Shekhar Mishra
- Department of Chemical and Biomolecular Engineering, Department of Energy Center for Advanced Bioenergy and Bioproducts Innovation, Carl R. Woese Institute for Genomic Biology, USA
| | | | - Michael J Volk
- Department of Chemical and Biomolecular Engineering, Department of Energy Center for Advanced Bioenergy and Bioproducts Innovation, Carl R. Woese Institute for Genomic Biology, USA
| | - Huimin Zhao
- Department of Chemical and Biomolecular Engineering, Department of Energy Center for Advanced Bioenergy and Bioproducts Innovation, Carl R. Woese Institute for Genomic Biology, USA; Department of Biochemistry, USA; Departments of Chemistry and Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
28
|
Fröhlich F. A Practical Guide for the Efficient Formulation and Calibration of Large, Energy- and Rule-Based Models of Cellular Signal Transduction. Methods Mol Biol 2023; 2634:59-86. [PMID: 37074574 DOI: 10.1007/978-1-0716-3008-2_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Aberrant signal transduction leads to complex diseases such as cancer. To rationally design treatment strategies with small molecule inhibitors, computational models have to be employed. Energy- and rule-based models allow the construction of mechanistic ordinary differential equation models based on structural insights. The detailed, energy-based description often generates large models, which are difficult to calibrate on experimental data. In this chapter, we provide a detailed, interactive protocol for the programmatic formulation and calibration of such large, energy- and rule-based models of cellular signal transduction based on an example model describing the action of RAF inhibitors on MAPK signaling. An interactive version of this chapter is available as Jupyter Notebook at github.com/FFroehlich/energy_modeling_chapter .
Collapse
Affiliation(s)
- Fabian Fröhlich
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Albadry M, Höpfl S, Ehteshamzad N, König M, Böttcher M, Neumann J, Lupp A, Dirsch O, Radde N, Christ B, Christ M, Schwen LO, Laue H, Klopfleisch R, Dahmen U. Periportal steatosis in mice affects distinct parameters of pericentral drug metabolism. Sci Rep 2022; 12:21825. [PMID: 36528753 PMCID: PMC9759570 DOI: 10.1038/s41598-022-26483-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Little is known about the impact of morphological disorders in distinct zones on metabolic zonation. It was described recently that periportal fibrosis did affect the expression of CYP proteins, a set of pericentrally located drug-metabolizing enzymes. Here, we investigated whether periportal steatosis might have a similar effect. Periportal steatosis was induced in C57BL6/J mice by feeding a high-fat diet with low methionine/choline content for either two or four weeks. Steatosis severity was quantified using image analysis. Triglycerides and CYP activity were quantified in photometric or fluorometric assay. The distribution of CYP3A4, CYP1A2, CYP2D6, and CYP2E1 was visualized by immunohistochemistry. Pharmacokinetic parameters of test drugs were determined after injecting a drug cocktail (caffeine, codeine, and midazolam). The dietary model resulted in moderate to severe mixed steatosis confined to periportal and midzonal areas. Periportal steatosis did not affect the zonal distribution of CYP expression but the activity of selected CYPs was associated with steatosis severity. Caffeine elimination was accelerated by microvesicular steatosis, whereas midazolam elimination was delayed in macrovesicular steatosis. In summary, periportal steatosis affected parameters of pericentrally located drug metabolism. This observation calls for further investigations of the highly complex interrelationship between steatosis and drug metabolism and underlying signaling mechanisms.
Collapse
Affiliation(s)
- Mohamed Albadry
- grid.275559.90000 0000 8517 6224Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany ,grid.411775.10000 0004 0621 4712Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkom, Menoufia, Egypt
| | - Sebastian Höpfl
- grid.5719.a0000 0004 1936 9713Institute for Systems Theory and Automatic Control, Faculty of Engineering Design, Production Engineering and Automotive Engineering, University of Stuttgart, Stuttgart, Germany
| | - Nadia Ehteshamzad
- grid.275559.90000 0000 8517 6224Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Matthias König
- grid.7468.d0000 0001 2248 7639Institute for Theoretical Biology, Institute of Biology, Humboldt-University, Berlin, Germany
| | - Michael Böttcher
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstraße 6, 06847 Dessau-Roßlau, Germany
| | - Jasna Neumann
- MVZ Medizinische Labore Dessau Kassel GmbH, Bauhüttenstraße 6, 06847 Dessau-Roßlau, Germany
| | - Amelie Lupp
- grid.275559.90000 0000 8517 6224Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Olaf Dirsch
- grid.459629.50000 0004 0389 4214Institute of Pathology, Klinikum Chemnitz, Chemnitz, Germany
| | - Nicole Radde
- grid.5719.a0000 0004 1936 9713Institute for Systems Theory and Automatic Control, Faculty of Engineering Design, Production Engineering and Automotive Engineering, University of Stuttgart, Stuttgart, Germany
| | - Bruno Christ
- grid.9647.c0000 0004 7669 9786Cell Transplantation/Molecular Hepatology Lab, Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig Medical Center, Leipzig, Germany
| | - Madlen Christ
- grid.9647.c0000 0004 7669 9786Cell Transplantation/Molecular Hepatology Lab, Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig Medical Center, Leipzig, Germany
| | - Lars Ole Schwen
- grid.428590.20000 0004 0496 8246Fraunhofer MEVIS, Max-Von-Laue-Str. 2, 28359 Bremen, Germany
| | - Hendrik Laue
- grid.428590.20000 0004 0496 8246Fraunhofer MEVIS, Max-Von-Laue-Str. 2, 28359 Bremen, Germany
| | - Robert Klopfleisch
- grid.14095.390000 0000 9116 4836Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Uta Dahmen
- grid.275559.90000 0000 8517 6224Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| |
Collapse
|
30
|
Thomaseth C, Radde NE. The effect of normalisation and error model choice on the distribution of the maximum likelihood estimator for a biochemical reaction. IET Syst Biol 2022; 17:1-13. [PMID: 36440585 PMCID: PMC9931059 DOI: 10.1049/syb2.12055] [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: 09/29/2021] [Revised: 09/13/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022] Open
Abstract
Sparse and noisy measurements make parameter estimation for biochemical reaction networks difficult and might lead to ill-posed optimisation problems. This is potentiated if the data has to be normalised, and only fold changes rather than absolute amounts are available. Here, the authors consider the propagation of measurement noise to the distribution of the maximum likelihood (ML) estimator in an in silico study. Therefore, a model of a reversible reaction is considered, for which reaction rate constants using fold changes is estimated. Noise propagation is analysed for different normalisation strategies and different error models. In particular, accuracy, precision, and asymptotic properties of the ML estimator is investigated. Results show that normalisation by the mean of a time series outperforms normalisation by a single time point in the example provided by the authors. Moreover, the error model with a heavy-tail distribution is slightly more robust to large measurement noise, but, beyond this, the choice of the error model did not have a significant impact on the estimation results provided by the authors.
Collapse
Affiliation(s)
- Caterina Thomaseth
- University of StuttgartInstitute for Systems Theory and Automatic ControlD‐70569 StuttgartGermany
| | - Nicole E. Radde
- University of StuttgartInstitute for Systems Theory and Automatic ControlD‐70569 StuttgartGermany
| |
Collapse
|
31
|
Linden NJ, Kramer B, Rangamani P. Bayesian parameter estimation for dynamical models in systems biology. PLoS Comput Biol 2022; 18:e1010651. [PMID: 36269772 PMCID: PMC9629650 DOI: 10.1371/journal.pcbi.1010651] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/02/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
Abstract
Dynamical systems modeling, particularly via systems of ordinary differential equations, has been used to effectively capture the temporal behavior of different biochemical components in signal transduction networks. Despite the recent advances in experimental measurements, including sensor development and '-omics' studies that have helped populate protein-protein interaction networks in great detail, modeling in systems biology lacks systematic methods to estimate kinetic parameters and quantify associated uncertainties. This is because of multiple reasons, including sparse and noisy experimental measurements, lack of detailed molecular mechanisms underlying the reactions, and missing biochemical interactions. Additionally, the inherent nonlinearities with respect to the states and parameters associated with the system of differential equations further compound the challenges of parameter estimation. In this study, we propose a comprehensive framework for Bayesian parameter estimation and complete quantification of the effects of uncertainties in the data and models. We apply these methods to a series of signaling models of increasing mathematical complexity. Systematic analysis of these dynamical systems showed that parameter estimation depends on data sparsity, noise level, and model structure, including the existence of multiple steady states. These results highlight how focused uncertainty quantification can enrich systems biology modeling and enable additional quantitative analyses for parameter estimation.
Collapse
Affiliation(s)
- Nathaniel J. Linden
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, California, United States of America
| | - Boris Kramer
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, California, United States of America
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, California, United States of America
| |
Collapse
|
32
|
BlotIt—Optimal alignment of Western blot and qPCR experiments. PLoS One 2022; 17:e0264295. [PMID: 35947551 PMCID: PMC9365137 DOI: 10.1371/journal.pone.0264295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022] Open
Abstract
Biological systems are frequently analyzed by means of mechanistic mathematical models. In order to infer model parameters and provide a useful model that can be employed for systems understanding and hypothesis testing, the model is often calibrated on quantitative, time-resolved data. To do so, it is typically important to compare experimental measurements over broad time ranges and various experimental conditions, e.g. perturbations of the biological system. However, most of the established experimental techniques such as Western blot, or quantitative real-time polymerase chain reaction only provide measurements on a relative scale, since different sample volumes, experimental adjustments or varying development times of a gel lead to systematic shifts in the data. In turn, the number of measurements corresponding to the same scale enabling comparability is limited. Here, we present a new flexible method to align measurement data that obeys different scaling factors and compare it to existing normalization approaches. We propose an alignment model to estimate these scaling factors and provide the possibility to adapt this model depending on the measurement technique of interest. In addition, an error model can be specified to adequately weight the different data points and obtain scaling-model based confidence intervals of the finally scaled data points. Our approach is applicable to all sorts of relative measurements and does not need a particular experimental condition that has been measured over all available scales. An implementation of the method is provided with the R package blotIt including refined ways of visualization.
Collapse
|
33
|
Eriksson O, Bhalla US, Blackwell KT, Crook SM, Keller D, Kramer A, Linne ML, Saudargienė A, Wade RC, Hellgren Kotaleski J. Combining hypothesis- and data-driven neuroscience modeling in FAIR workflows. eLife 2022; 11:e69013. [PMID: 35792600 PMCID: PMC9259018 DOI: 10.7554/elife.69013] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/13/2022] [Indexed: 12/22/2022] Open
Abstract
Modeling in neuroscience occurs at the intersection of different points of view and approaches. Typically, hypothesis-driven modeling brings a question into focus so that a model is constructed to investigate a specific hypothesis about how the system works or why certain phenomena are observed. Data-driven modeling, on the other hand, follows a more unbiased approach, with model construction informed by the computationally intensive use of data. At the same time, researchers employ models at different biological scales and at different levels of abstraction. Combining these models while validating them against experimental data increases understanding of the multiscale brain. However, a lack of interoperability, transparency, and reusability of both models and the workflows used to construct them creates barriers for the integration of models representing different biological scales and built using different modeling philosophies. We argue that the same imperatives that drive resources and policy for data - such as the FAIR (Findable, Accessible, Interoperable, Reusable) principles - also support the integration of different modeling approaches. The FAIR principles require that data be shared in formats that are Findable, Accessible, Interoperable, and Reusable. Applying these principles to models and modeling workflows, as well as the data used to constrain and validate them, would allow researchers to find, reuse, question, validate, and extend published models, regardless of whether they are implemented phenomenologically or mechanistically, as a few equations or as a multiscale, hierarchical system. To illustrate these ideas, we use a classical synaptic plasticity model, the Bienenstock-Cooper-Munro rule, as an example due to its long history, different levels of abstraction, and implementation at many scales.
Collapse
Affiliation(s)
- Olivia Eriksson
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of TechnologyStockholmSweden
| | - Upinder Singh Bhalla
- National Center for Biological Sciences, Tata Institute of Fundamental ResearchBangaloreIndia
| | - Kim T Blackwell
- Department of Bioengineering, Volgenau School of Engineering, George Mason UniversityFairfaxUnited States
| | - Sharon M Crook
- School of Mathematical and Statistical Sciences, Arizona State UniversityTempeUnited States
| | - Daniel Keller
- Blue Brain Project, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Andrei Kramer
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of TechnologyStockholmSweden
- Department of Neuroscience, Karolinska InstituteStockholmSweden
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Ausra Saudargienė
- Neuroscience Institute, Lithuanian University of Health SciencesKaunasLithuania
- Department of Informatics, Vytautas Magnus UniversityKaunasLithuania
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS)HeidelbergGermany
- Center for Molecular Biology (ZMBH), ZMBH-DKFZ Alliance, University of HeidelbergHeidelbergGermany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg UniversityHeidelbergGermany
| | - Jeanette Hellgren Kotaleski
- Science for Life Laboratory, School of Electrical Engineering and Computer Science, KTH Royal Institute of TechnologyStockholmSweden
- Department of Neuroscience, Karolinska InstituteStockholmSweden
| |
Collapse
|
34
|
Fröhlich F, Sorger PK. Fides: Reliable trust-region optimization for parameter estimation of ordinary differential equation models. PLoS Comput Biol 2022; 18:e1010322. [PMID: 35830470 PMCID: PMC9312381 DOI: 10.1371/journal.pcbi.1010322] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/25/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Ordinary differential equation (ODE) models are widely used to study biochemical reactions in cellular networks since they effectively describe the temporal evolution of these networks using mass action kinetics. The parameters of these models are rarely known a priori and must instead be estimated by calibration using experimental data. Optimization-based calibration of ODE models on is often challenging, even for low-dimensional problems. Multiple hypotheses have been advanced to explain why biochemical model calibration is challenging, including non-identifiability of model parameters, but there are few comprehensive studies that test these hypotheses, likely because tools for performing such studies are also lacking. Nonetheless, reliable model calibration is essential for uncertainty analysis, model comparison, and biological interpretation. We implemented an established trust-region method as a modular Python framework (fides) to enable systematic comparison of different approaches to ODE model calibration involving a variety of Hessian approximation schemes. We evaluated fides on a recently developed corpus of biologically realistic benchmark problems for which real experimental data are available. Unexpectedly, we observed high variability in optimizer performance among different implementations of the same mathematical instructions (algorithms). Analysis of possible sources of poor optimizer performance identified limitations in the widely used Gauss-Newton, BFGS and SR1 Hessian approximation schemes. We addressed these drawbacks with a novel hybrid Hessian approximation scheme that enhances optimizer performance and outperforms existing hybrid approaches. When applied to the corpus of test models, we found that fides was on average more reliable and efficient than existing methods using a variety of criteria. We expect fides to be broadly useful for ODE constrained optimization problems in biochemical models and to be a foundation for future methods development.
Collapse
Affiliation(s)
- Fabian Fröhlich
- Laboratory of Systems Pharmacology and Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter K. Sorger
- Laboratory of Systems Pharmacology and Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
35
|
Deciphering signal transduction networks in the liver by mechanistic mathematical modelling. Biochem J 2022; 479:1361-1374. [PMID: 35748700 PMCID: PMC9246346 DOI: 10.1042/bcj20210548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022]
Abstract
In health and disease, liver cells are continuously exposed to cytokines and growth factors. While individual signal transduction pathways induced by these factors were studied in great detail, the cellular responses induced by repeated or combined stimulations are complex and less understood. Growth factor receptors on the cell surface of hepatocytes were shown to be regulated by receptor interactions, receptor trafficking and feedback regulation. Here, we exemplify how mechanistic mathematical modelling based on quantitative data can be employed to disentangle these interactions at the molecular level. Crucial is the analysis at a mechanistic level based on quantitative longitudinal data within a mathematical framework. In such multi-layered information, step-wise mathematical modelling using submodules is of advantage, which is fostered by sharing of standardized experimental data and mathematical models. Integration of signal transduction with metabolic regulation in the liver and mechanistic links to translational approaches promise to provide predictive tools for biology and personalized medicine.
Collapse
|
36
|
Kemmer S, Berdiel-Acer M, Reinz E, Sonntag J, Tarade N, Bernhardt S, Fehling-Kaschek M, Hasmann M, Korf U, Wiemann S, Timmer J. Disentangling ERBB Signaling in Breast Cancer Subtypes-A Model-Based Analysis. Cancers (Basel) 2022; 14:2379. [PMID: 35625984 PMCID: PMC9139462 DOI: 10.3390/cancers14102379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 01/27/2023] Open
Abstract
Targeted therapies have shown striking success in the treatment of cancer over the last years. However, their specific effects on an individual tumor appear to be varying and difficult to predict. Using an integrative modeling approach that combines mechanistic and regression modeling, we gained insights into the response mechanisms of breast cancer cells due to different ligand-drug combinations. The multi-pathway model, capturing ERBB receptor signaling as well as downstream MAPK and PI3K pathways was calibrated on time-resolved data of the luminal breast cancer cell lines MCF7 and T47D across an array of four ligands and five drugs. The same model was then successfully applied to triple negative and HER2-positive breast cancer cell lines, requiring adjustments mostly for the respective receptor compositions within these cell lines. The additional relevance of cell-line-specific mutations in the MAPK and PI3K pathway components was identified via L1 regularization, where the impact of these mutations on pathway activation was uncovered. Finally, we predicted and experimentally validated the proliferation response of cells to drug co-treatments. We developed a unified mathematical model that can describe the ERBB receptor and downstream signaling in response to therapeutic drugs targeting this clinically relevant signaling network in cell line that represent three major subtypes of breast cancer. Our data and model suggest that alterations in this network could render anti-HER therapies relevant beyond the HER2-positive subtype.
Collapse
Affiliation(s)
- Svenja Kemmer
- Institute of Physics, University of Freiburg, 79104 Freiburg, Germany; (S.K.); (M.F.-K.)
- FDM—Freiburg Center for Data Analysis and Modeling, University of Freiburg, 79104 Freiburg, Germany
| | - Mireia Berdiel-Acer
- Division of Molecular Genome Analysis, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.-A.); (E.R.); (J.S.); (N.T.); (S.B.); (U.K.)
| | - Eileen Reinz
- Division of Molecular Genome Analysis, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.-A.); (E.R.); (J.S.); (N.T.); (S.B.); (U.K.)
| | - Johanna Sonntag
- Division of Molecular Genome Analysis, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.-A.); (E.R.); (J.S.); (N.T.); (S.B.); (U.K.)
| | - Nooraldeen Tarade
- Division of Molecular Genome Analysis, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.-A.); (E.R.); (J.S.); (N.T.); (S.B.); (U.K.)
- Faculty of Biosciences, University of Heidelberg, 69117 Heidelberg, Germany
| | - Stephan Bernhardt
- Division of Molecular Genome Analysis, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.-A.); (E.R.); (J.S.); (N.T.); (S.B.); (U.K.)
| | - Mirjam Fehling-Kaschek
- Institute of Physics, University of Freiburg, 79104 Freiburg, Germany; (S.K.); (M.F.-K.)
- FDM—Freiburg Center for Data Analysis and Modeling, University of Freiburg, 79104 Freiburg, Germany
| | | | - Ulrike Korf
- Division of Molecular Genome Analysis, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.-A.); (E.R.); (J.S.); (N.T.); (S.B.); (U.K.)
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center, 69120 Heidelberg, Germany; (M.B.-A.); (E.R.); (J.S.); (N.T.); (S.B.); (U.K.)
| | - Jens Timmer
- Institute of Physics, University of Freiburg, 79104 Freiburg, Germany; (S.K.); (M.F.-K.)
- FDM—Freiburg Center for Data Analysis and Modeling, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
37
|
Dynamic modeling of Nrf2 pathway activation in liver cells after toxicant exposure. Sci Rep 2022; 12:7336. [PMID: 35513409 PMCID: PMC9072554 DOI: 10.1038/s41598-022-10857-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
Cells are exposed to oxidative stress and reactive metabolites every day. The Nrf2 signaling pathway responds to oxidative stress by upregulation of antioxidants like glutathione (GSH) to compensate the stress insult and re-establish homeostasis. Although mechanisms describing the interaction between the key pathway constituents Nrf2, Keap1 and p62 are widely reviewed and discussed in literature, quantitative dynamic models bringing together these mechanisms with time-resolved data are limited. Here, we present an ordinary differential equation (ODE) based dynamic model to describe the dynamic response of Nrf2, Keap1, Srxn1 and GSH to oxidative stress caused by the soft-electrophile diethyl maleate (DEM). The time-resolved data obtained by single-cell confocal microscopy of green fluorescent protein (GFP) reporters and qPCR of the Nrf2 pathway components complemented with siRNA knock down experiments, is accurately described by the calibrated mathematical model. We show that the quantitative model can describe the activation of the Nrf2 pathway by compounds with a different mechanism of activation, including drugs which are known for their ability to cause drug induced liver-injury (DILI) i.e., diclofenac (DCF) and omeprazole (OMZ). Finally, we show that our model can reveal differences in the processes leading to altered activation dynamics amongst DILI inducing drugs.
Collapse
|
38
|
Karr J, Malik-Sheriff RS, Osborne J, Gonzalez-Parra G, Forgoston E, Bowness R, Liu Y, Thompson R, Garira W, Barhak J, Rice J, Torres M, Dobrovolny HM, Tang T, Waites W, Glazier JA, Faeder JR, Kulesza A. Model Integration in Computational Biology: The Role of Reproducibility, Credibility and Utility. FRONTIERS IN SYSTEMS BIOLOGY 2022; 2:822606. [PMID: 36909847 PMCID: PMC10002468 DOI: 10.3389/fsysb.2022.822606] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
During the COVID-19 pandemic, mathematical modeling of disease transmission has become a cornerstone of key state decisions. To advance the state-of-the-art host viral modeling to handle future pandemics, many scientists working on related issues assembled to discuss the topics. These discussions exposed the reproducibility crisis that leads to inability to reuse and integrate models. This document summarizes these discussions, presents difficulties, and mentions existing efforts towards future solutions that will allow future model utility and integration. We argue that without addressing these challenges, scientists will have diminished ability to build, disseminate, and implement high-impact multi-scale modeling that is needed to understand the health crises we face.
Collapse
Affiliation(s)
- Jonathan Karr
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rahuman S. Malik-Sheriff
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridgeshire, United Kingdom
| | - James Osborne
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC, Australia
| | | | - Eric Forgoston
- Department of Applied Mathematics and Statistics, Montclair State University, Montclair, NJ, United States
| | - Ruth Bowness
- Department of Mathematical Sciences, University of Bath, Bath, United Kingdom
| | - Yaling Liu
- Department of Mechanical Engineering and Mechanics, Department of Bioengineering, Lehigh University, Bethlehem, PA, United States
| | - Robin Thompson
- Mathematics Institute and the Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| | - Winston Garira
- Department of Mathematics and Applied Mathematics, Modelling Health and Environmental Linkages Research Group, University of Venda, Limpopo, South Africa
| | - Jacob Barhak
- Jacob Barhak Analytics, Austin, TX, United States
| | - John Rice
- Independent Retired Working Group Volunteer, Virginia Beach, VA, United States
| | - Marcella Torres
- Department of Mathematics and Computer Science, University of Richmond, Richmond, VA, United States
| | - Hana M. Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States
| | - Tingting Tang
- Department of Mathematics and Statistics in San Diego State University (SDSU) and SDSU Imperial Valley, Calexico, CA, United States
| | - William Waites
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Computer and Information Sciences, University of Strathclyde, Glasgow, Scotland
| | - James A. Glazier
- Biocomplexity Institute, Indiana University, Bloomington, IN, United States
| | - James R. Faeder
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | | |
Collapse
|
39
|
Helleckes LM, Osthege M, Wiechert W, von Lieres E, Oldiges M. Bayesian calibration, process modeling and uncertainty quantification in biotechnology. PLoS Comput Biol 2022; 18:e1009223. [PMID: 35255090 PMCID: PMC8939798 DOI: 10.1371/journal.pcbi.1009223] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 03/22/2022] [Accepted: 01/16/2022] [Indexed: 12/23/2022] Open
Abstract
High-throughput experimentation has revolutionized data-driven experimental sciences and opened the door to the application of machine learning techniques. Nevertheless, the quality of any data analysis strongly depends on the quality of the data and specifically the degree to which random effects in the experimental data-generating process are quantified and accounted for. Accordingly calibration, i.e. the quantitative association between observed quantities and measurement responses, is a core element of many workflows in experimental sciences. Particularly in life sciences, univariate calibration, often involving non-linear saturation effects, must be performed to extract quantitative information from measured data. At the same time, the estimation of uncertainty is inseparably connected to quantitative experimentation. Adequate calibration models that describe not only the input/output relationship in a measurement system but also its inherent measurement noise are required. Due to its mathematical nature, statistically robust calibration modeling remains a challenge for many practitioners, at the same time being extremely beneficial for machine learning applications. In this work, we present a bottom-up conceptual and computational approach that solves many problems of understanding and implementing non-linear, empirical calibration modeling for quantification of analytes and process modeling. The methodology is first applied to the optical measurement of biomass concentrations in a high-throughput cultivation system, then to the quantification of glucose by an automated enzymatic assay. We implemented the conceptual framework in two Python packages, calibr8 and murefi, with which we demonstrate how to make uncertainty quantification for various calibration tasks more accessible. Our software packages enable more reproducible and automatable data analysis routines compared to commonly observed workflows in life sciences. Subsequently, we combine the previously established calibration models with a hierarchical Monod-like ordinary differential equation model of microbial growth to describe multiple replicates of Corynebacterium glutamicum batch cultures. Key process model parameters are learned by both maximum likelihood estimation and Bayesian inference, highlighting the flexibility of the statistical and computational framework.
Collapse
Affiliation(s)
- Laura Marie Helleckes
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
- Institute of Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Michael Osthege
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
- Institute of Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Wolfgang Wiechert
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
- Computational Systems Biotechnology (AVT.CSB), RWTH Aachen University, Aachen, Germany
| | - Eric von Lieres
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Marco Oldiges
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
- Institute of Biotechnology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
40
|
Villaverde AF, Pathirana D, Fröhlich F, Hasenauer J, Banga JR. A protocol for dynamic model calibration. Brief Bioinform 2022; 23:bbab387. [PMID: 34619769 PMCID: PMC8769694 DOI: 10.1093/bib/bbab387] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/06/2021] [Accepted: 08/29/2021] [Indexed: 12/23/2022] Open
Abstract
Ordinary differential equation models are nowadays widely used for the mechanistic description of biological processes and their temporal evolution. These models typically have many unknown and nonmeasurable parameters, which have to be determined by fitting the model to experimental data. In order to perform this task, known as parameter estimation or model calibration, the modeller faces challenges such as poor parameter identifiability, lack of sufficiently informative experimental data and the existence of local minima in the objective function landscape. These issues tend to worsen with larger model sizes, increasing the computational complexity and the number of unknown parameters. An incorrectly calibrated model is problematic because it may result in inaccurate predictions and misleading conclusions. For nonexpert users, there are a large number of potential pitfalls. Here, we provide a protocol that guides the user through all the steps involved in the calibration of dynamic models. We illustrate the methodology with two models and provide all the code required to reproduce the results and perform the same analysis on new models. Our protocol provides practitioners and researchers in biological modelling with a one-stop guide that is at the same time compact and sufficiently comprehensive to cover all aspects of the problem.
Collapse
Affiliation(s)
- Alejandro F Villaverde
- Universidade de Vigo, Department of Systems Engineering & Control, Vigo 36310, Galicia, Spain
| | - Dilan Pathirana
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn 53115, Germany
| | - Fabian Fröhlich
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Jan Hasenauer
- Center for Mathematics, Technische Universität München, Garching 85748, Germany
- Harvard Medical School, Cambridge, MA 02115, USA
| | - Julio R Banga
- Bioprocess Engineering Group, IIM-CSIC, Vigo 36208, Galicia, Spain
| |
Collapse
|
41
|
Stapor P, Schmiester L, Wierling C, Merkt S, Pathirana D, Lange BMH, Weindl D, Hasenauer J. Mini-batch optimization enables training of ODE models on large-scale datasets. Nat Commun 2022; 13:34. [PMID: 35013141 PMCID: PMC8748893 DOI: 10.1038/s41467-021-27374-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 11/11/2021] [Indexed: 11/09/2022] Open
Abstract
Quantitative dynamic models are widely used to study cellular signal processing. A critical step in modelling is the estimation of unknown model parameters from experimental data. As model sizes and datasets are steadily growing, established parameter optimization approaches for mechanistic models become computationally extremely challenging. Mini-batch optimization methods, as employed in deep learning, have better scaling properties. In this work, we adapt, apply, and benchmark mini-batch optimization for ordinary differential equation (ODE) models, thereby establishing a direct link between dynamic modelling and machine learning. On our main application example, a large-scale model of cancer signaling, we benchmark mini-batch optimization against established methods, achieving better optimization results and reducing computation by more than an order of magnitude. We expect that our work will serve as a first step towards mini-batch optimization tailored to ODE models and enable modelling of even larger and more complex systems than what is currently possible.
Collapse
Affiliation(s)
- Paul Stapor
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, 85764, Neuherberg, Germany
- Technische Universität München, Center for Mathematics, Chair of Mathematical Modeling of Biological Systems, 85748, Garching, Germany
| | - Leonard Schmiester
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, 85764, Neuherberg, Germany
- Technische Universität München, Center for Mathematics, Chair of Mathematical Modeling of Biological Systems, 85748, Garching, Germany
| | | | - Simon Merkt
- Universität Bonn, Faculty of Mathematics and Natural Sciences, 53115, Bonn, Germany
| | - Dilan Pathirana
- Universität Bonn, Faculty of Mathematics and Natural Sciences, 53115, Bonn, Germany
| | | | - Daniel Weindl
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, 85764, Neuherberg, Germany
| | - Jan Hasenauer
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, 85764, Neuherberg, Germany.
- Technische Universität München, Center for Mathematics, Chair of Mathematical Modeling of Biological Systems, 85748, Garching, Germany.
- Universität Bonn, Faculty of Mathematics and Natural Sciences, 53115, Bonn, Germany.
| |
Collapse
|
42
|
Schmiester L, Weindl D, Hasenauer J. Efficient gradient-based parameter estimation for dynamic models using qualitative data. BIOINFORMATICS (OXFORD, ENGLAND) 2021. [PMID: 34260697 DOI: 10.1101/2021.02.06.430039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
MOTIVATION Unknown parameters of dynamical models are commonly estimated from experimental data. However, while various efficient optimization and uncertainty analysis methods have been proposed for quantitative data, methods for qualitative data are rare and suffer from bad scaling and convergence. RESULTS Here, we propose an efficient and reliable framework for estimating the parameters of ordinary differential equation models from qualitative data. In this framework, we derive a semi-analytical algorithm for gradient calculation of the optimal scaling method developed for qualitative data. This enables the use of efficient gradient-based optimization algorithms. We demonstrate that the use of gradient information improves performance of optimization and uncertainty quantification on several application examples. On average, we achieve a speedup of more than one order of magnitude compared to gradient-free optimization. In addition, in some examples, the gradient-based approach yields substantially improved objective function values and quality of the fits. Accordingly, the proposed framework substantially improves the parameterization of models from qualitative data. AVAILABILITY AND IMPLEMENTATION The proposed approach is implemented in the open-source Python Parameter EStimation TOolbox (pyPESTO). pyPESTO is available at https://github.com/ICB-DCM/pyPESTO. All application examples and code to reproduce this study are available at https://doi.org/10.5281/zenodo.4507613. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Leonard Schmiester
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
- Center for Mathematics, Technische Universität München, Garching 85748, Germany
| | - Daniel Weindl
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Jan Hasenauer
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
- Center for Mathematics, Technische Universität München, Garching 85748, Germany
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn 53113, Germany
| |
Collapse
|
43
|
Schmiester L, Weindl D, Hasenauer J. Efficient gradient-based parameter estimation for dynamic models using qualitative data. Bioinformatics 2021; 37:4493-4500. [PMID: 34260697 PMCID: PMC8652033 DOI: 10.1093/bioinformatics/btab512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 11/22/2022] Open
Abstract
Motivation Unknown parameters of dynamical models are commonly estimated from experimental data. However, while various efficient optimization and uncertainty analysis methods have been proposed for quantitative data, methods for qualitative data are rare and suffer from bad scaling and convergence. Results Here, we propose an efficient and reliable framework for estimating the parameters of ordinary differential equation models from qualitative data. In this framework, we derive a semi-analytical algorithm for gradient calculation of the optimal scaling method developed for qualitative data. This enables the use of efficient gradient-based optimization algorithms. We demonstrate that the use of gradient information improves performance of optimization and uncertainty quantification on several application examples. On average, we achieve a speedup of more than one order of magnitude compared to gradient-free optimization. In addition, in some examples, the gradient-based approach yields substantially improved objective function values and quality of the fits. Accordingly, the proposed framework substantially improves the parameterization of models from qualitative data. Availability and implementation The proposed approach is implemented in the open-source Python Parameter EStimation TOolbox (pyPESTO). pyPESTO is available at https://github.com/ICB-DCM/pyPESTO. All application examples and code to reproduce this study are available at https://doi.org/10.5281/zenodo.4507613. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Leonard Schmiester
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, 85764, Germany.,Center for Mathematics, Technische Universität München, Garching, 85748, Germany
| | - Daniel Weindl
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Jan Hasenauer
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, 85764, Germany.,Center for Mathematics, Technische Universität München, Garching, 85748, Germany.,Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, 53113, Germany
| |
Collapse
|
44
|
Fröhlich F, Weindl D, Schälte Y, Pathirana D, Paszkowski Ł, Lines GT, Stapor P, Hasenauer J. AMICI: High-Performance Sensitivity Analysis for Large Ordinary Differential Equation Models. Bioinformatics 2021; 37:3676-3677. [PMID: 33821950 PMCID: PMC8545331 DOI: 10.1093/bioinformatics/btab227] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 11/14/2022] Open
Abstract
Summary Ordinary differential equation models facilitate the understanding of cellular signal transduction and other biological processes. However, for large and comprehensive models, the computational cost of simulating or calibrating can be limiting. AMICI is a modular toolbox implemented in C++/Python/MATLAB that provides efficient simulation and sensitivity analysis routines tailored for scalable, gradient-based parameter estimation and uncertainty quantification. Availabilityand implementation AMICI is published under the permissive BSD-3-Clause license with source code publicly available on https://github.com/AMICI-dev/AMICI. Citeable releases are archived on Zenodo. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Fabian Fröhlich
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Weindl
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Yannik Schälte
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany.,Center for Mathematics, Technische Universität München, 85748 Garching, Germany
| | - Dilan Pathirana
- Faculty of Mathematics and Natural Sciences, University of Bonn, 53113 Bonn, Germany
| | | | | | - Paul Stapor
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany.,Center for Mathematics, Technische Universität München, 85748 Garching, Germany
| | - Jan Hasenauer
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany.,Center for Mathematics, Technische Universität München, 85748 Garching, Germany.,Faculty of Mathematics and Natural Sciences, University of Bonn, 53113 Bonn, Germany
| |
Collapse
|
45
|
Raimúndez E, Dudkin E, Vanhoefer J, Alamoudi E, Merkt S, Fuhrmann L, Bai F, Hasenauer J. COVID-19 outbreak in Wuhan demonstrates the limitations of publicly available case numbers for epidemiological modeling. Epidemics 2021; 34:100439. [PMID: 33556763 PMCID: PMC7845523 DOI: 10.1016/j.epidem.2021.100439] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 01/12/2023] Open
Abstract
Epidemiological models are widely used to analyze the spread of diseases such as the global COVID-19 pandemic caused by SARS-CoV-2. However, all models are based on simplifying assumptions and often on sparse data. This limits the reliability of parameter estimates and predictions. In this manuscript, we demonstrate the relevance of these limitations and the pitfalls associated with the use of overly simplistic models. We considered the data for the early phase of the COVID-19 outbreak in Wuhan, China, as an example, and perform parameter estimation, uncertainty analysis and model selection for a range of established epidemiological models. Amongst others, we employ Markov chain Monte Carlo sampling, parameter and prediction profile calculation algorithms. Our results show that parameter estimates and predictions obtained for several established models on the basis of reported case numbers can be subject to substantial uncertainty. More importantly, estimates were often unrealistic and the confidence/credibility intervals did not cover plausible values of critical parameters obtained using different approaches. These findings suggest, amongst others, that standard compartmental models can be overly simplistic and that the reported case numbers provide often insufficient information for obtaining reliable and realistic parameter values, and for forecasting the evolution of epidemics.
Collapse
Affiliation(s)
- Elba Raimúndez
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany; Technische Universität München, Center for Mathematics, Garching, Germany
| | - Erika Dudkin
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany
| | - Jakob Vanhoefer
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany
| | - Emad Alamoudi
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany
| | - Simon Merkt
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany
| | - Lara Fuhrmann
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany
| | - Fan Bai
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany
| | - Jan Hasenauer
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany; Technische Universität München, Center for Mathematics, Garching, Germany; Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany.
| |
Collapse
|
46
|
Schmiester L, Weindl D, Hasenauer J. Parameterization of mechanistic models from qualitative data using an efficient optimal scaling approach. J Math Biol 2020; 81:603-623. [PMID: 32696085 PMCID: PMC7427713 DOI: 10.1007/s00285-020-01522-w] [Citation(s) in RCA: 6] [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: 12/10/2019] [Revised: 05/28/2020] [Indexed: 12/21/2022]
Abstract
Quantitative dynamical models facilitate the understanding of biological processes and the prediction of their dynamics. These models usually comprise unknown parameters, which have to be inferred from experimental data. For quantitative experimental data, there are several methods and software tools available. However, for qualitative data the available approaches are limited and computationally demanding. Here, we consider the optimal scaling method which has been developed in statistics for categorical data and has been applied to dynamical systems. This approach turns qualitative variables into quantitative ones, accounting for constraints on their relation. We derive a reduced formulation for the optimization problem defining the optimal scaling. The reduced formulation possesses the same optimal points as the established formulation but requires less degrees of freedom. Parameter estimation for dynamical models of cellular pathways revealed that the reduced formulation improves the robustness and convergence of optimizers. This resulted in substantially reduced computation times. We implemented the proposed approach in the open-source Python Parameter EStimation TOolbox (pyPESTO) to facilitate reuse and extension. The proposed approach enables efficient parameterization of quantitative dynamical models using qualitative data.
Collapse
Affiliation(s)
- Leonard Schmiester
- Institute of Computational Biology, Helmholtz Zentrum München–German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Center for Mathematics, Technische Universität München, 85748 Garching, Germany
| | - Daniel Weindl
- Institute of Computational Biology, Helmholtz Zentrum München–German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Jan Hasenauer
- Institute of Computational Biology, Helmholtz Zentrum München–German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Center for Mathematics, Technische Universität München, 85748 Garching, Germany
- Faculty of Mathematics and Natural Sciences, University of Bonn, 53113 Bonn, Germany
| |
Collapse
|