1
|
Osborne JM. An adaptive numerical method for multi-cellular simulations of tissue development and maintenance. J Theor Biol 2024; 594:111922. [PMID: 39111542 DOI: 10.1016/j.jtbi.2024.111922] [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: 01/17/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024]
Abstract
In recent years, multi-cellular models, where cells are represented as individual interacting entities, are becoming ever popular. This has led to a proliferation of novel methods and simulation tools. The first aim of this paper is to review the numerical methods utilised by multi-cellular modelling tools and to demonstrate which numerical methods are appropriate for simulations of tissue and organ development, maintenance, and disease. The second aim is to introduce an adaptive time-stepping algorithm and to demonstrate it's efficiency and accuracy. We focus on off-lattice, mechanics based, models where cell movement is defined by a series of first order ordinary differential equations, derived by assuming over-damped motion and balancing forces. We see that many numerical methods have been used, ranging from simple Forward Euler approaches through to higher order single-step methods like Runge-Kutta 4 and multi-step methods like Adams-Bashforth 2. Through a series of exemplar multi-cellular simulations, we see that if: care is taken to have events (births deaths and re-meshing/re-arrangements) occur on common time-steps; and boundaries are imposed on all sub-steps of numerical methods or implemented using forces, then all numerical methods can converge with the correct order. We introduce an adaptive time-stepping method and demonstrate that the best compromise between L∞ error and run-time is to use Runge-Kutta 4 with an increased time-step and moderate adaptivity. We see that a judicious choice of numerical method can speed the simulation up by a factor of 10-60 from the Forward Euler methods seen in Osborne et al. (2017), and a further speed up by a factor of 4 can be achieved by using an adaptive time-step.
Collapse
Affiliation(s)
- James M Osborne
- School of Mathematics and Statistics, University of Melbourne, Melbourne, 3010, Victoria, Australia.
| |
Collapse
|
2
|
Kemkar S, Tao M, Ghosh A, Stamatakos G, Graf N, Poorey K, Balakrishnan U, Trask N, Radhakrishnan R. Towards verifiable cancer digital twins: tissue level modeling protocol for precision medicine. Front Physiol 2024; 15:1473125. [PMID: 39507514 PMCID: PMC11537925 DOI: 10.3389/fphys.2024.1473125] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Cancer exhibits substantial heterogeneity, manifesting as distinct morphological and molecular variations across tumors, which frequently undermines the efficacy of conventional oncological treatments. Developments in multiomics and sequencing technologies have paved the way for unraveling this heterogeneity. Nevertheless, the complexity of the data gathered from these methods cannot be fully interpreted through multimodal data analysis alone. Mathematical modeling plays a crucial role in delineating the underlying mechanisms to explain sources of heterogeneity using patient-specific data. Intra-tumoral diversity necessitates the development of precision oncology therapies utilizing multiphysics, multiscale mathematical models for cancer. This review discusses recent advancements in computational methodologies for precision oncology, highlighting the potential of cancer digital twins to enhance patient-specific decision-making in clinical settings. We review computational efforts in building patient-informed cellular and tissue-level models for cancer and propose a computational framework that utilizes agent-based modeling as an effective conduit to integrate cancer systems models that encode signaling at the cellular scale with digital twin models that predict tissue-level response in a tumor microenvironment customized to patient information. Furthermore, we discuss machine learning approaches to building surrogates for these complex mathematical models. These surrogates can potentially be used to conduct sensitivity analysis, verification, validation, and uncertainty quantification, which is especially important for tumor studies due to their dynamic nature.
Collapse
Affiliation(s)
- Sharvari Kemkar
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Mengdi Tao
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Alokendra Ghosh
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Georgios Stamatakos
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Zografos, Greece
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Kunal Poorey
- Department of Systems Biology, Sandia National Laboratories, Livermore, CA, United States
| | - Uma Balakrishnan
- Department of Quant Modeling and SW Eng, Sandia National Laboratories, Livermore, CA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Nathaniel Trask
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA, United States
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
3
|
Krix S, Wilczynski E, Falgàs N, Sánchez-Valle R, Yoles E, Nevo U, Baruch K, Fröhlich H. Towards early diagnosis of Alzheimer's disease: advances in immune-related blood biomarkers and computational approaches. Front Immunol 2024; 15:1343900. [PMID: 38720902 PMCID: PMC11078023 DOI: 10.3389/fimmu.2024.1343900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
Alzheimer's disease has an increasing prevalence in the population world-wide, yet current diagnostic methods based on recommended biomarkers are only available in specialized clinics. Due to these circumstances, Alzheimer's disease is usually diagnosed late, which contrasts with the currently available treatment options that are only effective for patients at an early stage. Blood-based biomarkers could fill in the gap of easily accessible and low-cost methods for early diagnosis of the disease. In particular, immune-based blood-biomarkers might be a promising option, given the recently discovered cross-talk of immune cells of the central nervous system with those in the peripheral immune system. Here, we give a background on recent advances in research on brain-immune system cross-talk in Alzheimer's disease and review machine learning approaches, which can combine multiple biomarkers with further information (e.g. age, sex, APOE genotype) into predictive models supporting an earlier diagnosis. In addition, mechanistic modeling approaches, such as agent-based modeling open the possibility to model and analyze cell dynamics over time. This review aims to provide an overview of the current state of immune-system related blood-based biomarkers and their potential for the early diagnosis of Alzheimer's disease.
Collapse
Affiliation(s)
- Sophia Krix
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for Information Technology (b-it), University of Bonn, Bonn, Germany
| | - Ella Wilczynski
- Department of Biomedical Engineering, The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Neus Falgàs
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FCRB-IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FCRB-IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Eti Yoles
- ImmunoBrain Checkpoint Ltd., Rechovot, Israel
| | - Uri Nevo
- Department of Biomedical Engineering, The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Kuti Baruch
- ImmunoBrain Checkpoint Ltd., Rechovot, Israel
| | - Holger Fröhlich
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for Information Technology (b-it), University of Bonn, Bonn, Germany
| |
Collapse
|
4
|
Thibault Greugny E, Bensaci J, Fages F, Stamatas GN. Computational modelling predicts impaired barrier function and higher sensitivity to skin inflammation following pH elevation. Exp Dermatol 2023; 32:177-185. [PMID: 36321871 DOI: 10.1111/exd.14698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/04/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
Abstract
Skin surface pH has been identified as a key regulator of the epidermal homeostasis through its action on serine protease activity. These enzymes, like kallikreins (KLK), are responsible for the degradation of corneodesmosomes, the protein structures linking together corneocytes, and are regulated by Lympho-Epithelial Kazal-Type-related Inhibitor (LEKTI). KLK activity increases at pH levels higher than physiological. An increase in skin surface pH has been observed in patients suffering from skin diseases characterized by impaired barrier function, like atopic dermatitis. In this work, we introduce an agent-based model of the epidermis to study the impact of a change in skin surface pH on the structural and physiological properties of the epidermis, through the LEKTI-KLK mechanism. We demonstrate that a less acidic pH, compared to the slightly acidic pH observed in healthy skin, is sufficient to significantly affect the water loss at the surface and the amount of irritant permeating through the epidermis. This weakening of the skin barrier function eventually results in a more intense skin inflammation following exposure to an external irritant. This work provides additional evidence that skin surface pH and serine proteases can be therapeutic targets to improve skin barrier integrity.
Collapse
Affiliation(s)
- Eléa Thibault Greugny
- Essential Health Translational Science, Johnson & Johnson Santé Beauté France, Issy-les-Moulineaux, France.,Inria Saclay Île-de-France, Lifeware Team, Palaiseau, France
| | - Jalil Bensaci
- Essential Health Translational Science, Johnson & Johnson Santé Beauté France, Issy-les-Moulineaux, France
| | - François Fages
- Inria Saclay Île-de-France, Lifeware Team, Palaiseau, France
| | - Georgios N Stamatas
- Essential Health Translational Science, Johnson & Johnson Santé Beauté France, Issy-les-Moulineaux, France
| |
Collapse
|
5
|
Novel Ground-Up 3D Multicellular Simulators for Synthetic Biology CAD Integrating Stochastic Gillespie Simulations Benchmarked with Topologically Variable SBML Models. Genes (Basel) 2023; 14:genes14010154. [PMID: 36672895 PMCID: PMC9859520 DOI: 10.3390/genes14010154] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023] Open
Abstract
The elevation of Synthetic Biology from single cells to multicellular simulations would be a significant scale-up. The spatiotemporal behavior of cellular populations has the potential to be prototyped in silico for computer assisted design through ergonomic interfaces. Such a platform would have great practical potential across medicine, industry, research, education and accessible archiving in bioinformatics. Existing Synthetic Biology CAD systems are considered limited regarding population level behavior, and this work explored the in silico challenges posed from biological and computational perspectives. Retaining the connection to Synthetic Biology CAD, an extension of the Infobiotics Workbench Suite was considered, with potential for the integration of genetic regulatory models and/or chemical reaction networks through Next Generation Stochastic Simulator (NGSS) Gillespie algorithms. These were executed using SBML models generated by in-house SBML-Constructor over numerous topologies and benchmarked in association with multicellular simulation layers. Regarding multicellularity, two ground-up multicellular solutions were developed, including the use of Unreal Engine 4 contrasted with CPU multithreading and Blender visualization, resulting in a comparison of real-time versus batch-processed simulations. In conclusion, high-performance computing and client-server architectures could be considered for future works, along with the inclusion of numerous biologically and physically informed features, whilst still pursuing ergonomic solutions.
Collapse
|
6
|
Mathias S, Coulier A, Hellander A. CBMOS: a GPU-enabled Python framework for the numerical study of center-based models. BMC Bioinformatics 2022; 23:55. [PMID: 35100968 PMCID: PMC8805507 DOI: 10.1186/s12859-022-04575-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/11/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Cell-based models are becoming increasingly popular for applications in developmental biology. However, the impact of numerical choices on the accuracy and efficiency of the simulation of these models is rarely meticulously tested. Without concrete studies to differentiate between solid model conclusions and numerical artifacts, modelers are at risk of being misled by their experiments' results. Most cell-based modeling frameworks offer a feature-rich environment, providing a wide range of biological components, but are less suitable for numerical studies. There is thus a need for software specifically targeted at this use case. RESULTS We present CBMOS, a Python framework for the simulation of the center-based or cell-centered model. Contrary to other implementations, CBMOS' focus is on facilitating numerical study of center-based models by providing access to multiple ordinary differential equation solvers and force functions through a flexible, user-friendly interface and by enabling rapid testing through graphics processing unit (GPU) acceleration. We show-case its potential by illustrating two common workflows: (1) comparison of the numerical properties of two solvers within a Jupyter notebook and (2) measuring average wall times of both solvers on a high performance computing cluster. More specifically, we confirm that although for moderate accuracy levels the backward Euler method allows for larger time step sizes than the commonly used forward Euler method, its additional computational cost due to being an implicit method prohibits its use for practical test cases. CONCLUSIONS CBMOS is a flexible, easy-to-use Python implementation of the center-based model, exposing both basic model assumptions and numerical components to the user. It is available on GitHub and PyPI under an MIT license. CBMOS allows for fast prototyping on a central processing unit for small systems through the use of NumPy. Using CuPy on a GPU, cell populations of up to 10,000 cells can be simulated within a few seconds. As such, it will substantially lower the time investment for any modeler to check the crucial assumption that model conclusions are independent of numerical issues.
Collapse
Affiliation(s)
- Sonja Mathias
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Adrien Coulier
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Andreas Hellander
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
7
|
Fletcher AG, Osborne JM. Seven challenges in the multiscale modeling of multicellular tissues. WIREs Mech Dis 2022; 14:e1527. [PMID: 35023326 PMCID: PMC11478939 DOI: 10.1002/wsbm.1527] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/23/2020] [Accepted: 03/25/2021] [Indexed: 11/11/2022]
Abstract
The growth and dynamics of multicellular tissues involve tightly regulated and coordinated morphogenetic cell behaviors, such as shape changes, movement, and division, which are governed by subcellular machinery and involve coupling through short- and long-range signals. A key challenge in the fields of developmental biology, tissue engineering and regenerative medicine is to understand how relationships between scales produce emergent tissue-scale behaviors. Recent advances in molecular biology, live-imaging and ex vivo techniques have revolutionized our ability to study these processes experimentally. To fully leverage these techniques and obtain a more comprehensive understanding of the causal relationships underlying tissue dynamics, computational modeling approaches are increasingly spanning multiple spatial and temporal scales, and are coupling cell shape, growth, mechanics, and signaling. Yet such models remain challenging: modeling at each scale requires different areas of technical skills, while integration across scales necessitates the solution to novel mathematical and computational problems. This review aims to summarize recent progress in multiscale modeling of multicellular tissues and to highlight ongoing challenges associated with the construction, implementation, interrogation, and validation of such models. This article is categorized under: Reproductive System Diseases > Computational Models Metabolic Diseases > Computational Models Cancer > Computational Models.
Collapse
Affiliation(s)
- Alexander G. Fletcher
- School of Mathematics and StatisticsUniversity of SheffieldSheffieldUK
- Bateson CentreUniversity of SheffieldSheffieldUK
| | - James M. Osborne
- School of Mathematics and StatisticsUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
8
|
Aghaallaei N, Dick AM, Tsingos E, Inoue D, Hasel E, Thumberger T, Toyoda A, Leptin M, Wittbrodt J, Bajoghli B. αβ/γδ T cell lineage outcome is regulated by intrathymic cell localization and environmental signals. SCIENCE ADVANCES 2021; 7:7/29/eabg3613. [PMID: 34261656 PMCID: PMC8279519 DOI: 10.1126/sciadv.abg3613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/28/2021] [Indexed: 05/07/2023]
Abstract
αβ and γδ T cells are two distinct sublineages that develop in the vertebrate thymus. Thus far, their differentiation from a common progenitor is mostly understood to be regulated by intrinsic mechanisms. However, the proportion of αβ/γδ T cells varies in different vertebrate taxa. How this process is regulated in species that tend to produce a high frequency of γδ T cells is unstudied. Using an in vivo teleost model, the medaka, we report that progenitors first enter a thymic niche where their development into γδ T cells is favored. Translocation from this niche, mediated by chemokine receptor Ccr9b, is a prerequisite for their differentiation into αβ T cells. On the other hand, the thymic niche also generates opposing gradients of the cytokine interleukin-7 and chemokine Ccl25a, and, together, they influence the lineage outcome. We propose a previously unknown mechanism that determines the proportion of αβ/γδ lineages within species.
Collapse
Affiliation(s)
- Narges Aghaallaei
- Department of Hematology, Oncology, Immunology, and Rheumatology, University Hospital of Tübingen, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Advaita M Dick
- Department of Hematology, Oncology, Immunology, and Rheumatology, University Hospital of Tübingen, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Erika Tsingos
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Daigo Inoue
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Eva Hasel
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Thomas Thumberger
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Atsushi Toyoda
- Comparative Genomics Laboratory, Department of Genomics and Evolutionary Biology, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan
| | - Maria Leptin
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
- EMBO, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Joachim Wittbrodt
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Baubak Bajoghli
- Department of Hematology, Oncology, Immunology, and Rheumatology, University Hospital of Tübingen, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany.
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| |
Collapse
|
9
|
Stamatas GN, Bensaci J, Greugny E, Kaur S, Wang H, Dizon MV, Cork MJ, Friedman AJ, Oddos T. A Predictive Self-Organizing Multicellular Computational Model of Infant Skin Permeability to Topically Applied Substances. J Invest Dermatol 2021; 141:2049-2055.e1. [PMID: 33705796 DOI: 10.1016/j.jid.2021.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 11/16/2022]
Abstract
Computational models of skin permeability are typically based on assumptions of fixed geometry and homogeneity of the whole epidermis or of epidermal strata and are often limited to adult skin. Infant skin differs quantitatively from that of the adult in its structure and its functional properties, including its barrier function to permeation. To address this problem, we developed a self-organizing multicellular epidermis model of barrier formation with realistic cell morphology. By modulating the parameters relating to cell turnover reflecting those in adult or infant epidermis, we were able to generate accordingly two distinct models. Emerging properties of these models reflect the corresponding experimentally measured values of epidermal and stratum corneum thickness. Diffusion of an externally applied substance (e.g., caffeine) was simulated by a molecular exchange between the model agents, defined by the individual cells and their surrounding extracellular space. By adjusting the surface concentration and the intercellular exchange rate, the model can recapitulate experimental permeability data after topical exposure. By applying these parameters to an infant model, we were able to predict the caffeine concentration profile in infant skin, closely matching experimental results. This work paves the way for a better understanding of skin physiology and function during the first years of life.
Collapse
Affiliation(s)
- Georgios N Stamatas
- Essential Health R&D, Johnson & Johnson Santé Beauté France, Issy-les-Moulineaux, France.
| | - Jalil Bensaci
- Essential Health R&D, Johnson & Johnson Santé Beauté France, Issy-les-Moulineaux, France
| | - Elea Greugny
- Essential Health R&D, Johnson & Johnson Santé Beauté France, Issy-les-Moulineaux, France
| | - Simarna Kaur
- Skin Health R&D, Johnson & Johnson Consumer Inc, Skillman, New Jersey, USA
| | - Hequn Wang
- Skin Health R&D, Johnson & Johnson Consumer Inc, Skillman, New Jersey, USA
| | - Maria Victoria Dizon
- Department of Dermatology, Faculty of Medicine and Surgery, University of Santo Tomas, Manila, The Philippines
| | - Michael J Cork
- The Academic Unit of Dermatology Research, Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Adam J Friedman
- The Department of Dermatology, George Washington School of Medicine & Health Sciences, Washington, District of Columbia, USA
| | - Thierry Oddos
- Essential Health R&D, Johnson & Johnson Santé Beauté France, Issy-les-Moulineaux, France
| |
Collapse
|
10
|
Mathias S, Coulier A, Bouchnita A, Hellander A. Impact of Force Function Formulations on the Numerical Simulation of Centre-Based Models. Bull Math Biol 2020; 82:132. [PMID: 33025278 PMCID: PMC7538447 DOI: 10.1007/s11538-020-00810-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Centre-based or cell-centre models are a framework for the computational study of multicellular systems with widespread use in cancer modelling and computational developmental biology. At the core of these models are the numerical method used to update cell positions and the force functions that encode the pairwise mechanical interactions of cells. For the latter, there are multiple choices that could potentially affect both the biological behaviour captured, and the robustness and efficiency of simulation. For example, available open-source software implementations of centre-based models rely on different force functions for their default behaviour and it is not straightforward for a modeller to know if these are interchangeable. Our study addresses this problem and contributes to the understanding of the potential and limitations of three popular force functions from a numerical perspective. We show empirically that choosing the force parameters such that the relaxation time for two cells after cell division is consistent between different force functions results in good agreement of the population radius of a two-dimensional monolayer relaxing mechanically after intense cell proliferation. Furthermore, we report that numerical stability is not sufficient to prevent unphysical cell trajectories following cell division, and consequently, that too large time steps can cause geometrical differences at the population level.
Collapse
Affiliation(s)
- Sonja Mathias
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Adrien Coulier
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Anass Bouchnita
- Department of Information Technology, Uppsala University, Uppsala, Sweden
- Present Address: Ecole Centrale Casablanca, Bouskoura, Morocco
| | - Andreas Hellander
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
11
|
Cooper FR, Baker RE, Bernabeu MO, Bordas R, Bowler L, Bueno-Orovio A, Byrne HM, Carapella V, Cardone-Noott L, Jonatha C, Dutta S, Evans BD, Fletcher AG, Grogan JA, Guo W, Harvey DG, Hendrix M, Kay D, Kursawe J, Maini PK, McMillan B, Mirams GR, Osborne JM, Pathmanathan P, Pitt-Francis JM, Robinson M, Rodriguez B, Spiteri RJ, Gavaghan DJ. Chaste: Cancer, Heart and Soft Tissue Environment. JOURNAL OF OPEN SOURCE SOFTWARE 2020; 5:1848. [PMID: 37192932 PMCID: PMC7614534 DOI: 10.21105/joss.01848] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Chaste (Cancer, Heart And Soft Tissue Environment) is an open source simulation package for the numerical solution of mathematical models arising in physiology and biology. To date, Chaste development has been driven primarily by applications that include continuum modelling of cardiac electrophysiology ('Cardiac Chaste'), discrete cell-based modelling of soft tissues ('Cell-based Chaste'), and modelling of ventilation in lungs ('Lung Chaste'). Cardiac Chaste addresses the need for a high-performance, generic, and verified simulation framework for cardiac electrophysiology that is freely available to the scientific community. Cardiac chaste provides a software package capable of realistic heart simulations that is efficient, rigorously tested, and runs on HPC platforms. Cell-based Chaste addresses the need for efficient and verified implementations of cell-based modelling frameworks, providing a set of extensible tools for simulating biological tissues. Computational modelling, along with live imaging techniques, plays an important role in understanding the processes of tissue growth and repair. A wide range of cell-based modelling frameworks have been developed that have each been successfully applied in a range of biological applications. Cell-based Chaste includes implementations of the cellular automaton model, the cellular Potts model, cell-centre models with cell representations as overlapping spheres or Voronoi tessellations, and the vertex model. Lung Chaste addresses the need for a novel, generic and efficient lung modelling software package that is both tested and verified. It aims to couple biophysically-detailed models of airway mechanics with organ-scale ventilation models in a package that is freely available to the scientific community. Chaste is designed to be modular and extensible, providing libraries for common scientific computing infrastructure such as linear algebra operations, finite element meshes, and ordinary and partial differential equation solvers. This infrastructure is used by libraries for specific applications, such as continuum mechanics, cardiac models, and cell-based models. The software engineering techniques used to develop Chaste are intended to ensure code quality, re-usability and reliability. Primary applications of the software include cardiac and respiratory physiology, cancer and developmental biology.
Collapse
Affiliation(s)
- Fergus R Cooper
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Ruth E Baker
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Miguel O Bernabeu
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rafel Bordas
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Louise Bowler
- Department of Computer Science, University of Oxford, Oxford, UK
| | | | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Valentina Carapella
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | - Cooper Jonatha
- Research IT Services, University College London, London, UK
| | - Sara Dutta
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Benjamin D Evans
- Centre for Biomedical Modelling and Analysis, Living Systems Institute, University of Exeter, Exeter, UK
- School of Psychological Science, University of Bristol, Bristol, UK
| | - Alexander G Fletcher
- School of Mathematics & Statistics, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - James A Grogan
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Wenxian Guo
- Department of Computer Science, University of Saskatchewan, Canada
| | - Daniel G Harvey
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Maurice Hendrix
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
- Digital Research Service, University of Nottingham, Nottingham, UK
| | - David Kay
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Jochen Kursawe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Beth McMillan
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - James M Osborne
- School of Mathematics and Statistics, University of Melbourne, Victoria, Australia
| | - Pras Pathmanathan
- Office of Science and Engineering Laboratories (OSEL), Center for Devices and Radiological Health (CDRH), U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | | | - Martin Robinson
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Blanca Rodriguez
- Department of Computer Science, University of Oxford, Oxford, UK
| | | | - David J Gavaghan
- Department of Computer Science, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Bravo RR, Baratchart E, West J, Schenck RO, Miller AK, Gallaher J, Gatenbee CD, Basanta D, Robertson-Tessi M, Anderson ARA. Hybrid Automata Library: A flexible platform for hybrid modeling with real-time visualization. PLoS Comput Biol 2020; 16:e1007635. [PMID: 32155140 PMCID: PMC7105119 DOI: 10.1371/journal.pcbi.1007635] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/30/2020] [Accepted: 01/06/2020] [Indexed: 12/12/2022] Open
Abstract
The Hybrid Automata Library (HAL) is a Java Library developed for use in mathematical oncology modeling. It is made of simple, efficient, generic components that can be used to model complex spatial systems. HAL's components can broadly be classified into: on- and off-lattice agent containers, finite difference diffusion fields, a GUI building system, and additional tools and utilities for computation and data collection. These components are designed to operate independently and are standardized to make them easy to interface with one another. As a demonstration of how modeling can be simplified using our approach, we have included a complete example of a hybrid model (a spatial model with interacting agent-based and PDE components). HAL is a useful asset for researchers who wish to build performant 1D, 2D and 3D hybrid models in Java, while not starting entirely from scratch. It is available on GitHub at https://github.com/MathOnco/HAL under the MIT License. HAL requires the Java JDK version 1.8 or later to compile and run the source code.
Collapse
Affiliation(s)
- Rafael R. Bravo
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Etienne Baratchart
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Jeffrey West
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Ryan O. Schenck
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna K. Miller
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Jill Gallaher
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Chandler D. Gatenbee
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - David Basanta
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Mark Robertson-Tessi
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Alexander R. A. Anderson
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida, United States of America
| |
Collapse
|
13
|
Argentati C, Tortorella I, Bazzucchi M, Morena F, Martino S. Harnessing the Potential of Stem Cells for Disease Modeling: Progress and Promises. J Pers Med 2020; 10:E8. [PMID: 32041088 PMCID: PMC7151621 DOI: 10.3390/jpm10010008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/18/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022] Open
Abstract
Ex vivo cell/tissue-based models are an essential step in the workflow of pathophysiology studies, assay development, disease modeling, drug discovery, and development of personalized therapeutic strategies. For these purposes, both scientific and pharmaceutical research have adopted ex vivo stem cell models because of their better predictive power. As matter of a fact, the advancing in isolation and in vitro expansion protocols for culturing autologous human stem cells, and the standardization of methods for generating patient-derived induced pluripotent stem cells has made feasible to generate and investigate human cellular disease models with even greater speed and efficiency. Furthermore, the potential of stem cells on generating more complex systems, such as scaffold-cell models, organoids, or organ-on-a-chip, allowed to overcome the limitations of the two-dimensional culture systems as well as to better mimic tissues structures and functions. Finally, the advent of genome-editing/gene therapy technologies had a great impact on the generation of more proficient stem cell-disease models and on establishing an effective therapeutic treatment. In this review, we discuss important breakthroughs of stem cell-based models highlighting current directions, advantages, and limitations and point out the need to combine experimental biology with computational tools able to describe complex biological systems and deliver results or predictions in the context of personalized medicine.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy
| |
Collapse
|
14
|
Dalle Nogare D, Chitnis AB. NetLogo agent-based models as tools for understanding the self-organization of cell fate, morphogenesis and collective migration of the zebrafish posterior Lateral Line primordium. Semin Cell Dev Biol 2019; 100:186-198. [PMID: 31901312 DOI: 10.1016/j.semcdb.2019.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 01/25/2023]
Abstract
Interactions between primordium cells and their environment determines the self-organization of the zebrafish posterior Lateral Line primordium as it migrates under the skin from the ear to the tip of the tail forming and depositing neuromasts to spearhead formation of the posterior Lateral Line sensory system. In this review we describe how the NetLogo agent-based programming environment has been used in our lab to visualize and explore how self-generated chemokine gradients determine collective migration, how the dynamics of Wnt signaling can be used to predict patterns of neuromast deposition, and how previously defined interactions between Wnt and Fgf signaling systems have the potential to determine the periodic formation of center-biased Fgf signaling centers in the wake of a shrinking Wnt system. We also describe how NetLogo was used as a database for storing and visualizing the results of in toto lineage analysis of all cells in the migrating primordium. Together, the models illustrate how this programming environment can be used in diverse ways to integrate what has been learnt from biological experiments about the nature of interactions between cells and their environment, and explore how these interactions could potentially determine emergent patterns of cell fate specification, morphogenesis and collective migration of the zebrafish posterior Lateral Line primordium.
Collapse
Affiliation(s)
- Damian Dalle Nogare
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD USA
| | - Ajay B Chitnis
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD USA.
| |
Collapse
|
15
|
Bozorgmehr F, Hommertgen A, Krisam J, Lasitschka F, Kuon J, Maenz M, Huber PE, König L, Kieser M, Debus J, Thomas M, Rieken S. Fostering efficacy of anti-PD-1-treatment: Nivolumab plus radiotherapy in advanced non-small cell lung cancer - study protocol of the FORCE trial. BMC Cancer 2019; 19:1074. [PMID: 31703637 PMCID: PMC6842256 DOI: 10.1186/s12885-019-6205-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 09/25/2019] [Indexed: 12/02/2022] Open
Abstract
Background Hypofractionated palliative radiotherapy for metastatic lung cancer patients is frequently used in order to ease pain, to increase bone stability, to treat local mass effects, or to prolong progression-free survival at critical sites. Recently introduced, immunotherapy for patients with non-squamous non-small cell lung carcinoma (NSCLC) has significantly improved outcome in this cohort. Preclinical and early clinical data suggest that the combination of photon radiation with programmed death-1 (PD-1) targeting immunotherapies may promote a strong and durable immune response against tumor manifestations both within and beyond radiation targets. Methods/design In the present prospective, two-group, non-randomized, open-label phase II trial, 130 patients with stage IV non-squamous NSCLC in 2nd-line or 3rd-line treatment will be included. 65 patients with a clinical indication for palliative radiotherapy to non-cerebral/non-pulmonary metastatic sites will receive 240 mg nivolumab followed by palliative radiotherapy with 5 × 4 Gray (Gy) = 20 Gy photon radiation, which will be initiated within 72 h after first nivolumab administration (Group A). 65 patients without an indication for radiotherapy will only receive nivolumab (Group B). Nivolumab will be further administered every two weeks in both groups and will be continued until progression and loss of clinical benefit or until occurrence of limiting toxicities. The primary endpoint will be the objective response rate (ORR) according to response evaluation criteria in solid tumors (RECIST) 1.1. Secondary endpoints will be progression-free survival (PFS) according to RECIST 1.1, overall survival, descriptive subgroup analyses according to PD-L1 expression, toxicity and quality of life. Since response patterns following immunotherapies differ from those after conventional cytostatic agents, both objective response rate and progression-free survival will additionally be assessed according to immune-related RECIST (irRECIST) criteria. Discussion The FORCE study will prospectively investigate response rates, progression-free and overall survival (OS), and toxicity of nivolumab with and without hypofractionated palliative radiotherapy in a group of 130 patients with metastatic non-small cell lung cancer (non-squamous histology) in 2nd-line or 3rd-line treatment. This trial will contribute prospective data to the repeatedly published observation that the combination of hypofractionated photon radiotherapy and medical immunotherapy is not only safe but will also promote antitumoral immune responses. Trial registration Clinicaltrials.gov identifier: NCT03044626 (Date of initial registration: 05 January 2017). Eudra-CT Number: 2015–005741-31 (Date of initial registration: 18 December 2015).
Collapse
Affiliation(s)
- Farastuk Bozorgmehr
- Department of Thoracic Oncology, Thoraxklinik at University Hospital of Heidelberg, Röntgenstraße 1, 69126, Heidelberg, Germany.,Translational Lung Research Center Heidelberg TLRCH, Member of the German Center for Lung Research DZL, Im Neuenheimer Feld 156, 69120, Heidelberg, Germany
| | - Adriane Hommertgen
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.,German Cancer Research Center, Abteilung für Molekulare Radioonkologie, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology HIRO, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Johannes Krisam
- Institute of Medical Biometry and Informatics, University Hospital of Heidelberg, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Felix Lasitschka
- Translational Lung Research Center Heidelberg TLRCH, Member of the German Center for Lung Research DZL, Im Neuenheimer Feld 156, 69120, Heidelberg, Germany.,University Hospital of Heidelberg, Institute of Pathology, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Jonas Kuon
- Department of Thoracic Oncology, Thoraxklinik at University Hospital of Heidelberg, Röntgenstraße 1, 69126, Heidelberg, Germany
| | | | - Peter E Huber
- German Cancer Research Center, Abteilung für Molekulare Radioonkologie, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology HIRO, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Laila König
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology HIRO, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Meinhard Kieser
- Institute of Medical Biometry and Informatics, University Hospital of Heidelberg, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Juergen Debus
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.,German Cancer Research Center, Abteilung für Molekulare Radioonkologie, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology HIRO, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik at University Hospital of Heidelberg, Röntgenstraße 1, 69126, Heidelberg, Germany.,Translational Lung Research Center Heidelberg TLRCH, Member of the German Center for Lung Research DZL, Im Neuenheimer Feld 156, 69120, Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany. .,Heidelberg Institute of Radiation Oncology HIRO, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
16
|
Tsingos E, Höckendorf B, Sütterlin T, Kirchmaier S, Grabe N, Centanin L, Wittbrodt J. Retinal stem cells modulate proliferative parameters to coordinate post-embryonic morphogenesis in the eye of fish. eLife 2019; 8:42646. [PMID: 30910010 PMCID: PMC6486154 DOI: 10.7554/elife.42646] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 03/13/2019] [Indexed: 12/22/2022] Open
Abstract
Combining clonal analysis with a computational agent based model, we investigate how tissue-specific stem cells for neural retina (NR) and retinal pigmented epithelium (RPE) of the teleost medaka (Oryzias latipes) coordinate their growth rates. NR cell division timing is less variable, consistent with an upstream role as growth inducer. RPE cells divide with greater variability, consistent with a downstream role responding to inductive signals. Strikingly, the arrangement of the retinal ciliary marginal zone niche results in a spatially biased random lineage loss, where stem- and progenitor cell domains emerge spontaneously. Further, our data indicate that NR cells orient division axes to regulate organ shape and retinal topology. We highlight an unappreciated mechanism for growth coordination, where one tissue integrates cues to synchronize growth of nearby tissues. This strategy may enable evolution to modulate cell proliferation parameters in one tissue to adapt whole-organ morphogenesis in a complex vertebrate organ. By the time babies reach adulthood, they have grown many times larger than they were at birth. This development is driven by an increase in the number and size of cells in the body. In particular, special types of cells, called stem cells, act as a reservoir for tissues: they divide to create new cells that will mature into various specialized structures. The retina is the light-sensitive part of the eye. It consists of the neural retina, a tissue that contains light-detecting cells, which is supported by the retinal pigment epithelium or RPE. In fish, the RPE and neural retina are replenished by distinct groups of stem cells that do not mix, despite the tissues being close together. Unlike humans, fish grow throughout adulthood, and their eyes must then keep pace with the body. This means that the different tissues in the retina must somehow coordinate to expand at the same rate: otherwise, the retina would get wrinkled and not work properly. Tsingos et al. therefore wanted to determine how stem cells in the neural retina and RPE co-operated to produce the right number of new cells at the right time. First, stem cells in the eyes of newly hatched fish were labelled with a visible marker so that their divisions could be tracked over time to build cell family trees. This showed that stem cells behaved differently in the neural retina and the RPE. Computer simulations of the growing retina explained this behavior: stem cells in the neural retina were telling the RPE stem cells when it was time to divide. Combining results from the simulations with data from the experiments revealed that a stem cell decided to keep up dividing partly because of its position in the tissue, and partly because of random chance. To be healthy, the body needs to fine-tune the number of cells it produces: creating too few cells may make it difficult to heal after injury, but making too many could lead to diseases such as cancer. Understanding how tissues normally agree to grow together could therefore open new avenues of treatment for these conditions.
Collapse
Affiliation(s)
- Erika Tsingos
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Burkhard Höckendorf
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Thomas Sütterlin
- National Center for Tumor Diseases, Hamamatsu TIGA Center, Bioquant, Heidelberg University, Heidelberg, Germany
| | - Stephan Kirchmaier
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Niels Grabe
- National Center for Tumor Diseases, Hamamatsu TIGA Center, Bioquant, Heidelberg University, Heidelberg, Germany
| | - Lazaro Centanin
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Joachim Wittbrodt
- Centre for Organismal Studies Heidelberg, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
17
|
ya||a: GPU-Powered Spheroid Models for Mesenchyme and Epithelium. Cell Syst 2019; 8:261-266.e3. [DOI: 10.1016/j.cels.2019.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/03/2018] [Accepted: 02/19/2019] [Indexed: 11/20/2022]
|
18
|
Metzcar J, Wang Y, Heiland R, Macklin P. A Review of Cell-Based Computational Modeling in Cancer Biology. JCO Clin Cancer Inform 2019; 3:1-13. [PMID: 30715927 PMCID: PMC6584763 DOI: 10.1200/cci.18.00069] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer biology involves complex, dynamic interactions between cancer cells and their tissue microenvironments. Single-cell effects are critical drivers of clinical progression. Chemical and mechanical communication between tumor and stromal cells can co-opt normal physiologic processes to promote growth and invasion. Cancer cell heterogeneity increases cancer's ability to test strategies to adapt to microenvironmental stresses. Hypoxia and treatment can select for cancer stem cells and drive invasion and resistance. Cell-based computational models (also known as discrete models, agent-based models, or individual-based models) simulate individual cells as they interact in virtual tissues, which allows us to explore how single-cell behaviors lead to the dynamics we observe and work to control in cancer systems. In this review, we introduce the broad range of techniques available for cell-based computational modeling. The approaches can range from highly detailed models of just a few cells and their morphologies to millions of simpler cells in three-dimensional tissues. Modeling individual cells allows us to directly translate biologic observations into simulation rules. In many cases, individual cell agents include molecular-scale models. Most models also simulate the transport of oxygen, drugs, and growth factors, which allow us to link cancer development to microenvironmental conditions. We illustrate these methods with examples drawn from cancer hypoxia, angiogenesis, invasion, stem cells, and immunosurveillance. An ecosystem of interoperable cell-based simulation tools is emerging at a time when cloud computing resources make software easier to access and supercomputing resources make large-scale simulation studies possible. As the field develops, we anticipate that high-throughput simulation studies will allow us to rapidly explore the space of biologic possibilities, prescreen new therapeutic strategies, and even re-engineer tumor and stromal cells to bring cancer systems under control.
Collapse
|
19
|
Abstract
Being concerned by the understanding of the mechanism underlying chronic degenerative diseases , we presented in the previous chapter the medical systems biology conceptual framework that we present for that purpose in this volume. More specifically, we argued there the clear advantages offered by a state-space perspective when applied to the systems-level description of the biomolecular machinery that regulates complex degenerative diseases. We also discussed the importance of the dynamical interplay between the risk factors and the network of interdependencies that characterizes the biochemical, cellular, and tissue-level biomolecular reactions that underlie the physiological processes in health and disease. As we pointed out in the previous chapter, the understanding of this interplay (articulated around cellular phenotypic plasticity properties, regulated by specific kinds of gene regulatory networks) is necessary if prevention is chosen as the human-health improvement strategy (potentially involving the modulation of the patient's lifestyle). In this chapter we provide the medical systems biology mathematical and computational modeling tools required for this task.
Collapse
|
20
|
Liberman A, Kario D, Mussel M, Brill J, Buetow K, Efroni S, Nevo U. Cell studio: A platform for interactive, 3D graphical simulation of immunological processes. APL Bioeng 2018; 2:026107. [PMID: 31069304 PMCID: PMC6481718 DOI: 10.1063/1.5039473] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 05/04/2018] [Indexed: 12/27/2022] Open
Abstract
The field of computer modeling and simulation of biological systems is rapidly advancing, backed by significant progress in the fields of experimentation techniques, computer hardware, and programming software. The result of a simulation may be delivered in several ways, from numerical results, through graphs of the simulated run, to a visualization of the simulation. The vision of an in-silico experiment mimicking an in-vitro or in-vivo experiment as it is viewed under a microscope is appealing but technically demanding and computationally intensive. Here, we report “Cell Studio,” a generic, hybrid platform to simulate an immune microenvironment with biological and biophysical rules. We use game engines—generic programs for game creation which offer ready-made assets and tools—to create a visualized, interactive 3D simulation. We also utilize a scalable architecture that delegates the computational load to a server. The user may view the simulation, move the “camera” around, stop, fast-forward, and rewind it and inject soluble molecules into the extracellular medium at any point in time. During simulation, graphs are created in real time for a broad view of system-wide processes. The model is parametrized using a user-friendly Graphical User Interface (GUI). We show a simple validation simulation and compare its results with those from a “classical” simulation, validated against a “wet” experiment. We believe that interactive, real-time 3D visualization may aid in generating insights from the model and encourage intuition about the immunological scenario.
Collapse
Affiliation(s)
- Asaf Liberman
- The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | - Matan Mussel
- Physics Department, TU Dortmund University, Dortmund 44227, Germany
| | - Jacob Brill
- Arizona State University, Tempe, Arizona 85281, USA
| | | | - Sol Efroni
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 52900, Israel
| | | |
Collapse
|
21
|
Mayorga LS, Verma M, Hontecillas R, Hoops S, Bassaganya-Riera J. Agents and networks to model the dynamic interactions of intracellular transport. CELLULAR LOGISTICS 2017; 7:e1392401. [PMID: 29296512 DOI: 10.1080/21592799.2017.1392401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/03/2017] [Accepted: 10/10/2017] [Indexed: 01/28/2023]
Abstract
Cell biology is increasingly evolving to become a more formal and quantitative science. The field of intracellular transport is no exception. However, it is extremely challenging to formulate mathematical and computational models for processes that involve dynamic structures that continuously change their shape, position and composition, leading to information transfer and functional outcomes. The two major strategies employed to represent intracellular trafficking are based on "ordinary differential equations" and "agent-" based modeling. Both approaches have advantages and drawbacks. Combinations of both modeling strategies have promising characteristics to generate meaningful simulations for intracellular transport and allow the formulation of new hypotheses and provide new insights. In the near future, cell biologists will encounter and hopefully overcome the challenge of translating descriptive cartoon representations of biological systems into mathematical network models.
Collapse
Affiliation(s)
- Luis S Mayorga
- IHEM (Universidad Nacional de Cuyo, CONICET), Facultad de Ciencias Médicas, Facultad de Ciencias Exactas y Naturales, Mendoza, Argentina
| | - Meghna Verma
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| | - Stefan Hoops
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
22
|
Grogan JA, Connor AJ, Markelc B, Muschel RJ, Maini PK, Byrne HM, Pitt-Francis JM. Microvessel Chaste: An Open Library for Spatial Modeling of Vascularized Tissues. Biophys J 2017; 112:1767-1772. [PMID: 28494948 PMCID: PMC5425404 DOI: 10.1016/j.bpj.2017.03.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/22/2017] [Accepted: 03/27/2017] [Indexed: 11/29/2022] Open
Abstract
Spatial models of vascularized tissues are widely used in computational physiology. We introduce a software library for composing multiscale, multiphysics models for applications including tumor growth, angiogenesis, osteogenesis, coronary perfusion, and oxygen delivery. Composition of such models is time consuming, with many researchers writing custom software. Recent advances in imaging have produced detailed three-dimensional (3D) datasets of vascularized tissues at the scale of individual cells. To fully exploit such data there is an increasing need for software that allows user-friendly composition of efficient, 3D models of vascularized tissues, and comparison of predictions with in vivo or in vitro experiments and alternative computational formulations. Microvessel Chaste can be used to build simulations of vessel growth and adaptation in response to mechanical and chemical stimuli; intra- and extravascular transport of nutrients, growth factors and drugs; and cell proliferation in complex 3D geometries. In addition, it can be used to develop custom software for integrating modeling with experimental data processing workflows, facilitated by a comprehensive Python interface to solvers implemented in C++. This article links to two reproducible example problems, showing how the library can be used to build simulations of tumor growth and angiogenesis with realistic vessel networks.
Collapse
Affiliation(s)
- James A Grogan
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom.
| | - Anthony J Connor
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom; Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Bostjan Markelc
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Ruth J Muschel
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Joe M Pitt-Francis
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
23
|
Sütterlin T, Tsingos E, Bensaci J, Stamatas GN, Grabe N. A 3D self-organizing multicellular epidermis model of barrier formation and hydration with realistic cell morphology based on EPISIM. Sci Rep 2017; 7:43472. [PMID: 28262741 PMCID: PMC5338006 DOI: 10.1038/srep43472] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/24/2017] [Indexed: 11/09/2022] Open
Abstract
The epidermis and the stratum corneum (SC) as its outermost layer have evolved to protect the body from evaporative water loss to the environment. To morphologically represent the extremely flattened cells of the SC - and thereby the epidermal barrier - in a multicellular computational model, we developed a 3D biomechanical model (BM) based on ellipsoid cell shapes. We integrated the BM in the multicellular modelling and simulation platform EPISIM. We created a cell behavioural model (CBM) with EPISIM encompassing regulatory feedback loops between the epidermal barrier, water loss to the environment, and water and calcium flow within the tissue. This CBM allows a small number of stem cells to initiate self-organizing epidermal stratification, yielding the spontaneous emergence of water and calcium gradients comparable to experimental data. We find that the 3D in silico epidermis attains homeostasis most quickly at high ambient humidity, and once in homeostasis the epidermal barrier robustly buffers changes in humidity. Our model yields an in silico epidermis with a previously unattained realistic morphology, whose cell neighbour topology is validated with experimental data obtained from in vivo images. This work paves the way to computationally investigate how an impaired SC barrier precipitates disease.
Collapse
Affiliation(s)
- Thomas Sütterlin
- Hamamatsu TIGA Center, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,National Center for Tumor Diseases, Dept. of Medical Oncology, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Erika Tsingos
- Hamamatsu TIGA Center, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,Centre for Organismal Studies, Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Jalil Bensaci
- Emerging Science &Innovation, Johnson &Johnson, Santé Beauté France, 1 rue Camille Desmoulins, 92130 Issy les Moulineaux, France
| | - Georgios N Stamatas
- Emerging Science &Innovation, Johnson &Johnson, Santé Beauté France, 1 rue Camille Desmoulins, 92130 Issy les Moulineaux, France
| | - Niels Grabe
- Hamamatsu TIGA Center, BioQuant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,National Center for Tumor Diseases, Dept. of Medical Oncology, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| |
Collapse
|
24
|
Mc Auley MT, Guimera AM, Hodgson D, Mcdonald N, Mooney KM, Morgan AE, Proctor CJ. Modelling the molecular mechanisms of aging. Biosci Rep 2017; 37:BSR20160177. [PMID: 28096317 PMCID: PMC5322748 DOI: 10.1042/bsr20160177] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/15/2016] [Accepted: 01/16/2017] [Indexed: 01/09/2023] Open
Abstract
The aging process is driven at the cellular level by random molecular damage that slowly accumulates with age. Although cells possess mechanisms to repair or remove damage, they are not 100% efficient and their efficiency declines with age. There are many molecular mechanisms involved and exogenous factors such as stress also contribute to the aging process. The complexity of the aging process has stimulated the use of computational modelling in order to increase our understanding of the system, test hypotheses and make testable predictions. As many different mechanisms are involved, a wide range of models have been developed. This paper gives an overview of the types of models that have been developed, the range of tools used, modelling standards and discusses many specific examples of models that have been grouped according to the main mechanisms that they address. We conclude by discussing the opportunities and challenges for future modelling in this field.
Collapse
Affiliation(s)
- Mark T Mc Auley
- Faculty of Science and Engineering, University of Chester, Chester, U.K
| | - Alvaro Martinez Guimera
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), Newcastle University, Newcastle upon Tyne, Ormskirk, U.K
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, U.K
| | - David Hodgson
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), Newcastle University, Newcastle upon Tyne, Ormskirk, U.K
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - Neil Mcdonald
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), Newcastle University, Newcastle upon Tyne, Ormskirk, U.K
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, U.K
| | | | - Amy E Morgan
- Faculty of Science and Engineering, University of Chester, Chester, U.K
| | - Carole J Proctor
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), Newcastle University, Newcastle upon Tyne, Ormskirk, U.K.
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| |
Collapse
|
25
|
Osborne JM, Fletcher AG, Pitt-Francis JM, Maini PK, Gavaghan DJ. Comparing individual-based approaches to modelling the self-organization of multicellular tissues. PLoS Comput Biol 2017; 13:e1005387. [PMID: 28192427 PMCID: PMC5330541 DOI: 10.1371/journal.pcbi.1005387] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 02/28/2017] [Accepted: 01/28/2017] [Indexed: 12/28/2022] Open
Abstract
The coordinated behaviour of populations of cells plays a central role in tissue growth and renewal. Cells react to their microenvironment by modulating processes such as movement, growth and proliferation, and signalling. Alongside experimental studies, computational models offer a useful means by which to investigate these processes. To this end a variety of cell-based modelling approaches have been developed, ranging from lattice-based cellular automata to lattice-free models that treat cells as point-like particles or extended shapes. However, it remains unclear how these approaches compare when applied to the same biological problem, and what differences in behaviour are due to different model assumptions and abstractions. Here, we exploit the availability of an implementation of five popular cell-based modelling approaches within a consistent computational framework, Chaste (http://www.cs.ox.ac.uk/chaste). This framework allows one to easily change constitutive assumptions within these models. In each case we provide full details of all technical aspects of our model implementations. We compare model implementations using four case studies, chosen to reflect the key cellular processes of proliferation, adhesion, and short- and long-range signalling. These case studies demonstrate the applicability of each model and provide a guide for model usage.
Collapse
Affiliation(s)
- James M. Osborne
- School of Mathematics and Statistics, University of Melbourne, Parkville, Victoria, Australia
| | - Alexander G. Fletcher
- School of Mathematics and Statistics, University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joe M. Pitt-Francis
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Philip K. Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - David J. Gavaghan
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
26
|
Verma M, Hontecillas R, Abedi V, Leber A, Tubau-Juni N, Philipson C, Carbo A, Bassaganya-Riera J. Modeling-Enabled Systems Nutritional Immunology. Front Nutr 2016; 3:5. [PMID: 26909350 PMCID: PMC4754447 DOI: 10.3389/fnut.2016.00005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/01/2016] [Indexed: 12/14/2022] Open
Abstract
This review highlights the fundamental role of nutrition in the maintenance of health, the immune response, and disease prevention. Emerging global mechanistic insights in the field of nutritional immunology cannot be gained through reductionist methods alone or by analyzing a single nutrient at a time. We propose to investigate nutritional immunology as a massively interacting system of interconnected multistage and multiscale networks that encompass hidden mechanisms by which nutrition, microbiome, metabolism, genetic predisposition, and the immune system interact to delineate health and disease. The review sets an unconventional path to apply complex science methodologies to nutritional immunology research, discovery, and development through “use cases” centered around the impact of nutrition on the gut microbiome and immune responses. Our systems nutritional immunology analyses, which include modeling and informatics methodologies in combination with pre-clinical and clinical studies, have the potential to discover emerging systems-wide properties at the interface of the immune system, nutrition, microbiome, and metabolism.
Collapse
Affiliation(s)
- Meghna Verma
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA; The Center for Modeling Immunity to Enteric Pathogens, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA; The Center for Modeling Immunity to Enteric Pathogens, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| | - Vida Abedi
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA; The Center for Modeling Immunity to Enteric Pathogens, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| | - Andrew Leber
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA; The Center for Modeling Immunity to Enteric Pathogens, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| | - Nuria Tubau-Juni
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA; The Center for Modeling Immunity to Enteric Pathogens, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| | | | | | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA; The Center for Modeling Immunity to Enteric Pathogens, Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
27
|
Mei Y, Abedi V, Carbo A, Zhang X, Lu P, Philipson C, Hontecillas R, Hoops S, Liles N, Bassaganya-Riera J. Multiscale modeling of mucosal immune responses. BMC Bioinformatics 2015; 16 Suppl 12:S2. [PMID: 26329787 PMCID: PMC4705510 DOI: 10.1186/1471-2105-16-s12-s2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Computational modeling techniques are playing increasingly important roles in advancing a systems-level mechanistic understanding of biological processes. Computer simulations guide and underpin experimental and clinical efforts. This study presents ENteric Immune Simulator (ENISI), a multiscale modeling tool for modeling the mucosal immune responses. ENISI's modeling environment can simulate in silico experiments from molecular signaling pathways to tissue level events such as tissue lesion formation. ENISI's architecture integrates multiple modeling technologies including ABM (agent-based modeling), ODE (ordinary differential equations), SDE (stochastic modeling equations), and PDE (partial differential equations). This paper focuses on the implementation and developmental challenges of ENISI. A multiscale model of mucosal immune responses during colonic inflammation, including CD4+ T cell differentiation and tissue level cell-cell interactions was developed to illustrate the capabilities, power and scope of ENISI MSM.
Collapse
|
28
|
|
29
|
D'Alessandro LA, Hoehme S, Henney A, Drasdo D, Klingmüller U. Unraveling liver complexity from molecular to organ level: challenges and perspectives. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 117:78-86. [PMID: 25433231 DOI: 10.1016/j.pbiomolbio.2014.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/28/2014] [Accepted: 11/19/2014] [Indexed: 12/13/2022]
Abstract
Biological responses are determined by information processing at multiple and highly interconnected scales. Within a tissue the individual cells respond to extracellular stimuli by regulating intracellular signaling pathways that in turn determine cell fate decisions and influence the behavior of neighboring cells. As a consequence the cellular responses critically impact tissue composition and architecture. Understanding the regulation of these mechanisms at different scales is key to unravel the emergent properties of biological systems. In this perspective, a multidisciplinary approach combining experimental data with mathematical modeling is introduced. We report the approach applied within the Virtual Liver Network to analyze processes that regulate liver functions from single cell responses to the organ level using a number of examples. By facilitating interdisciplinary collaborations, the Virtual Liver Network studies liver regeneration and inflammatory processes as well as liver metabolic functions at multiple scales, and thus provides a suitable example to identify challenges and point out potential future application of multi-scale systems biology.
Collapse
Affiliation(s)
- L A D'Alessandro
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany
| | - S Hoehme
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, Germany
| | - A Henney
- Obsidian Biomedical Consulting Ltd., Macclesfield, UK; The German Virtual Liver Network, University of Heidelberg, 69120 Heidelberg, Germany
| | - D Drasdo
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, Germany; Institut National de Recherche en Informatique et en Automatique (INRIA), Domaine de Voluceau, 78150 Rocquencourt, France; University Pierre and Marie Curie and CNRS UMR 7598, LJLL, F-75005 Paris, France; CNRS, 7598 Paris, France
| | - U Klingmüller
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany.
| |
Collapse
|
30
|
Safferling K, Sütterlin T, Westphal K, Ernst C, Breuhahn K, James M, Jäger D, Halama N, Grabe N. Wound healing revised: a novel reepithelialization mechanism revealed by in vitro and in silico models. ACTA ACUST UNITED AC 2014; 203:691-709. [PMID: 24385489 PMCID: PMC3840932 DOI: 10.1083/jcb.201212020] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Experimental analysis and computational modeling of epidermal wound closure in 3D suggests an important role for surrounding tissue in determining epithelial cell movement and fate. Wound healing is a complex process in which a tissue’s individual cells have to be orchestrated in an efficient and robust way. We integrated multiplex protein analysis, immunohistochemical analysis, and whole-slide imaging into a novel medium-throughput platform for quantitatively capturing proliferation, differentiation, and migration in large numbers of organotypic skin cultures comprising epidermis and dermis. Using fluorescent time-lag staining, we were able to infer source and final destination of keratinocytes in the healing epidermis. This resulted in a novel extending shield reepithelialization mechanism, which we confirmed by computational multicellular modeling and perturbation of tongue extension. This work provides a consistent experimental and theoretical model for epidermal wound closure in 3D, negating the previously proposed concepts of epidermal tongue extension and highlighting the so far underestimated role of the surrounding tissue. Based on our findings, epidermal wound closure is a process in which cell behavior is orchestrated by a higher level of tissue control that 2D monolayer assays are not able to capture.
Collapse
Affiliation(s)
- Kai Safferling
- Hamamatsu Tissue Imaging and Analysis Center, BIOQUANT, and 2 Department of Medical Oncology, National Center for Tumor Diseases, University of Heidelberg, 69117 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Starruß J, de Back W, Brusch L, Deutsch A. Morpheus: a user-friendly modeling environment for multiscale and multicellular systems biology. ACTA ACUST UNITED AC 2014; 30:1331-2. [PMID: 24443380 PMCID: PMC3998129 DOI: 10.1093/bioinformatics/btt772] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Summary: Morpheus is a modeling environment for the simulation and integration of cell-based models with ordinary differential equations and reaction-diffusion systems. It allows rapid development of multiscale models in biological terms and mathematical expressions rather than programming code. Its graphical user interface supports the entire workflow from model construction and simulation to visualization, archiving and batch processing. Availability and implementation: Binary packages are available at http://imc.zih.tu-dresden.de/wiki/morpheus for Linux, Mac OSX and MS Windows. Contact:walter.deback@tu-dresden.de Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jörn Starruß
- Center for Information Services and High Performance Computing, Technische Universität Dresden, 01062 Dresden, Germany
| | | | | | | |
Collapse
|
32
|
|