1
|
Chiang H, Chung CA. Simulation of Soluble and Bound VEGF-stimulated in vitro Capillary-like Network Formation on Deformed Substrate. PLoS Comput Biol 2024; 20:e1012281. [PMID: 39038038 PMCID: PMC11262697 DOI: 10.1371/journal.pcbi.1012281] [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: 12/08/2023] [Accepted: 06/26/2024] [Indexed: 07/24/2024] Open
Abstract
Capillary plexus cultivation is crucial in tissue engineering and regenerative medicine. Theoretical simulations have been conducted to supplement the expensive experimental works. However, the mechanisms connecting mechanical and chemical stimuli remained undefined, and the functions of the different VEGF forms in the culture environment were still unclear. In this paper, we developed a hybrid model for simulating short-term in vitro capillary incubations. We used the Cellular Potts model to predict individual cell migration, morphology change, and continuum mechanics to quantify biogel deformation and VEGF transport dynamics. By bridging the mechanical regulation and chemical stimulation in the model, the results showed good agreement between the predicted network topology and experiments, in which elongated cells connected, forming the network cords and round cells gathered, creating cobblestone-like aggregates. The results revealed that the capillary-like networks could develop in high integrity only when the mechanical and chemical couplings worked adequately, with the cell morphology and haptotaxis driven by the soluble and bound forms of VEGF, respectively, functioning simultaneously.
Collapse
Affiliation(s)
- Hsun Chiang
- Department of Mechanical Engineering, National Central University, Taoyuan, Taiwan
| | - Chih-Ang Chung
- Department of Mechanical Engineering, National Central University, Taoyuan, Taiwan
| |
Collapse
|
2
|
Crossley RM, Johnson S, Tsingos E, Bell Z, Berardi M, Botticelli M, Braat QJS, Metzcar J, Ruscone M, Yin Y, Shuttleworth R. Modeling the extracellular matrix in cell migration and morphogenesis: a guide for the curious biologist. Front Cell Dev Biol 2024; 12:1354132. [PMID: 38495620 PMCID: PMC10940354 DOI: 10.3389/fcell.2024.1354132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The extracellular matrix (ECM) is a highly complex structure through which biochemical and mechanical signals are transmitted. In processes of cell migration, the ECM also acts as a scaffold, providing structural support to cells as well as points of potential attachment. Although the ECM is a well-studied structure, its role in many biological processes remains difficult to investigate comprehensively due to its complexity and structural variation within an organism. In tandem with experiments, mathematical models are helpful in refining and testing hypotheses, generating predictions, and exploring conditions outside the scope of experiments. Such models can be combined and calibrated with in vivo and in vitro data to identify critical cell-ECM interactions that drive developmental and homeostatic processes, or the progression of diseases. In this review, we focus on mathematical and computational models of the ECM in processes such as cell migration including cancer metastasis, and in tissue structure and morphogenesis. By highlighting the predictive power of these models, we aim to help bridge the gap between experimental and computational approaches to studying the ECM and to provide guidance on selecting an appropriate model framework to complement corresponding experimental studies.
Collapse
Affiliation(s)
- Rebecca M. Crossley
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Samuel Johnson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Erika Tsingos
- Computational Developmental Biology Group, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Zoe Bell
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Optics11 life, Amsterdam, Netherlands
| | | | - Quirine J. S. Braat
- Department of Applied Physics and Science Education, Eindhoven University of Technology, Eindhoven, Netherlands
| | - John Metzcar
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
- Department of Informatics, Indiana University, Bloomington, IN, United States
| | | | - Yuan Yin
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
3
|
Stepanova D, Byrne HM, Maini PK, Alarcón T. Computational modeling of angiogenesis: The importance of cell rearrangements during vascular growth. WIREs Mech Dis 2024; 16:e1634. [PMID: 38084799 DOI: 10.1002/wsbm.1634] [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: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 03/16/2024]
Abstract
Angiogenesis is the process wherein endothelial cells (ECs) form sprouts that elongate from the pre-existing vasculature to create new vascular networks. In addition to its essential role in normal development, angiogenesis plays a vital role in pathologies such as cancer, diabetes and atherosclerosis. Mathematical and computational modeling has contributed to unraveling its complexity. Many existing theoretical models of angiogenic sprouting are based on the "snail-trail" hypothesis. This framework assumes that leading ECs positioned at sprout tips migrate toward low-oxygen regions while other ECs in the sprout passively follow the leaders' trails and proliferate to maintain sprout integrity. However, experimental results indicate that, contrary to the snail-trail assumption, ECs exchange positions within developing vessels, and the elongation of sprouts is primarily driven by directed migration of ECs. The functional role of cell rearrangements remains unclear. This review of the theoretical modeling of angiogenesis is the first to focus on the phenomenon of cell mixing during early sprouting. We start by describing the biological processes that occur during early angiogenesis, such as phenotype specification, cell rearrangements and cell interactions with the microenvironment. Next, we provide an overview of various theoretical approaches that have been employed to model angiogenesis, with particular emphasis on recent in silico models that account for the phenomenon of cell mixing. Finally, we discuss when cell mixing should be incorporated into theoretical models and what essential modeling components such models should include in order to investigate its functional role. This article is categorized under: Cardiovascular Diseases > Computational Models Cancer > Computational Models.
Collapse
Affiliation(s)
- Daria Stepanova
- Laboratorio Subterráneo de Canfranc, Canfranc-Estación, Huesca, Spain
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Tomás Alarcón
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
- Centre de Recerca Matemàtica, Bellaterra, Barcelona, Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
4
|
Rojek KO, Wrzos A, Żukowski S, Bogdan M, Lisicki M, Szymczak P, Guzowski J. Long-term day-by-day tracking of microvascular networks sprouting in fibrin gels: From detailed morphological analyses to general growth rules. APL Bioeng 2024; 8:016106. [PMID: 38327714 PMCID: PMC10849774 DOI: 10.1063/5.0180703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/04/2024] [Indexed: 02/09/2024] Open
Abstract
Understanding and controlling of the evolution of sprouting vascular networks remains one of the basic challenges in tissue engineering. Previous studies on the vascularization dynamics have typically focused only on the phase of intense growth and often lacked spatial control over the initial cell arrangement. Here, we perform long-term day-by-day analysis of tens of isolated microvasculatures sprouting from endothelial cell-coated spherical beads embedded in an external fibrin gel. We systematically study the topological evolution of the sprouting networks over their whole lifespan, i.e., for at least 14 days. We develop a custom image analysis toolkit and quantify (i) the overall length and area of the sprouts, (ii) the distributions of segment lengths and branching angles, and (iii) the average number of branch generations-a measure of network complexity. We show that higher concentrations of vascular endothelial growth factor (VEGF) lead to earlier sprouting and more branched networks, yet without significantly affecting the speed of growth of individual sprouts. We find that the mean branching angle is weakly dependent on VEGF and typically in the range of 60°-75°, suggesting that, by comparison with the available diffusion-limited growth models, the bifurcating tips tend to follow local VEGF gradients. At high VEGF concentrations, we observe exponential distributions of segment lengths, which signify purely stochastic branching. Our results-due to their high statistical relevance-may serve as a benchmark for predictive models, while our new image analysis toolkit, offering unique features and high speed of operation, could be exploited in future angiogenic drug tests.
Collapse
Affiliation(s)
- Katarzyna O. Rojek
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Antoni Wrzos
- Institute of Theoretical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | | | - Michał Bogdan
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Lisicki
- Institute of Theoretical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Piotr Szymczak
- Institute of Theoretical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Jan Guzowski
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
5
|
Davidson CD, Midekssa FS, DePalma SJ, Kamen JL, Wang WY, Jayco DKP, Wieger ME, Baker BM. Mechanical Intercellular Communication via Matrix-Borne Cell Force Transmission During Vascular Network Formation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306210. [PMID: 37997199 PMCID: PMC10797481 DOI: 10.1002/advs.202306210] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Indexed: 11/25/2023]
Abstract
Intercellular communication is critical to the formation and homeostatic function of all tissues. Previous work has shown that cells can communicate mechanically via the transmission of cell-generated forces through their surrounding extracellular matrix, but this process is not well understood. Here, mechanically defined, synthetic electrospun fibrous matrices are utilized in conjunction with a microfabrication-based cell patterning approach to examine mechanical intercellular communication (MIC) between endothelial cells (ECs) during their assembly into interconnected multicellular networks. It is found that cell force-mediated matrix displacements in deformable fibrous matrices underly directional extension and migration of neighboring ECs toward each other prior to the formation of stable cell-cell connections enriched with vascular endothelial cadherin (VE-cadherin). A critical role is also identified for calcium signaling mediated by focal adhesion kinase and mechanosensitive ion channels in MIC that extends to multicellular assembly of 3D vessel-like networks when ECs are embedded within fibrin hydrogels. These results illustrate a role for cell-generated forces and ECM mechanical properties in multicellular assembly of capillary-like EC networks and motivates the design of biomaterials that promote MIC for vascular tissue engineering.
Collapse
Affiliation(s)
| | - Firaol S. Midekssa
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Samuel J. DePalma
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Jordan L. Kamen
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - William Y. Wang
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | | | - Megan E. Wieger
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Brendon M. Baker
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMI48109USA
| |
Collapse
|
6
|
Noerr PS, Zamora Alvarado JE, Golnaraghi F, McCloskey KE, Gopinathan A, Dasbiswas K. Optimal mechanical interactions direct multicellular network formation on elastic substrates. Proc Natl Acad Sci U S A 2023; 120:e2301555120. [PMID: 37910554 PMCID: PMC10636364 DOI: 10.1073/pnas.2301555120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 09/09/2023] [Indexed: 11/03/2023] Open
Abstract
Cells self-organize into functional, ordered structures during tissue morphogenesis, a process that is evocative of colloidal self-assembly into engineered soft materials. Understanding how intercellular mechanical interactions may drive the formation of ordered and functional multicellular structures is important in developmental biology and tissue engineering. Here, by combining an agent-based model for contractile cells on elastic substrates with endothelial cell culture experiments, we show that substrate deformation-mediated mechanical interactions between cells can cluster and align them into branched networks. Motivated by the structure and function of vasculogenic networks, we predict how measures of network connectivity like percolation probability and fractal dimension as well as local morphological features including junctions, branches, and rings depend on cell contractility and density and on substrate elastic properties including stiffness and compressibility. We predict and confirm with experiments that cell network formation is substrate stiffness dependent, being optimal at intermediate stiffness. We also show the agreement between experimental data and predicted cell cluster types by mapping a combined phase diagram in cell density substrate stiffness. Overall, we show that long-range, mechanical interactions provide an optimal and general strategy for multicellular self-organization, leading to more robust and efficient realizations of space-spanning networks than through just local intercellular interactions.
Collapse
Affiliation(s)
- Patrick S. Noerr
- Department of Physics, University of California, Merced, CA95343
| | - Jose E. Zamora Alvarado
- Department of Materials and Biomaterials Science and Engineering, University of California, Merced, CA95343
| | | | - Kara E. McCloskey
- Department of Materials and Biomaterials Science and Engineering, University of California, Merced, CA95343
| | - Ajay Gopinathan
- Department of Physics, University of California, Merced, CA95343
| | - Kinjal Dasbiswas
- Department of Physics, University of California, Merced, CA95343
| |
Collapse
|
7
|
Dazzi C, Mehl J, Benamar M, Gerhardt H, Knaus P, Duda GN, Checa S. External mechanical loading overrules cell-cell mechanical communication in sprouting angiogenesis during early bone regeneration. PLoS Comput Biol 2023; 19:e1011647. [PMID: 37956208 PMCID: PMC10681321 DOI: 10.1371/journal.pcbi.1011647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 11/27/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Sprouting angiogenesis plays a key role during bone regeneration. For example, insufficient early revascularization of the injured site can lead to delayed or non-healing. During sprouting, endothelial cells are known to be mechano-sensitive and respond to local mechanical stimuli. Endothelial cells interact and communicate mechanically with their surroundings, such as outer-vascular stromal cells, through cell-induced traction forces. In addition, external physiological loads act at the healing site, resulting in tissue deformations and impacting cellular arrangements. How these two distinct mechanical cues (cell-induced and external) impact angiogenesis and sprout patterning in early bone healing remains however largely unknown. Therefore, the aim of this study was to investigate the relative role of externally applied and cell-induced mechanical signals in driving sprout patterning at the onset of bone healing. To investigate cellular self-organisation in early bone healing, an in silico model accounting for the mechano-regulation of sprouting angiogenesis and stromal cell organization was developed. Computer model predictions were compared to in vivo experiments of a mouse osteotomy model stabilized with a rigid or a semirigid fixation system. We found that the magnitude and orientation of principal strains within the healing region can explain experimentally observed sprout patterning, under both fixation conditions. Furthermore, upon simulating the selective inhibition of either cell-induced or externally applied mechanical cues, external mechanical signals appear to overrule the mechanical communication acting on a cell-cell interaction level. Such findings illustrate the relevance of external mechanical signals over the local cell-mediated mechanical cues and could be used in the design of fracture treatment strategies for bone regeneration.
Collapse
Affiliation(s)
- Chiara Dazzi
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Mehl
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Mounir Benamar
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
8
|
Tsingos E, Bakker BH, Keijzer KAE, Hupkes HJ, Merks RMH. Hybrid cellular Potts and bead-spring modeling of cells in fibrous extracellular matrix. Biophys J 2023; 122:2609-2622. [PMID: 37183398 PMCID: PMC10397577 DOI: 10.1016/j.bpj.2023.05.013] [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: 07/08/2022] [Revised: 02/17/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023] Open
Abstract
The mechanical interaction between cells and the extracellular matrix (ECM) is fundamental to coordinate collective cell behavior in tissues. Relating individual cell-level mechanics to tissue-scale collective behavior is a challenge that cell-based models such as the cellular Potts model (CPM) are well-positioned to address. These models generally represent the ECM with mean-field approaches, which assume substrate homogeneity. This assumption breaks down with fibrous ECM, which has nontrivial structure and mechanics. Here, we extend the CPM with a bead-spring model of ECM fiber networks modeled using molecular dynamics. We model a contractile cell pulling with discrete focal adhesion-like sites on the fiber network and demonstrate agreement with experimental spatiotemporal fiber densification and displacement. We show that at high network cross-linking, contractile cell forces propagate over at least eight cell diameters, decaying with distance with power law exponent n= 0.35 - 0.65 typical of viscoelastic ECMs. Further, we use in silico atomic force microscopy to measure local cell-induced network stiffening consistent with experiments. Our model lays the foundation for investigating how local and long-ranged cell-ECM mechanobiology contributes to multicellular morphogenesis.
Collapse
Affiliation(s)
- Erika Tsingos
- Mathematical Institute, Leiden University, Leiden, the Netherlands.
| | | | - Koen A E Keijzer
- Mathematical Institute, Leiden University, Leiden, the Netherlands
| | | | - Roeland M H Merks
- Mathematical Institute, Leiden University, Leiden, the Netherlands; Institute for Biology Leiden, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
9
|
Ferre-Torres J, Noguera-Monteagudo A, Lopez-Canosa A, Romero-Arias JR, Barrio R, Castaño O, Hernandez-Machado A. Modelling of chemotactic sprouting endothelial cells through an extracellular matrix. Front Bioeng Biotechnol 2023; 11:1145550. [PMID: 37362221 PMCID: PMC10285466 DOI: 10.3389/fbioe.2023.1145550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Sprouting angiogenesis is a core biological process critical to vascular development. Its accurate simulation, relevant to multiple facets of human health, is of broad, interdisciplinary appeal. This study presents an in-silico model replicating a microfluidic assay where endothelial cells sprout into a biomimetic extracellular matrix, specifically, a large-pore, low-concentration fibrin-based porous hydrogel, influenced by chemotactic factors. We introduce a novel approach by incorporating the extracellular matrix and chemotactic factor effects into a unified term using a single parameter, primarily focusing on modelling sprouting dynamics and morphology. This continuous model naturally describes chemotactic-induced sprouting with no need for additional rules. In addition, we extended our base model to account for matrix sensing and degradation, crucial aspects of angiogenesis. We validate our model via a hybrid in-silico experimental method, comparing the model predictions with experimental results derived from the microfluidic setup. Our results underscore the intricate relationship between the extracellular matrix structure and angiogenic sprouting, proposing a promising method for predicting the influence of the extracellular matrix on angiogenesis.
Collapse
Affiliation(s)
- Josep Ferre-Torres
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | | | - Adrian Lopez-Canosa
- Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Spain
| | - J Roberto Romero-Arias
- Institute for Research in Applied Mathematics and Systems, National Autonomous University of Mexico , Mexico City, Mexico
| | - Rafael Barrio
- Institute of Physics, National Autonomous University of Mexico, Mexico City, Mexico
| | - Oscar Castaño
- Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona (UB), Barcelona, Spain
| | - Aurora Hernandez-Machado
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona (UB), Barcelona, Spain
| |
Collapse
|
10
|
Ahmed RK, Abdalrahman T, Davies NH, Vermolen F, Franz T. Mathematical model of mechano-sensing and mechanically induced collective motility of cells on planar elastic substrates. Biomech Model Mechanobiol 2023; 22:809-824. [PMID: 36814004 DOI: 10.1007/s10237-022-01682-2] [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: 08/04/2022] [Accepted: 12/28/2022] [Indexed: 02/24/2023]
Abstract
Cells mechanically interact with their environment to sense, for example, topography, elasticity and mechanical cues from other cells. Mechano-sensing has profound effects on cellular behaviour, including motility. The current study aims to develop a mathematical model of cellular mechano-sensing on planar elastic substrates and demonstrate the model's predictive capabilities for the motility of individual cells in a colony. In the model, a cell is assumed to transmit an adhesion force, derived from a dynamic focal adhesion integrin density, that locally deforms a substrate, and to sense substrate deformation originating from neighbouring cells. The substrate deformation from multiple cells is expressed as total strain energy density with a spatially varying gradient. The magnitude and direction of the gradient at the cell location define the cell motion. Cell-substrate friction, partial motion randomness, and cell death and division are included. The substrate deformation by a single cell and the motility of two cells are presented for several substrate elasticities and thicknesses. The collective motility of 25 cells on a uniform substrate mimicking the closure of a circular wound of 200 µm is predicted for deterministic and random motion. Cell motility on substrates with varying elasticity and thickness is explored for four cells and 15 cells, the latter again mimicking wound closure. Wound closure by 45 cells is used to demonstrate the simulation of cell death and division during migration. The mathematical model can adequately simulate the mechanically induced collective cell motility on planar elastic substrates. The model is suitable for extension to other cell and substrates shapes and the inclusion of chemotactic cues, offering the potential to complement in vitro and in vivo studies.
Collapse
Affiliation(s)
- Riham K Ahmed
- Division of Biomedical Engineering, Department of Human Biology, Biomedical Engineering Research Centre, University of Cape Town, Observatory, South Africa.
| | - Tamer Abdalrahman
- Division of Biomedical Engineering, Department of Human Biology, Biomedical Engineering Research Centre, University of Cape Town, Observatory, South Africa
- Computational Mechanobiology, Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin, Berlin, Germany
| | - Neil H Davies
- Cardiovascular Research Unit, Chris Barnard Division of Cardiothoracic Surgery, MRC IUCHRU, University of Cape Town, Observatory, South Africa
| | - Fred Vermolen
- Computational Mathematics Group, Department of Mathematics and Statistics, University of Hasselt, Diepenbeek, Belgium
| | - Thomas Franz
- Division of Biomedical Engineering, Department of Human Biology, Biomedical Engineering Research Centre, University of Cape Town, Observatory, South Africa
- Bioengineering Science Research Group, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
11
|
Odagiri K, Fujisaki H, Takada H, Ogawa R. Mathematical model for promotion of wound closure with ATP release. Biophys Physicobiol 2023; 20:e200023. [PMID: 38496238 PMCID: PMC10941958 DOI: 10.2142/biophysico.bppb-v20.0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/22/2023] [Indexed: 03/19/2024] Open
Abstract
To computationally investigate the recent experimental finding such that extracellular ATP release caused by exogeneous mechanical forces promote wound closure, we introduce a mathematical model, the Cellular Potts Model (CPM), which is a popular discretized model on a lattice, where the movement of a "cell" is determined by a Monte Carlo procedure. In the experiment, it was observed that there is mechanosensitive ATP release from the leading cells facing the wound gap and the subsequent extracellular Ca2+ influx. To model these phenomena, the Reaction-Diffusion equations for extracellular ATP and intracellular Ca2+ concentrations are adopted and combined with CPM, where we also add a polarity term because the cell migration is enhanced in the case of ATP release. From the numerical simulations using this hybrid model, we discuss effects of the collective cell migration due to the ATP release and the Ca2+ influx caused by the mechanical forces and the consequent promotion of wound closure.
Collapse
Affiliation(s)
- Kenta Odagiri
- School of Network and Information, Senshu University, Kawasaki, Kanagawa 214-8580, Japan
- AMED-CREST, Bunkyo, Tokyo 113-8603, Japan
| | - Hiroshi Fujisaki
- AMED-CREST, Bunkyo, Tokyo 113-8603, Japan
- Department of Physics, Nippon Medical School, Musashino, Tokyo 180-0023, Japan
| | - Hiroya Takada
- AMED-CREST, Bunkyo, Tokyo 113-8603, Japan
- Department of Anti-Aging and Preventive Medicine, Nippon Medical School, Bunkyo, Tokyo 113-8603, Japan
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Bunkyo, Tokyo 113-8603, Japan
| | - Rei Ogawa
- AMED-CREST, Bunkyo, Tokyo 113-8603, Japan
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Bunkyo, Tokyo 113-8603, Japan
| |
Collapse
|
12
|
Carrasco-Mantis A, Alarcón T, Sanz-Herrera JA. An in silico study on the influence of extracellular matrix mechanics on vasculogenesis. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 231:107369. [PMID: 36738607 DOI: 10.1016/j.cmpb.2023.107369] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND AND OBJECTIVES Blood vessels form a network of capillaries throughout the body that perform essential functions for life. Vasculogenesis, i.e. the formation of new blood vessels, is regulated by many factors, biochemical ones being among the most important. However, others such as the biomechanical influence on shape, organization and structure of vessel networks require further investigation. In this paper, we develop a 3D agent-based mechanobiological model of vasculogenesis with the aim of analyzing how the mechanics of the extracellular matrix (ECM) affects vasculogenesis. METHODS For this purpose, we consider a growing domain composed of different cells: tip cells, which are the driving cells located at the end of the vessels and stalk cells, which are found in the interior of the vascular network. ECM is considered as particles (agents) that surround the growth of the vascular network. Depending on the cell type, different sets of forces are considered, such as chemotactic, mechanical, random and viscoelastic forces among others. RESULTS The growth of the network is iteratively analyzed and updated at each time step based on a mechanically-driven proliferation rule. The influence of different biomechanical factors, such as ECM stiffness or viscoelasticity are explored through in silico simulations. A number of indicators are defined along the algorithm, like number of cells, branches, tortuosity and anisotropy, in order to compare topological differences of the vascular network during vasculogenesis under different ECM conditions. The obtained results are qualitatively compared with other related works in the literature. CONCLUSIONS The present study sheds some light and partially explain, from an in silico perspective, the role of ECM mechanics on vasculogenesis. The main conclusions of this work are: (i) increased stiffness increases proliferation, (ii) the network tends to migrate towards stiffer areas, and (iii) increased viscoelasticity decreases proliferation.
Collapse
Affiliation(s)
- A Carrasco-Mantis
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla, Spain
| | - T Alarcón
- ICREA (Institució Catalana de Recerca i Estudis Avançats), Centre de Recerca Matemàtica, Barcelona, Spain
| | - J A Sanz-Herrera
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla, Spain.
| |
Collapse
|
13
|
Liu C, Nguyen RY, Pizzurro GA, Zhang X, Gong X, Martinez AR, Mak M. Self-assembly of mesoscale collagen architectures and applications in 3D cell migration. Acta Biomater 2023; 155:167-181. [PMID: 36371004 PMCID: PMC9805527 DOI: 10.1016/j.actbio.2022.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022]
Abstract
3D in vitro tumor models have recently been investigated as they can recapitulate key features in the tumor microenvironment. Reconstruction of a biomimetic scaffold is critical in these models. However, most current methods focus on modulating local properties, e.g. micro- and nano-scaled topographies, without capturing the global millimeter or intermediate mesoscale features. Here we introduced a method for modulating the collagen I-based extracellular matrix structure by disruption of fibrillogenesis and the gelation process through mechanical agitation. With this method, we generated collagen scaffolds that are thickened and wavy at a larger scale while featuring global softness. Thickened collagen patches were interconnected with loose collagen networks, highly resembling collagen architecture in the tumor stroma. This thickened collagen network promoted tumor cell dissemination. In addition, this novel modified scaffold triggered differences in morphology and migratory behaviors of tumor cells. Altogether, our method for altered collagen architecture paves new ways for studying in detail cell behavior in physiologically relevant biological processes. STATEMENT OF SIGNIFICANCE: Tumor progression usually involves chronic tissue damage and repair processes. Hallmarks of tumors are highly overlapped with those of wound healing. To mimic the tumor milieu, collagen-based scaffolds are widely used. These scaffolds focus on modulating microscale topographies and mechanics, lacking global architecture similarity compared with in vivo architecture. Here we introduced one type of thick collagen bundles that mimics ECM architecture in human skin scars. These thickened collagen bundles are long and wavy while featuring global softness. This collagen architecture imposes fewer steric restraints and promotes tumor cell dissemination. Our findings demonstrate a distinct picture of cell behaviors and intercellular interactions, highlighting the importance of collagen architecture and spatial heterogeneity of the tumor microenvironment.
Collapse
Affiliation(s)
- Chang Liu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | - Ryan Y Nguyen
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | - Gabriela A Pizzurro
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | - Xingjian Zhang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States
| | | | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, United States.
| |
Collapse
|
14
|
Morozov AM, Sergeev AN, Sungurova AV, Morozov DV, Belyak MA, Domracheva AS. Computer simulation of the wound process (review of literature). BULLETIN OF THE MEDICAL INSTITUTE "REAVIZ" (REHABILITATION, DOCTOR AND HEALTH) 2022. [DOI: 10.20340/vmi-rvz.2023.1.ictm.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Relevance. Computer simulation is a mathematical modeling process performed on a computer that is designed to predict the behavior or results of a real or physical system. Computer simulation has a number of advantages over classical models of animal experiments: the cheapness of the method (the need to acquire and maintain animals disappears by itself), the speed of obtaining results, the absence of bioethical problems, the ability to change the conditions of the experiment, etc.he purpose of this study is to review the methods of computer simulation of the wound process, to identify the shortcomings of the models and propose ways to solve them, as well as to select the best existing model for describing wound regeneration.Material and methods. In the course of this work, an analysis was made of foreign and domestic literature on the problem of computer modeling of the wound process.Results. After analyzing the relevant literature on this topic, the problem is seen precisely in the insufficiently studied process of wound regeneration, since many different cells, cytokines, growth factors, enzymes, fibrillar proteins, etc. take part in it. The models that currently exist describe wound regeneration only in an extremely generalized way, which does not allow us to apply them in clinical situations. Analyzing literature sources, we came to the conclusion that both numerical approaches, both cellular-biochemical (the first type of models) and phenomenological (the second type) are applicable in the case of wound modeling and can be used very successfully. The problem is that on the basis of one approach it is impossible to display a complete picture of wound healing, in this way it is possible to predict only individual regeneration parameters necessary for certain purposes due to the complexity and versatility of this typical pathophysiological process.Conclusion. Computer modeling of wounds is still a controversial and complex topic. Existing models are not intended to describe all the processes occurring in a healing wound. It is much more productive to describe the various phenomena during healing separately. This is due to the fact that many elements are involved in the regeneration of the skin, which are almost impossible to take into account in full. The available models are of exclusively scientific value, consisting in attempts to understand all complex processes and interactions. Practical application is difficult, since existing models require specific input data that require highly specialized equipment. If we abstract from all this, then the best existing model of the first type is the model of the authors Yangyang Wang, Christian F. Guerrero-Juarez, Yuchi Qiu and co-authors, in addition to it, any of the described phenomenological models will do.
Collapse
|
15
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
16
|
Fernandes A, Miéville A, Grob F, Yamashita T, Mehl J, Hosseini V, Emmert MY, Falk V, Vogel V. Endothelial-Smooth Muscle Cell Interactions in a Shear-Exposed Intimal Hyperplasia on-a-Dish Model to Evaluate Therapeutic Strategies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202317. [PMID: 35971167 PMCID: PMC9534971 DOI: 10.1002/advs.202202317] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Indexed: 05/25/2023]
Abstract
Intimal hyperplasia (IH) represents a major challenge following cardiovascular interventions. While mechanisms are poorly understood, the inefficient preventive methods incentivize the search for novel therapies. A vessel-on-a-dish platform is presented, consisting of direct-contact cocultures with human primary endothelial cells (ECs) and smooth muscle cells (SMCs) exposed to both laminar pulsatile and disturbed flow on an orbital shaker. With contractile SMCs sitting below a confluent EC layer, a model that successfully replicates the architecture of a quiescent vessel wall is created. In the novel IH model, ECs are seeded on synthetic SMCs at low density, mimicking reendothelization after vascular injury. Over 3 days of coculture, ECs transition from a network conformation to confluent 2D islands, as promoted by pulsatile flow, resulting in a "defected" EC monolayer. In defected regions, SMCs incorporated plasma fibronectin into fibers, increased proliferation, and formed multilayers, similarly to IH in vivo. These phenomena are inhibited under confluent EC layers, supporting therapeutic approaches that focus on endothelial regeneration rather than inhibiting proliferation, as illustrated in a proof-of-concept experiment with Paclitaxel. Thus, this in vitro system offers a new tool to study EC-SMC communication in IH pathophysiology, while providing an easy-to-use translational disease model platform for low-cost and high-content therapeutic development.
Collapse
Affiliation(s)
- Andreia Fernandes
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Arnaud Miéville
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Franziska Grob
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Tadahiro Yamashita
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
- Present address:
Department of System Design EngineeringKeio University108‐8345YokohamaJapan
| | - Julia Mehl
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
- Present address:
Julius Wolff InstituteBerlin Institute of HealthCharité Universitätsmedizin Berlin10117BerlinGermany
| | - Vahid Hosseini
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Maximilian Y. Emmert
- Department of Cardiovascular SurgeryCharité Universitätsmedizin Berlin10117BerlinGermany
- Department of Cardiothoracic and Vascular SurgeryGerman Heart Center Berlin13353BerlinGermany
- Institute for Regenerative Medicine (IREM)University of Zurich8006ZurichSwitzerland
| | - Volkmar Falk
- Department of Cardiovascular SurgeryCharité Universitätsmedizin Berlin10117BerlinGermany
- Department of Cardiothoracic and Vascular SurgeryGerman Heart Center Berlin13353BerlinGermany
- Department of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| | - Viola Vogel
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH Zurich8093ZurichSwitzerland
| |
Collapse
|
17
|
Intercellular communication in the tumour microecosystem: Mediators and therapeutic approaches for hepatocellular carcinoma. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166528. [PMID: 36007784 DOI: 10.1016/j.bbadis.2022.166528] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022]
Abstract
Hepatocellular carcinoma (HCC), one of the most common tumours worldwide, is one of the main causes of mortality in cancer patients. There are still numerous problems hindering its early diagnosis, which lead to late patients receiving treatment, and these problems need to be solved urgently. The tumour microecosystem is a complex network system comprising seven parts: the hypoxia niche, immune microenvironment, metabolic microenvironment, acidic niche, innervated niche, mechanical microenvironment, and microbial microenvironment. Intercellular communication is divided into direct contact and indirect communication. Direct contact communication includes gap junctions, tunneling nanotubes, and receptor-ligand interactions, whereas indirect communication includes exosomes, apoptotic vesicles, and soluble factors. Mechanical communication and cytoplasmic exchange are further means of intercellular communication. Intercellular communication mediates the crosstalk between the tumour microecosystem and the host as well as that between cells and cell-free components in the tumour microecosystem, causing changes in the tumour hallmarks of the HCC microecosystem such as changes in tumour proliferation, invasion, apoptosis, angiogenesis, metastasis, inflammatory response, gene mutation, immune escape, metabolic reprogramming, and therapeutic resistance. Here, we review the role of the above-mentioned intercellular communication in the HCC microecosystem and discuss the advantages of targeted intercellular communication in the clinical diagnosis and treatment of HCC. Finally, the current problems and prospects are discussed.
Collapse
|
18
|
He X, Lee B, Jiang Y. Extracellular matrix in cancer progression and therapy. MEDICAL REVIEW (2021) 2022; 2:125-139. [PMID: 37724245 PMCID: PMC10471113 DOI: 10.1515/mr-2021-0028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/31/2022] [Indexed: 09/20/2023]
Abstract
The tumor ecosystem with heterogeneous cellular compositions and the tumor microenvironment has increasingly become the focus of cancer research in recent years. The extracellular matrix (ECM), the major component of the tumor microenvironment, and its interactions with the tumor cells and stromal cells have also enjoyed tremendously increased attention. Like the other components of the tumor microenvironment, the ECM in solid tumors differs significantly from that in normal organs and tissues. We review recent studies of the complex roles the tumor ECM plays in cancer progression, from tumor initiation, growth to angiogenesis and invasion. We highlight that the biomolecular, biophysical, and mechanochemical interactions between the ECM and cells not only regulate the steps of cancer progression, but also affect the efficacy of systemic cancer treatment. We further discuss the strategies to target and modify the tumor ECM to improve cancer therapy.
Collapse
Affiliation(s)
- Xiuxiu He
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Byoungkoo Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
19
|
Post JN, Loerakker S, Merks R, Carlier A. Implementing computational modeling in tissue engineering: where disciplines meet. Tissue Eng Part A 2022; 28:542-554. [PMID: 35345902 DOI: 10.1089/ten.tea.2021.0215] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In recent years, the mathematical and computational sciences have developed novel methodologies and insights that can aid in designing advanced bioreactors, microfluidic set-ups or organ-on-chip devices, in optimizing culture conditions, or predicting long-term behavior of engineered tissues in vivo. In this review, we introduce the concept of computational models and how they can be integrated in an interdisciplinary workflow for Tissue Engineering and Regenerative Medicine (TERM). We specifically aim this review of general concepts and examples at experimental scientists with little or no computational modeling experience. We also describe the contribution of computational models in understanding TERM processes and in advancing the TERM field by providing novel insights.
Collapse
Affiliation(s)
- Janine Nicole Post
- University of Twente, 3230, Tissue Regeneration, Enschede, Overijssel, Netherlands;
| | - Sandra Loerakker
- Eindhoven University of Technology, 3169, Department of Biomedical Engineering, Eindhoven, Noord-Brabant, Netherlands.,Eindhoven University of Technology, 3169, Institute for Complex Molecular Systems, Eindhoven, Noord-Brabant, Netherlands;
| | - Roeland Merks
- Leiden University, 4496, Institute for Biology Leiden and Mathematical Institute, Leiden, Zuid-Holland, Netherlands;
| | - Aurélie Carlier
- Maastricht University, 5211, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER Maastricht, Maastricht, Netherlands, 6200 MD;
| |
Collapse
|
20
|
Laranjeira S, Pellegrino G, Bhangra KS, Phillips JB, Shipley RJ. In silico framework to inform the design of repair constructs for peripheral nerve injury repair. J R Soc Interface 2022; 19:20210824. [PMID: 35232275 PMCID: PMC8889181 DOI: 10.1098/rsif.2021.0824] [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] [Indexed: 12/22/2022] Open
Abstract
Peripheral nerve injuries affect millions of people per year and cause loss of sensation and muscle control alongside chronic pain. The most severe injuries are treated through a nerve autograft; however, donor site morbidity and poor outcomes mean alternatives are required. One option is to engineer nerve replacement tissues to provide a supportive microenvironment to encourage nerve regeneration as an alternative to nerve grafts. Currently, progress is hampered due to a lack of consensus on how to arrange materials and cells in space to maximize rate of regeneration. This is compounded by a reliance on experimental testing, which precludes extensive investigations of multiple parameters due to time and cost limitations. Here, a computational framework is proposed to simulate the growth of repairing neurites, captured using a random walk approach and parameterized against literature data. The framework is applied to a specific scenario where the engineered tissue comprises a collagen hydrogel with embedded biomaterial fibres. The size and number of fibres are optimized to maximize neurite regrowth, and the robustness of model predictions is tested through sensitivity analyses. The approach provides an in silico tool to inform the design of engineered replacement tissues, with the opportunity for further development to multi-cue environments.
Collapse
Affiliation(s)
- S. Laranjeira
- UCL Mechanical Engineering, London, UK,UCL Centre for Nerve Engineering, UK
| | | | - K. S. Bhangra
- Department of Pharmacology, UCL School of Pharmacy, London, UK,UCL Centre for Nerve Engineering, UK
| | - J. B. Phillips
- Department of Pharmacology, UCL School of Pharmacy, London, UK,UCL Centre for Nerve Engineering, UK
| | - R. J. Shipley
- UCL Mechanical Engineering, London, UK,UCL Centre for Nerve Engineering, UK
| |
Collapse
|
21
|
Hirway SU, Weinberg SH. A review of computational modeling, machine learning and image analysis in cancer metastasis dynamics. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2022. [DOI: 10.1002/cso2.1044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Shreyas U. Hirway
- Department of Biomedical Engineering The Ohio State University Columbus Ohio USA
| | - Seth H. Weinberg
- Department of Biomedical Engineering The Ohio State University Columbus Ohio USA
| |
Collapse
|
22
|
van Steijn L, Wortel IMN, Sire C, Dupré L, Theraulaz G, Merks RMH. Computational modelling of cell motility modes emerging from cell-matrix adhesion dynamics. PLoS Comput Biol 2022; 18:e1009156. [PMID: 35157694 PMCID: PMC8880896 DOI: 10.1371/journal.pcbi.1009156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 02/25/2022] [Accepted: 01/18/2022] [Indexed: 11/18/2022] Open
Abstract
Lymphocytes have been described to perform different motility patterns such as Brownian random walks, persistent random walks, and Lévy walks. Depending on the conditions, such as confinement or the distribution of target cells, either Brownian or Lévy walks lead to more efficient interaction with the targets. The diversity of these motility patterns may be explained by an adaptive response to the surrounding extracellular matrix (ECM). Indeed, depending on the ECM composition, lymphocytes either display a floating motility without attaching to the ECM, or sliding and stepping motility with respectively continuous or discontinuous attachment to the ECM, or pivoting behaviour with sustained attachment to the ECM. Moreover, on the long term, lymphocytes either perform a persistent random walk or a Brownian-like movement depending on the ECM composition. How the ECM affects cell motility is still incompletely understood. Here, we integrate essential mechanistic details of the lymphocyte-matrix adhesions and lymphocyte intrinsic cytoskeletal induced cell propulsion into a Cellular Potts model (CPM). We show that the combination of de novo cell-matrix adhesion formation, adhesion growth and shrinkage, adhesion rupture, and feedback of adhesions onto cell propulsion recapitulates multiple lymphocyte behaviours, for different lymphocyte subsets and various substrates. With an increasing attachment area and increased adhesion strength, the cells’ speed and persistence decreases. Additionally, the model predicts random walks with short-term persistent but long-term subdiffusive properties resulting in a pivoting type of motility. For small adhesion areas, the spatial distribution of adhesions emerges as a key factor influencing cell motility. Small adhesions at the front allow for more persistent motility than larger clusters at the back, despite a similar total adhesion area. In conclusion, we present an integrated framework to simulate the effects of ECM proteins on cell-matrix adhesion dynamics. The model reveals a sufficient set of principles explaining the plasticity of lymphocyte motility. During immunosurveillance, lymphocytes patrol through tissues to interact with cancer cells, other immune cells, and pathogens. The efficiency of this process depends on the kinds of trajectories taken, ranging from simple Brownian walks to Lévy walks. The composition of the extracellular matrix (ECM), a network of macromolecules, affects the formation of cell-matrix adhesions, thus strongly influencing the way lymphocytes move. Here, we present a model of lymphocyte motility driven by adhesions that grow, shrink and rupture in response to the ECM and cellular forces. Compared to other models, our model is computationally light making it suitable for generating long term cell track data, while still capturing actin dynamics and adhesion turnover. Our model suggests that cell motility is affected by the force required to break adhesions and the rate at which new adhesions form. Adhesions can promote cell protrusion by inhibiting retrograde actin flow. After introducing this effect into the model, we found that it reduces the cellular diffusivity and that it promotes stick-slip behaviour. Furthermore, location and size of adhesion clusters determined cell persistence. Overall, our model explains the plasticity of lymphocyte behaviour in response to the ECM.
Collapse
Affiliation(s)
| | - Inge M. N. Wortel
- Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, The Netherlands
| | - Clément Sire
- Laboratoire de Physique Théorique, Centre National de la Recherche Scientifique (CNRS) & Université de Toulouse—Paul Sabatier, Toulouse, France
| | - Loïc Dupré
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Université de Toulouse, Toulouse, France
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Guy Theraulaz
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Centre National de la Recherche Scientifique (CNRS) & Université de Toulouse—Paul Sabatier, Toulouse, France
- Centre for Ecological Sciences, Indian Institute of Science, Bengaluru, India
| | - Roeland M. H. Merks
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
23
|
Villa C, Gerisch A, Chaplain MAJ. A novel nonlocal partial differential equation model of endothelial progenitor cell cluster formation during the early stages of vasculogenesis. J Theor Biol 2022; 534:110963. [PMID: 34838584 DOI: 10.1016/j.jtbi.2021.110963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/03/2021] [Accepted: 11/12/2021] [Indexed: 11/18/2022]
Abstract
The formation of new vascular networks is essential for tissue development and regeneration, in addition to playing a key role in pathological settings such as ischemia and tumour development. Experimental findings in the past two decades have led to the identification of a new mechanism of neovascularisation, known as cluster-based vasculogenesis, during which endothelial progenitor cells (EPCs) mobilised from the bone marrow are capable of bridging distant vascular beds in a variety of hypoxic settings in vivo. This process is characterised by the formation of EPC clusters during its early stages and, while much progress has been made in identifying various mechanisms underlying cluster formation, we are still far from a comprehensive description of such spatio-temporal dynamics. In order to achieve this, we propose a novel mathematical model of the early stages of cluster-based vasculogenesis, comprising of a system of nonlocal partial differential equations including key mechanisms such as endogenous chemotaxis, matrix degradation, cell proliferation and cell-to-cell adhesion. We conduct a linear stability analysis on the system and solve the equations numerically. We then conduct a parametric analysis of the numerical solutions of the one-dimensional problem to investigate the role of underlying dynamics on the speed of cluster formation and the size of clusters, measured via appropriate metrics for the cluster width and compactness. We verify the key results of the parametric analysis with simulations of the two-dimensional problem. Our results, which qualitatively compare with data from in vitro experiments, elucidate the complementary role played by endogenous chemotaxis and matrix degradation in the formation of clusters, suggesting chemotaxis is responsible for the cluster topology while matrix degradation is responsible for the speed of cluster formation. Our results also indicate that the nonlocal cell-to-cell adhesion term in our model, even though it initially causes cells to aggregate, is not sufficient to ensure clusters are stable over long time periods. Consequently, new modelling strategies for cell-to-cell adhesion are required to stabilise in silico clusters. We end the paper with a thorough discussion of promising, fruitful future modelling and experimental research perspectives.
Collapse
Affiliation(s)
- Chiara Villa
- School of Mathematics and Statistics, University of St Andrews, St Andrews KY16 9SS, UK.
| | - Alf Gerisch
- Fachbereich Mathematik, Technische Universität Darmstadt, Dolivostr. 15, 64293 Darmstadt, Germany
| | - Mark A J Chaplain
- School of Mathematics and Statistics, University of St Andrews, St Andrews KY16 9SS, UK
| |
Collapse
|
24
|
Mukhopadhyay D, De R. Growth kinetics and power laws indicate distinct mechanisms of cell-cell interactions in the aggregation process. Biophys J 2022; 121:481-490. [PMID: 34968426 PMCID: PMC8822615 DOI: 10.1016/j.bpj.2021.12.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/20/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023] Open
Abstract
Cellular aggregation is a complex process orchestrated by various kinds of interactions depending on the environment. Different interactions give rise to different pathways of cellular rearrangement and the development of specialized tissues. To distinguish the underlying mechanisms, in this theoretical work, we investigate the spontaneous emergence of tissue patterns from an ensemble of single cells on a substrate following three leading pathways of cell-cell interactions, namely, direct cell adhesion contacts, matrix-mediated mechanical interaction, and chemical signaling. Our analysis shows that the growth kinetics of the aggregation process are distinctly different for each pathway and bear the signature of the specific cell-cell interactions. Interestingly, we find that the average domain size and the mass of the clusters exhibit a power law growth in time under certain interaction mechanisms hitherto unexplored. Further, as observed in experiments, the cluster size distribution can be characterized by stretched exponential functions showing distinct cellular organization processes.
Collapse
Affiliation(s)
- Debangana Mukhopadhyay
- Department of Physical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Rumi De
- Department of Physical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India.
| |
Collapse
|
25
|
Multicellular mechanochemical hybrid cellular Potts model of tissue formation during epithelial‐mesenchymal transition. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2021. [DOI: 10.1002/cso2.1031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
26
|
Yang L, Pijuan-Galito S, Rho HS, Vasilevich AS, Eren AD, Ge L, Habibović P, Alexander MR, de Boer J, Carlier A, van Rijn P, Zhou Q. High-Throughput Methods in the Discovery and Study of Biomaterials and Materiobiology. Chem Rev 2021; 121:4561-4677. [PMID: 33705116 PMCID: PMC8154331 DOI: 10.1021/acs.chemrev.0c00752] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 02/07/2023]
Abstract
The complex interaction of cells with biomaterials (i.e., materiobiology) plays an increasingly pivotal role in the development of novel implants, biomedical devices, and tissue engineering scaffolds to treat diseases, aid in the restoration of bodily functions, construct healthy tissues, or regenerate diseased ones. However, the conventional approaches are incapable of screening the huge amount of potential material parameter combinations to identify the optimal cell responses and involve a combination of serendipity and many series of trial-and-error experiments. For advanced tissue engineering and regenerative medicine, highly efficient and complex bioanalysis platforms are expected to explore the complex interaction of cells with biomaterials using combinatorial approaches that offer desired complex microenvironments during healing, development, and homeostasis. In this review, we first introduce materiobiology and its high-throughput screening (HTS). Then we present an in-depth of the recent progress of 2D/3D HTS platforms (i.e., gradient and microarray) in the principle, preparation, screening for materiobiology, and combination with other advanced technologies. The Compendium for Biomaterial Transcriptomics and high content imaging, computational simulations, and their translation toward commercial and clinical uses are highlighted. In the final section, current challenges and future perspectives are discussed. High-throughput experimentation within the field of materiobiology enables the elucidation of the relationships between biomaterial properties and biological behavior and thereby serves as a potential tool for accelerating the development of high-performance biomaterials.
Collapse
Affiliation(s)
- Liangliang Yang
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sara Pijuan-Galito
- School
of Pharmacy, Biodiscovery Institute, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Hoon Suk Rho
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Aliaksei S. Vasilevich
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aysegul Dede Eren
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lu Ge
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Pamela Habibović
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Morgan R. Alexander
- School
of Pharmacy, Boots Science Building, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Jan de Boer
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aurélie Carlier
- Department
of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick van Rijn
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Qihui Zhou
- Institute
for Translational Medicine, Department of Stomatology, The Affiliated
Hospital of Qingdao University, Qingdao
University, Qingdao 266003, China
| |
Collapse
|
27
|
Bose S, Dasbiswas K, Gopinath A. Matrix Stiffness Modulates Mechanical Interactions and Promotes Contact between Motile Cells. Biomedicines 2021; 9:biomedicines9040428. [PMID: 33920918 PMCID: PMC8077938 DOI: 10.3390/biomedicines9040428] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 02/07/2023] Open
Abstract
The mechanical micro-environment of cells and tissues influences key aspects of cell structure and function, including cell motility. For proper tissue development, cells need to migrate, interact, and form contacts. Cells are known to exert contractile forces on underlying soft substrates and sense deformations in them. Here, we propose and analyze a minimal biophysical model for cell migration and long-range cell–cell interactions through mutual mechanical deformations of the substrate. We compute key metrics of cell motile behavior, such as the number of cell-cell contacts over a given time, the dispersion of cell trajectories, and the probability of permanent cell contact, and analyze how these depend on a cell motility parameter and substrate stiffness. Our results elucidate how cells may sense each other mechanically and generate coordinated movements and provide an extensible framework to further address both mechanical and short-range biophysical interactions.
Collapse
Affiliation(s)
- Subhaya Bose
- Department of Physics, University of California Merced, Merced, CA 95343, USA; (S.B.); (K.D.)
| | - Kinjal Dasbiswas
- Department of Physics, University of California Merced, Merced, CA 95343, USA; (S.B.); (K.D.)
| | - Arvind Gopinath
- Department of Bioengineering, University of California Merced, Merced, CA 95343, USA
- Correspondence:
| |
Collapse
|
28
|
Anomalous Angiogenesis in Retina. Biomedicines 2021; 9:biomedicines9020224. [PMID: 33671578 PMCID: PMC7927046 DOI: 10.3390/biomedicines9020224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/14/2022] Open
Abstract
Age-related macular degeneration (AMD) may cause severe loss of vision or blindness, particularly in elderly people. Exudative AMD is characterized by the angiogenesis of blood vessels growing from underneath the macula, crossing the blood–retina barrier (which comprises Bruch’s membrane (BM) and the retinal pigmentation epithelium (RPE)), leaking blood and fluid into the retina and knocking off photoreceptors. Here, we simulate a computational model of angiogenesis from the choroid blood vessels via a cellular Potts model, as well as BM, RPE cells, drusen deposits and photoreceptors. Our results indicate that improving AMD may require fixing the impaired lateral adhesion between RPE cells and with BM, as well as diminishing Vessel Endothelial Growth Factor (VEGF) and Jagged proteins that affect the Notch signaling pathway. Our numerical simulations suggest that anti-VEGF and anti-Jagged therapies could temporarily halt exudative AMD while addressing impaired cellular adhesion, which could be more effective over a longer time-span.
Collapse
|
29
|
Stepanova D, Byrne HM, Maini PK, Alarcón T. A multiscale model of complex endothelial cell dynamics in early angiogenesis. PLoS Comput Biol 2021; 17:e1008055. [PMID: 33411727 PMCID: PMC7817011 DOI: 10.1371/journal.pcbi.1008055] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 01/20/2021] [Accepted: 11/19/2020] [Indexed: 12/30/2022] Open
Abstract
We introduce a hybrid two-dimensional multiscale model of angiogenesis, the process by which endothelial cells (ECs) migrate from a pre-existing vascular bed in response to local environmental cues and cell-cell interactions, to create a new vascular network. Recent experimental studies have highlighted a central role of cell rearrangements in the formation of angiogenic networks. Our model accounts for this phenomenon via the heterogeneous response of ECs to their microenvironment. These cell rearrangements, in turn, dynamically remodel the local environment. The model reproduces characteristic features of angiogenic sprouting that include branching, chemotactic sensitivity, the brush border effect, and cell mixing. These properties, rather than being hardwired into the model, emerge naturally from the gene expression patterns of individual cells. After calibrating and validating our model against experimental data, we use it to predict how the structure of the vascular network changes as the baseline gene expression levels of the VEGF-Delta-Notch pathway, and the composition of the extracellular environment, vary. In order to investigate the impact of cell rearrangements on the vascular network structure, we introduce the mixing measure, a scalar metric that quantifies cell mixing as the vascular network grows. We calculate the mixing measure for the simulated vascular networks generated by ECs of different lineages (wild type cells and mutant cells with impaired expression of a specific receptor). Our results show that the time evolution of the mixing measure is directly correlated to the generic features of the vascular branching pattern, thus, supporting the hypothesis that cell rearrangements play an essential role in sprouting angiogenesis. Furthermore, we predict that lower cell rearrangement leads to an imbalance between branching and sprout elongation. Since the computation of this statistic requires only individual cell trajectories, it can be computed for networks generated in biological experiments, making it a potential biomarker for pathological angiogenesis. Angiogenesis, the process by which new blood vessels are formed by sprouting from the pre-existing vascular bed, plays a key role in both physiological and pathological processes, including tumour growth. The structure of a growing vascular network is determined by the coordinated behaviour of endothelial cells in response to various signalling cues. Recent experimental studies have highlighted the importance of cell rearrangements as a driver for sprout elongation. However, the functional role of this phenomenon remains unclear. We formulate a new multiscale model of angiogenesis which, by accounting explicitly for the complex dynamics of endothelial cells within growing angiogenic sprouts, is able to reproduce generic features of angiogenic structures (branching, chemotactic sensitivity, cell mixing, etc.) as emergent properties of its dynamics. We validate our model against experimental data and then use it to quantify the phenomenon of cell mixing in vascular networks generated by endothelial cells of different lineages. Our results show that there is a direct correlation between the time evolution of cell mixing in a growing vascular network and its branching structure, thus paving the way for understanding the functional role of cell rearrangements in angiogenesis.
Collapse
Affiliation(s)
- Daria Stepanova
- Centre de Recerca Matemàtica, Bellaterra (Barcelona), Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
- * E-mail:
| | - Helen M. Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Philip K. Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Tomás Alarcón
- Centre de Recerca Matemàtica, Bellaterra (Barcelona), Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Barcelona Graduate School of Mathematics (BGSMath), Barcelona, Spain
| |
Collapse
|
30
|
Scott LE, Griggs LA, Narayanan V, Conway DE, Lemmon CA, Weinberg SH. A hybrid model of intercellular tension and cell-matrix mechanical interactions in a multicellular geometry. Biomech Model Mechanobiol 2020; 19:1997-2013. [PMID: 32193709 PMCID: PMC7502553 DOI: 10.1007/s10237-020-01321-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 03/13/2020] [Indexed: 12/18/2022]
Abstract
Epithelial cells form continuous sheets of cells that exist in tensional homeostasis. Homeostasis is maintained through cell-to-cell junctions that distribute tension and balance forces between cells and their underlying matrix. Disruption of tensional homeostasis can lead to epithelial-mesenchymal transition (EMT), a transdifferentiation process in which epithelial cells adopt a mesenchymal phenotype, losing cell-cell adhesion and enhancing cellular motility. This process is critical during embryogenesis and wound healing, but is also dysregulated in many disease states. To further understand the role of intercellular tension in spatial patterning of epithelial cell monolayers, we developed a multicellular computational model of cell-cell and cell-substrate forces. This work builds on a hybrid cellular Potts model (CPM)-finite element model to evaluate cell-matrix mechanical feedback of an adherent multicellular cluster. Cellular movement is governed by thermodynamic constraints from cell volume, cell-cell and cell-matrix contacts, and durotaxis, which arises from cell-generated traction forces on a finite element substrate. Junction forces at cell-cell contacts balance these traction forces, thereby producing a mechanically stable epithelial monolayer. Simulations were compared to in vitro experiments using fluorescence-based junction force sensors in clusters of cells undergoing EMT. Results indicate that the multicellular CPM model can reproduce many aspects of EMT, including epithelial monolayer formation dynamics, changes in cell geometry, and spatial patterning of cell-cell forces in an epithelial tissue.
Collapse
Affiliation(s)
- Lewis E Scott
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Lauren A Griggs
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Vani Narayanan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Christopher A Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Seth H Weinberg
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
31
|
Rens EG, Merks RM. Cell Shape and Durotaxis Explained from Cell-Extracellular Matrix Forces and Focal Adhesion Dynamics. iScience 2020; 23:101488. [PMID: 32896767 PMCID: PMC7482025 DOI: 10.1016/j.isci.2020.101488] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/13/2020] [Accepted: 08/18/2020] [Indexed: 12/26/2022] Open
Abstract
Many cells are small and rounded on soft extracellular matrices (ECM), elongated on stiffer ECMs, and flattened on hard ECMs. Cells also migrate up stiffness gradients (durotaxis). Using a hybrid cellular Potts and finite-element model extended with ODE-based models of focal adhesion (FA) turnover, we show that the full range of cell shape and durotaxis can be explained in unison from dynamics of FAs, in contrast to previous mathematical models. In our 2D cell-shape model, FAs grow due to cell traction forces. Forces develop faster on stiff ECMs, causing FAs to stabilize and, consequently, cells to spread on stiff ECMs. If ECM stress further stabilizes FAs, cells elongate on substrates of intermediate stiffness. We show that durotaxis follows from the same set of assumptions. Our model contributes to the understanding of the basic responses of cells to ECM stiffness, paving the way for future modeling of more complex cell-ECM interactions.
Collapse
Affiliation(s)
- Elisabeth G. Rens
- Scientific Computing, CWI, Science Park 123, 1098 XG Amsterdam, the Netherlands
- Mathematics Department, University of British Columbia, Mathematics Road 1984, Vancouver, BC V6T 1Z2, Canada
| | - Roeland M.H. Merks
- Scientific Computing, CWI, Science Park 123, 1098 XG Amsterdam, the Netherlands
- Mathematical Institute, Leiden University, Niels Bohrweg 1, 2333 CA Leiden, the Netherlands
| |
Collapse
|
32
|
Rens EG, Zeegers MT, Rabbers I, Szabó A, Merks RMH. Autocrine inhibition of cell motility can drive epithelial branching morphogenesis in the absence of growth. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190386. [PMID: 32713299 DOI: 10.1098/rstb.2019.0386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Epithelial branching morphogenesis drives the development of organs such as the lung, salivary gland, kidney and the mammary gland. It involves cell proliferation, cell differentiation and cell migration. An elaborate network of chemical and mechanical signals between the epithelium and the surrounding mesenchymal tissues regulates the formation and growth of branching organs. Surprisingly, when cultured in isolation from mesenchymal tissues, many epithelial tissues retain the ability to exhibit branching morphogenesis even in the absence of proliferation. In this work, we propose a simple, experimentally plausible mechanism that can drive branching morphogenesis in the absence of proliferation and cross-talk with the surrounding mesenchymal tissue. The assumptions of our mathematical model derive from in vitro observations of the behaviour of mammary epithelial cells. These data show that autocrine secretion of the growth factor TGF[Formula: see text]1 inhibits the formation of cell protrusions, leading to curvature-dependent inhibition of sprouting. Our hybrid cellular Potts and partial-differential equation model correctly reproduces the experimentally observed tissue-geometry-dependent determination of the sites of branching, and it suffices for the formation of self-avoiding branching structures in the absence and also in the presence of cell proliferation. This article is part of the theme issue 'Multi-scale analysis and modelling of collective migration in biological systems'.
Collapse
Affiliation(s)
- Elisabeth G Rens
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands.,Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - Mathé T Zeegers
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - Iraes Rabbers
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - András Szabó
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - Roeland M H Merks
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands.,Mathematical Institute, Leiden University, Leiden, The Netherlands
| |
Collapse
|
33
|
Schakenraad K, Ernst J, Pomp W, Danen EHJ, Merks RMH, Schmidt T, Giomi L. Mechanical interplay between cell shape and actin cytoskeleton organization. SOFT MATTER 2020; 16:6328-6343. [PMID: 32490503 DOI: 10.1039/d0sm00492h] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We investigate the mechanical interplay between the spatial organization of the actin cytoskeleton and the shape of animal cells adhering on micropillar arrays. Using a combination of analytical work, computer simulations and in vitro experiments, we demonstrate that the orientation of the stress fibers strongly influences the geometry of the cell edge. In the presence of a uniformly aligned cytoskeleton, the cell edge can be well approximated by elliptical arcs, whose eccentricity reflects the degree of anisotropy of the cell's internal stresses. Upon modeling the actin cytoskeleton as a nematic liquid crystal, we further show that the geometry of the cell edge feeds back on the organization of the stress fibers by altering the length scale at which these are confined. This feedback mechanism is controlled by a dimensionless number, the anchoring number, representing the relative weight of surface-anchoring and bulk-aligning torques. Our model allows to predict both cellular shape and the internal structure of the actin cytoskeleton and is in good quantitative agreement with experiments on fibroblastoid (GDβ1, GDβ3) and epithelioid (GEβ1, GEβ3) cells.
Collapse
Affiliation(s)
- Koen Schakenraad
- Instituut-Lorentz, Leiden University, P.O. Box 9506, 2300 RA Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
34
|
Li X, Ni Q, He X, Kong J, Lim SM, Papoian GA, Trzeciakowski JP, Trache A, Jiang Y. Tensile force-induced cytoskeletal remodeling: Mechanics before chemistry. PLoS Comput Biol 2020; 16:e1007693. [PMID: 32520928 PMCID: PMC7326277 DOI: 10.1371/journal.pcbi.1007693] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/30/2020] [Accepted: 04/21/2020] [Indexed: 12/31/2022] Open
Abstract
Understanding cellular remodeling in response to mechanical stimuli is a critical step in elucidating mechanical activation of biochemical signaling pathways. Experimental evidence indicates that external stress-induced subcellular adaptation is accomplished through dynamic cytoskeletal reorganization. To study the interactions between subcellular structures involved in transducing mechanical signals, we combined experimental data and computational simulations to evaluate real-time mechanical adaptation of the actin cytoskeletal network. Actin cytoskeleton was imaged at the same time as an external tensile force was applied to live vascular smooth muscle cells using a fibronectin-functionalized atomic force microscope probe. Moreover, we performed computational simulations of active cytoskeletal networks under an external tensile force. The experimental data and simulation results suggest that mechanical structural adaptation occurs before chemical adaptation during filament bundle formation: actin filaments first align in the direction of the external force by initializing anisotropic filament orientations, then the chemical evolution of the network follows the anisotropic structures to further develop the bundle-like geometry. Our findings present an alternative two-step explanation for the formation of actin bundles due to mechanical stimulation and provide new insights into the mechanism of mechanotransduction. Remodeling the cytoskeletal network in response to external force is key to cellular mechanotransduction. Despite much focus on cytoskeletal remodeling in recent years, a comprehensive understanding of actin remodeling in real-time in cells under mechanical stimuli is still lacking. We integrated tensile stress-induced 3D actin remodeling and 3D computational simulations of actin cytoskeleton to study how the actin cytoskeleton form bundles and how these bundles evolve over time upon external tensile stress. We found that actin network remodels through a two-step process in which rapid alignment of actin filaments is followed by slower actin bundling. Based on these results, we propose a “mechanics before chemistry” model of actin cytoskeleton remodeling under external tensile force.
Collapse
Affiliation(s)
- Xiaona Li
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia, United States of America
| | - Qin Ni
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland, United States of America
| | - Xiuxiu He
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia, United States of America
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia, United States of America
| | - Soon-Mi Lim
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Garegin A. Papoian
- Department of Chemistry & Biochemistry, University of Maryland, College Park, Maryland, United States of America
| | - Jerome P. Trzeciakowski
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Andreea Trache
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, Texas, United States of America
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
35
|
Malik AA, Wennberg B, Gerlee P. The Impact of Elastic Deformations of the Extracellular Matrix on Cell Migration. Bull Math Biol 2020; 82:49. [PMID: 32248312 PMCID: PMC7128007 DOI: 10.1007/s11538-020-00721-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 03/15/2020] [Indexed: 01/06/2023]
Abstract
The mechanical properties of the extracellular matrix, in particular its stiffness, are known to impact cell migration. In this paper, we develop a mathematical model of a single cell migrating on an elastic matrix, which accounts for the deformation of the matrix induced by forces exerted by the cell, and investigate how the stiffness impacts the direction and speed of migration. We model a cell in 1D as a nucleus connected to a number of adhesion sites through elastic springs. The cell migrates by randomly updating the position of its adhesion sites. We start by investigating the case where the cell springs are constant, and then go on to assuming that they depend on the matrix stiffness, on matrices of both uniform stiffness as well as those with a stiffness gradient. We find that the assumption that cell springs depend on the substrate stiffness is necessary and sufficient for an efficient durotactic response. We compare simulations to recent experimental observations of human cancer cells exhibiting durotaxis, which show good qualitative agreement.
Collapse
Affiliation(s)
- A A Malik
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, 412 96, Gothenburg, Sweden.
| | - B Wennberg
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, 412 96, Gothenburg, Sweden
| | - P Gerlee
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, 412 96, Gothenburg, Sweden
| |
Collapse
|
36
|
Alblawi A, Ranjani AS, Yasmin H, Gupta S, Bit A, Rahimi-Gorji M. Scaffold-free: A developing technique in field of tissue engineering. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 185:105148. [PMID: 31678793 DOI: 10.1016/j.cmpb.2019.105148] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/17/2019] [Accepted: 10/20/2019] [Indexed: 06/10/2023]
Abstract
Scaffold-free tissue engineering can be considered as a rapidly developing technique in the field of tissue engineering. In the areas of regenerative medicine and wound healing, there is a demand of techniques where no scaffolds are used for the development of desired tissue. These techniques will overcome the problems of rejection and tissue failure which are common with scaffolds. Main breakthrough of scaffold free tissue engineering was after invention of 3-D printers which made it possible to print complex tissues which were not possible by conventional methods. Mathematical modeling is a prediction technique is used in tissue engineering for simulation of the model to be constructed. Coming to scaffold-free technique, mathematical modeling is necessary for the processing of the model that has to be bio-printed so as to avoid and changes in the final construct. Tissue construct is developed by use of non-destructive imaging techniques i.e. computed tomography (CT) and magnetic resonance imaging (MRI).In this review, we discussed about various mathematical models and the models which are widely used in bioprinting techniques such as Cellular Potts Model (CPM) and Cellular Particle Dynamic (CPD) model. Later, developed of 3-D tissue construct using micro CT scan images was explained. Finally, we discussed about scaffold free techniques such as 3-D bioprinting and cell sheet technology. In this manuscript, we proposed a cell sheet based bioprinting technique where mesenchymal stem cells (MSCs) on the surface of thermoresponsive polymer were subjected to mechanosensing either by introducing acoustic energies or stress created by polymeric strain energy function. Mechanosensing stimulus will trigger the intracellular signal transduction pathway leading to differentiation of the MSCs into desired cells.
Collapse
Affiliation(s)
- Adel Alblawi
- Mechanical Engineering Department, College of Engineering, Shaqra University, Dawadmi, P.O. 11911, Ar Riyadh, Saudi Arabia.
| | - Achalla Sri Ranjani
- Department of Biomedical Engineering, National Institute of Technology, Raipur, India
| | - Humaira Yasmin
- Department of Mathematics, College of Science, Majmaah University, 11952, Saudi Arabia.
| | - Sharda Gupta
- Department of Biomedical Engineering, National Institute of Technology, Raipur, India
| | - Arindam Bit
- Department of Biomedical Engineering, National Institute of Technology, Raipur, India.
| | - Mohammad Rahimi-Gorji
- Faculty of Medicine and Health Science, Ghent University, 9000 Gent, Belgium; Biofluid, Tissue and Solid Mechanics for Medical Applications Lab (IBiTech, bioMMeda), Ghent University, Gent 9000, Belgium.
| |
Collapse
|
37
|
Nakano T, Okaie Y, Kinugasa Y, Koujin T, Suda T, Hiraoka Y, Haraguchi T. Roles of Remote and Contact Forces in Epithelial Cell Structure Formation. Biophys J 2020; 118:1466-1478. [PMID: 32097624 PMCID: PMC7091513 DOI: 10.1016/j.bpj.2020.01.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/25/2020] [Accepted: 01/29/2020] [Indexed: 12/28/2022] Open
Abstract
Cancer cells collectively form a large-scale structure for their growth. In this article, we report that HeLa cells, epithelial-like human cervical cancer cells, aggressively migrate on Matrigel and form a large-scale structure in a cell-density-dependent manner. To explain the experimental results, we develop a simple model in which cells interact and migrate using the two fundamentally different types of force, remote and contact forces, and show how cells form a large-scale structure. We demonstrate that the simple model reproduces experimental observations, suggesting that the remote and contact forces considered in this work play a major role in large-scale structure formation of HeLa cells. This article provides important evidence that cancer cells form a large-scale structure and develops an understanding into the poorly understood mechanisms of their structure formation.
Collapse
Affiliation(s)
- Tadashi Nakano
- Institute for Datability Science, Osaka University, Suita, Japan.
| | - Yutaka Okaie
- Institute for Datability Science, Osaka University, Suita, Japan
| | - Yasuha Kinugasa
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Takako Koujin
- Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | | | - Yasushi Hiraoka
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Tokuko Haraguchi
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| |
Collapse
|
38
|
Notch signaling and taxis mechanisms regulate early stage angiogenesis: A mathematical and computational model. PLoS Comput Biol 2020; 16:e1006919. [PMID: 31986145 PMCID: PMC7021322 DOI: 10.1371/journal.pcbi.1006919] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 02/14/2020] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
Abstract
During angiogenesis, new blood vessels sprout and grow from existing ones. This process plays a crucial role in organ development and repair, in wound healing and in numerous pathological processes such as cancer progression or diabetes. Here, we present a mathematical model of early stage angiogenesis that permits exploration of the relative importance of mechanical, chemical and cellular cues. Endothelial cells proliferate and move over an extracellular matrix by following external gradients of Vessel Endothelial Growth Factor, adhesion and stiffness, which are incorporated to a Cellular Potts model with a finite element description of elasticity. The dynamics of Notch signaling involving Delta-4 and Jagged-1 ligands determines tip cell selection and vessel branching. Through their production rates, competing Jagged-Notch and Delta-Notch dynamics determine the influence of lateral inhibition and lateral induction on the selection of cellular phenotypes, branching of blood vessels, anastomosis (fusion of blood vessels) and angiogenesis velocity. Anastomosis may be favored or impeded depending on the mechanical configuration of strain vectors in the ECM near tip cells. Numerical simulations demonstrate that increasing Jagged production results in pathological vasculatures with thinner and more abundant vessels, which can be compensated by augmenting the production of Delta ligands. Angiogenesis is the process by which new blood vessels grow from existing ones. This process plays a crucial role in organ development, in wound healing and in numerous pathological processes such as cancer growth or in diabetes. Angiogenesis is a complex, multi-step and well regulated process where biochemistry and physics are intertwined. The process entails signaling in vessel cells being driven by both chemical and mechanical mechanisms that result in vascular cell movement, deformation and proliferation. Mathematical models have the ability to bring together these mechanisms in order to explore their relative relevance in vessel growth. Here, we present a mathematical model of early stage angiogenesis that is able to explore the role of biochemical signaling and tissue mechanics. We use this model to unravel the regulating role of Jagged, Notch and Delta dynamics in vascular cells. These membrane proteins have an important part in determining the leading cell in each neo-vascular sprout. Numerical simulations demonstrate that increasing Jagged production results in pathological vasculatures with thinner and more abundant vessels, which can be compensated by augmenting the production of Delta ligands.
Collapse
|
39
|
Rens EG, Edelstein-Keshet L. From energy to cellular forces in the Cellular Potts Model: An algorithmic approach. PLoS Comput Biol 2019; 15:e1007459. [PMID: 31825952 PMCID: PMC6927661 DOI: 10.1371/journal.pcbi.1007459] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 12/23/2019] [Accepted: 10/05/2019] [Indexed: 11/30/2022] Open
Abstract
Single and collective cell dynamics, cell shape changes, and cell migration can be conveniently represented by the Cellular Potts Model, a computational platform based on minimization of a Hamiltonian. Using the fact that a force field is easily derived from a scalar energy (F = −∇H), we develop a simple algorithm to associate effective forces with cell shapes in the CPM. We predict the traction forces exerted by single cells of various shapes and sizes on a 2D substrate. While CPM forces are specified directly from the Hamiltonian on the cell perimeter, we approximate the force field inside the cell domain using interpolation, and refine the results with smoothing. Predicted forces compare favorably with experimentally measured cellular traction forces. We show that a CPM model with internal signaling (such as Rho-GTPase-related contractility) can be associated with retraction-protrusion forces that accompany cell shape changes and migration. We adapt the computations to multicellular systems, showing, for example, the forces that a pair of swirling cells exert on one another, demonstrating that our algorithm works equally well for interacting cells. Finally, we show forces exerted by cells on one another in classic cell-sorting experiments. Cells exert forces on their surroundings and on one another. In simulations of cell shape using the Cellular Potts Model (CPM), the dynamics of deforming cell shapes is traditionally represented by an energy-minimization method. We use this CPM energy, the Hamiltonian, to derive and visualize the corresponding forces exerted by the cells. We use the fact that force is the negative gradient of energy to assign forces to the CPM cell edges, and then extend the results to approximate interior forces by interpolation. We show that this method works for single as well as multiple interacting model cells, both static and motile. Finally, we show favorable comparison between predicted forces and real forces measured experimentally.
Collapse
Affiliation(s)
- Elisabeth G. Rens
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leah Edelstein-Keshet
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
40
|
Thüroff F, Goychuk A, Reiter M, Frey E. Bridging the gap between single-cell migration and collective dynamics. eLife 2019; 8:e46842. [PMID: 31808744 PMCID: PMC6992385 DOI: 10.7554/elife.46842] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 12/06/2019] [Indexed: 11/13/2022] Open
Abstract
Motivated by the wealth of experimental data recently available, we present a cellular-automaton-based modeling framework focussing on high-level cell functions and their concerted effect on cellular migration patterns. Specifically, we formulate a coarse-grained description of cell polarity through self-regulated actin organization and its response to mechanical cues. Furthermore, we address the impact of cell adhesion on collective migration in cell cohorts. The model faithfully reproduces typical cell shapes and movements down to the level of single cells, yet allows for the efficient simulation of confluent tissues. In confined circular geometries, we find that specific properties of individual cells (polarizability; contractility) influence the emerging collective motion of small cell cohorts. Finally, we study the properties of expanding cellular monolayers (front morphology; stress and velocity distributions) at the level of extended tissues.
Collapse
Affiliation(s)
- Florian Thüroff
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| | - Andriy Goychuk
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| | - Matthias Reiter
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| | - Erwin Frey
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| |
Collapse
|
41
|
Van Liedekerke P, Neitsch J, Johann T, Warmt E, Gonzàlez-Valverde I, Hoehme S, Grosser S, Kaes J, Drasdo D. A quantitative high-resolution computational mechanics cell model for growing and regenerating tissues. Biomech Model Mechanobiol 2019; 19:189-220. [PMID: 31749071 PMCID: PMC7005086 DOI: 10.1007/s10237-019-01204-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 07/16/2019] [Indexed: 12/19/2022]
Abstract
Mathematical models are increasingly designed to guide experiments in biology, biotechnology, as well as to assist in medical decision making. They are in particular important to understand emergent collective cell behavior. For this purpose, the models, despite still abstractions of reality, need to be quantitative in all aspects relevant for the question of interest. This paper considers as showcase example the regeneration of liver after drug-induced depletion of hepatocytes, in which the surviving and dividing hepatocytes must squeeze in between the blood vessels of a network to refill the emerged lesions. Here, the cells' response to mechanical stress might significantly impact the regeneration process. We present a 3D high-resolution cell-based model integrating information from measurements in order to obtain a refined and quantitative understanding of the impact of cell-biomechanical effects on the closure of drug-induced lesions in liver. Our model represents each cell individually and is constructed by a discrete, physically scalable network of viscoelastic elements, capable of mimicking realistic cell deformation and supplying information at subcellular scales. The cells have the capability to migrate, grow, and divide, and the nature and parameters of their mechanical elements can be inferred from comparisons with optical stretcher experiments. Due to triangulation of the cell surface, interactions of cells with arbitrarily shaped (triangulated) structures such as blood vessels can be captured naturally. Comparing our simulations with those of so-called center-based models, in which cells have a largely rigid shape and forces are exerted between cell centers, we find that the migration forces a cell needs to exert on its environment to close a tissue lesion, is much smaller than predicted by center-based models. To stress generality of the approach, the liver simulations were complemented by monolayer and multicellular spheroid growth simulations. In summary, our model can give quantitative insight in many tissue organization processes, permits hypothesis testing in silico, and guide experiments in situations in which cell mechanics is considered important.
Collapse
Affiliation(s)
- Paul Van Liedekerke
- Inria Paris & Sorbonne Université LJLL, 2 Rue Simone IFF, 75012, Paris, France. .,IfADo - Leibniz Research Centre for Working Environment and Human Factors, Ardeystrasse 67, Dortmund, Germany.
| | - Johannes Neitsch
- Interdisciplinary Centre for Bioinformatics, Leipzig University, Härtelstr. 16-18, 04107, Leipzig, Germany
| | - Tim Johann
- IfADo - Leibniz Research Centre for Working Environment and Human Factors, Ardeystrasse 67, Dortmund, Germany
| | - Enrico Warmt
- Faculty of Physics and Earth Science, Peter Debye Institute for Soft Matter Physics, Leipzig University, Linnéstraße 5, 04103, Leipzig, Germany
| | | | - Stefan Hoehme
- Interdisciplinary Centre for Bioinformatics, Leipzig University, Härtelstr. 16-18, 04107, Leipzig, Germany.,Institute for Computer Science, Leipzig University, Härtelstr. 16-18, 04107, Leipzig, Germany
| | - Steffen Grosser
- Faculty of Physics and Earth Science, Peter Debye Institute for Soft Matter Physics, Leipzig University, Linnéstraße 5, 04103, Leipzig, Germany
| | - Josef Kaes
- Faculty of Physics and Earth Science, Peter Debye Institute for Soft Matter Physics, Leipzig University, Linnéstraße 5, 04103, Leipzig, Germany
| | - Dirk Drasdo
- Inria Paris & Sorbonne Université LJLL, 2 Rue Simone IFF, 75012, Paris, France. .,IfADo - Leibniz Research Centre for Working Environment and Human Factors, Ardeystrasse 67, Dortmund, Germany. .,Interdisciplinary Centre for Bioinformatics, Leipzig University, Härtelstr. 16-18, 04107, Leipzig, Germany.
| |
Collapse
|
42
|
Akbarpour Ghazani M, Nouri Z, Saghafian M, Soltani M. Mathematical modeling reveals how the density of initial tumor and its distance to parent vessels alter the growth trend of vascular tumors. Microcirculation 2019; 27:e12584. [DOI: 10.1111/micc.12584] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 06/10/2019] [Accepted: 08/05/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Mehran Akbarpour Ghazani
- Department of Mechanical Engineering Isfahan University of Technology Isfahan Iran
- Faculty of Mechanical Engineering University of Tabriz Tabriz Iran
| | - Zahra Nouri
- Department of Mechanical Engineering Isfahan University of Technology Isfahan Iran
| | - Mohsen Saghafian
- Department of Mechanical Engineering Isfahan University of Technology Isfahan Iran
| | - Madjid Soltani
- Department of Mechanical Engineering K.N. Toosi University of Technology Tehran Iran
- Advanced Bioengineering Initiative Center Computational Medicine Center K. N. Toosi University of Technology Tehran Iran
- Cancer Biology Research Center Cancer Institute of Iran Tehran University of Medical Sciences Tehran Iran
- Centre for Biotechnology and Bioengineering (CBB) University of Waterloo Waterloo ON Canada
- Department of Electrical and Computer Engineering University of Waterloo Waterloo ON Canada
| |
Collapse
|
43
|
Yuan Y, Basu S, Lin MH, Shukla S, Sarkar D. Colloidal Gels for Guiding Endothelial Cell Organization via Microstructural Morphology. ACS APPLIED MATERIALS & INTERFACES 2019; 11:31709-31728. [PMID: 31403768 PMCID: PMC7219539 DOI: 10.1021/acsami.9b11293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
One of the fundamental challenges in vascular morphogenesis is to understand how the microstructural morphology of a 3D matrix can provide the spatial cues to organize the endothelial cells (ECs) into specific vascular structures. Colloidal gels can provide well-controlled distinct morphological matrices because these gels are formed by the aggregation of particles. By altering the aggregation mode, the spatial organization of the particles can be controlled to yield different microstructural morphology. To demonstrate this, colloidal aggregates and gels were developed by electrostatic interaction-mediated aggregation of cationic polyurethane (PU) colloidal particles by using low molecular weight electrolyte and polyelectrolyte to develop microstructurally different colloidal gels without altering their bulk elasticity. Compact dense colloidal aggregates with constricted voids were developed via electrolyte-mediated aggregation, whereas stranded branched networks with interconnected voids were formed via polyelectrolyte-mediated bridging interactions. Results show that the microstructure of aggregated colloids and gels can regulate EC organizations. Within endothelial matrices, ECs track the microstructure of particulate phase to interconnect with stranded colloidal network but cluster around compact colloidal aggregate. Similarly, in colloidal gels, ECs formed capillary-like structures by interconnecting along the stranded networks with enhanced cell-matrix interactions and increased cell extension but aggregated within the constricted voids of compact dense gel with enhanced cell-cell interaction. Both morphometric analysis and expression of EC markers corroborated the cell organizations in these gels. Using these colloidal gels, we demonstrated the role of 3D microstructural morphology as an important regulator for spatial guidance of ECs and simultaneously established the significance of colloidal gels as 3D matrix to regulate cellular morphogenesis.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Sukanya Basu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Meng Huisan Lin
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Shruti Shukla
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Debanjan Sarkar
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
- Correspondence to: D. Sarkar. Biomedical Engineering, University at Buffalo, Ph: 716-645-8497, Fax: 716-645-2207,
| |
Collapse
|
44
|
Soft culture substrates favor stem-like cellular phenotype and facilitate reprogramming of human mesenchymal stem/stromal cells (hMSCs) through mechanotransduction. Sci Rep 2019; 9:9086. [PMID: 31235788 PMCID: PMC6591285 DOI: 10.1038/s41598-019-45352-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 06/04/2019] [Indexed: 01/26/2023] Open
Abstract
Biophysical cues influence many aspects of cell behavior. Stiffness of the extracellular matrix is probed by cells and transduced into biochemical signals through mechanotransduction protein networks, strongly influencing stem cell behavior. Cellular stemness is intimately related with mechanical properties of the cell, like intracellular contractility and stiffness, which in turn are influenced by the microenvironment. Pluripotency is associated with soft and low-contractility cells. Hence, we postulated that soft cell culture substrates, presumably inducing low cellular contractility and stiffness, increase the reprogramming efficiency of mesenchymal stem/stromal cells (MSCs) into induced pluripotent stem cells (iPSCs). We demonstrate that soft substrates (1.5 or 15 kPa polydimethylsiloxane – PDMS) caused modulation of several cellular features of MSCs into a phenotype closer to pluripotent stem cells (PSCs). MSCs cultured on soft substrates presented more relaxed nuclei, lower maturation of focal adhesions and F-actin assembling, more euchromatic and less heterochromatic nuclear DNA regions, and increased expression of pluripotency-related genes. These changes correlate with the reprogramming of MSCs, with a positive impact on the kinetics, robustness of colony formation and reprogramming efficiency. Additionally, substrate stiffness influences several phenotypic features of iPS cells and colonies, and data indicates that soft substrates favor full iPSC reprogramming.
Collapse
|
45
|
Griffin BP, Largaespada CJ, Rinaldi NA, Lemmon CA. A novel method for quantifying traction forces on hexagonal micropatterned protein features on deformable poly-dimethyl siloxane sheets. MethodsX 2019; 6:1343-1352. [PMID: 31417850 PMCID: PMC6690417 DOI: 10.1016/j.mex.2019.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 05/09/2019] [Indexed: 11/24/2022] Open
Abstract
Many methods exist for quantifying cellular traction forces, including traction force microscopy and microfabricated post arrays. However, these methodologies have limitations, including a requirement to remove cells to determine undeflected particle locations and the inability to quantify forces of cells with low cytoskeletal stiffness, respectively. Here we present a novel method of traction force quantification that eliminates both of these limitations. Through the use of a hexagonal pattern of microcontact-printed protein spots, a novel computational algorithm, and thin surfaces of polydimethyl siloxane (PDMS) blends, we demonstrate a system that: •quantifies cellular forces on a homogeneous surface that is stable and easily manufactured.•utilizes hexagonal patterns of protein spots and computational geometry to quantify cellular forces without need for cell removal.•quantifies cellular forces in cells with low cytoskeletal rigidity.
Collapse
Affiliation(s)
- Brian P. Griffin
- Department of Biomedical Engineering, Virginia Commonwealth University, United States
| | | | - Nicole A. Rinaldi
- Department of Biomedical Engineering, University of Rochester, United States
| | - Christopher A. Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, United States
| |
Collapse
|
46
|
Bui J, Conway DE, Heise RL, Weinberg SH. Mechanochemical Coupling and Junctional Forces during Collective Cell Migration. Biophys J 2019; 117:170-183. [PMID: 31200935 DOI: 10.1016/j.bpj.2019.05.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/09/2019] [Accepted: 05/22/2019] [Indexed: 12/31/2022] Open
Abstract
Cell migration, a fundamental physiological process in which cells sense and move through their surrounding physical environment, plays a critical role in development and tissue formation, as well as pathological processes, such as cancer metastasis and wound healing. During cell migration, dynamics are governed by the bidirectional interplay between cell-generated mechanical forces and the activity of Rho GTPases, a family of small GTP-binding proteins that regulate actin cytoskeleton assembly and cellular contractility. These interactions are inherently more complex during the collective migration of mechanically coupled cells because of the additional regulation of cell-cell junctional forces. In this study, we adapted a recent minimal modeling framework to simulate the interactions between mechanochemical signaling in individual cells and interactions with cell-cell junctional forces during collective cell migration. We find that migration of individual cells depends on the feedback between mechanical tension and Rho GTPase activity in a biphasic manner. During collective cell migration, waves of Rho GTPase activity mediate mechanical contraction/extension and thus synchronization throughout the tissue. Further, cell-cell junctional forces exhibit distinct spatial patterns during collective cell migration, with larger forces near the leading edge. Larger junctional force magnitudes are associated with faster collective cell migration and larger tissue size. Simulations of heterogeneous tissue migration exhibit a complex dependence on the properties of both leading and trailing cells. Computational predictions demonstrate that collective cell migration depends on both the emergent dynamics and interactions between cellular-level Rho GTPase activity and contractility and multicellular-level junctional forces.
Collapse
Affiliation(s)
- Justin Bui
- Department of Chemical Engineering, University of California Berkeley, Berkeley, California
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Seth H Weinberg
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
47
|
Malik AA, Gerlee P. Mathematical modelling of cell migration: stiffness dependent jump rates result in durotaxis. J Math Biol 2019; 78:2289-2315. [PMID: 30972438 PMCID: PMC6534528 DOI: 10.1007/s00285-019-01344-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 01/25/2019] [Indexed: 12/17/2022]
Abstract
Durotaxis, the phenomena where cells migrate up a gradient in substrate stiffness, remains poorly understood. It has been proposed that durotaxis results from the reinforcement of focal adhesions on stiff substrates. In this paper we formulate a mathematical model of single cell migration on elastic substrates with spatially varying stiffness. We develop a stochastic model where the cell moves by updating the position of its adhesion sites at random times, and the rate of updates is determined by the local stiffness of the substrate. We investigate two physiologically motivated mechanisms of stiffness sensing. From the stochastic model of single cell migration we derive a population level description in the form of a partial differential equation for the time evolution of the density of cells. The equation is an advection–diffusion equation, where the advective velocity is proportional to the stiffness gradient. The model shows quantitative agreement with experimental results in which cells tend to cluster when seeded on a matrix with periodically varying stiffness.
Collapse
Affiliation(s)
- Adam A Malik
- Mathematical Sciences, Chalmers University of Technology, 41296, Göteborg, Sweden. .,Mathematical Sciences, University of Gothenburg, 41296, Göteborg, Sweden.
| | - Philip Gerlee
- Mathematical Sciences, Chalmers University of Technology, 41296, Göteborg, Sweden.,Mathematical Sciences, University of Gothenburg, 41296, Göteborg, Sweden
| |
Collapse
|
48
|
Kühn C, Checa S. Computational Modeling to Quantify the Contributions of VEGFR1, VEGFR2, and Lateral Inhibition in Sprouting Angiogenesis. Front Physiol 2019; 10:288. [PMID: 30971939 PMCID: PMC6445957 DOI: 10.3389/fphys.2019.00288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 03/05/2019] [Indexed: 12/25/2022] Open
Abstract
Sprouting angiogenesis is a necessary process in regeneration and development as well as in tumorigenesis. VEGF-A is the main pro-angiogenic chemoattractant and it can bind to the decoy receptor VEGFR1 or to VEGFR2 to induce sprouting. Active sprout cells express Dll4, which binds to Notch1 on neighboring cells, in turn inhibiting VEGFR2 expression. It is known that the balance between VEGFR2 and VEGFR1 determines tip selection and network architecture, however the quantitative interrelationship of the receptors and their interrelated balances, also with relation to Dll4-Notch1 signaling, remains yet largely unknown. Here, we present an agent-based computer model of sprouting angiogenesis, integrating VEGFR1 and VEGFR2 in a detailed model of cellular signaling. Our model reproduces experimental data on VEGFR1 knockout. We show that soluble VEGFR1 improves the efficiency of angiogenesis by directing sprouts away from existing cells over a wide range of parameters. Our analysis unravels the relevance of the stability of the active notch intracellular domain as a dominating hub in this regulatory network. Our analysis quantitatively dissects the regulatory interactions in sprouting angiogenesis. Because we use a detailed model of intracellular signaling, the results of our analysis are directly linked to biological entities. We provide our computational model and simulation engine for integration in complementary modeling approaches.
Collapse
Affiliation(s)
- Clemens Kühn
- Julius Wolff Institute, Charite - Universitätsmedizin Berlin, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute, Charite - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charite - UIniversitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
49
|
Palmieri B, Scanlon C, Worroll D, Grant M, Lee J. Substrate mediated interaction between pairs of keratocytes: Multipole traction force models describe their migratory behavior. PLoS One 2019; 14:e0212162. [PMID: 30822310 PMCID: PMC6396918 DOI: 10.1371/journal.pone.0212162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/29/2019] [Indexed: 11/18/2022] Open
Abstract
A series of traction force microscopy experiments involving pairs of keratocytes migrating on compliant substrates were analyzed. We observed several instances where keratocytes that are about to collide turn before they touch. We term this phenomenon collision avoidance behavior and we propose that the turning is caused by the substrate mediated elastic interactions between the cells. A multipole analysis of the cell traction reveals that the left-right symmetry of the keratocyte traction pattern is broken during collision avoidance events. The analysis further shows that the cell migration direction reorients before the principal traction dipoles as the cells turn. Linear elasticity theory is used to derive the cell-cell interaction energy between pairs of keratocytes. The traction force applied by each cell is modeled as a two points (dipole) or three points (tripod) force model. We show that both models predict that cells that are about to collide in a head-on manner will turn before touching. The tripod model is further able to account for the quadrupole components of the traction force profile that we observed experimentally. Also, the tripod model proposes a mechanism that may explain why cells tend to scatter with a finite angle after a collision avoidance event. A relationship between the scattering angle and the traction force quadrupole moment is also established. Dynamical simulations of migrating model cells are further used to explain the emergence of other cell pair trajectories that we observed experimentally.
Collapse
Affiliation(s)
- Benoit Palmieri
- Department of Physics, McGill University, Montréal, Québec, Canada
| | - Christine Scanlon
- Department of Molecular & Cell Biology, Storrs, CT, United States of America
| | - Daniel Worroll
- Department of Molecular & Cell Biology, Storrs, CT, United States of America
| | - Martin Grant
- Department of Physics, McGill University, Montréal, Québec, Canada
| | - Juliet Lee
- Department of Molecular & Cell Biology, Storrs, CT, United States of America
- * E-mail:
| |
Collapse
|
50
|
Schwager SC, Taufalele PV, Reinhart-King CA. Cell-Cell Mechanical Communication in Cancer. Cell Mol Bioeng 2019; 12:1-14. [PMID: 31565083 PMCID: PMC6764766 DOI: 10.1007/s12195-018-00564-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Communication between cancer cells enables cancer progression and metastasis. While cell-cell communication in cancer has primarily been examined through chemical mechanisms, recent evidence suggests that mechanical communication through cell-cell junctions and cell-ECM linkages is also an important mediator of cancer progression. Cancer and stromal cells remodel the ECM through a variety of mechanisms, including matrix degradation, cross-linking, deposition, and physical remodeling. Cancer cells sense these mechanical environmental changes through cell-matrix adhesion complexes and subsequently alter their tension between both neighboring cells and the surrounding matrix, thereby altering the force landscape within the microenvironment. This communication not only allows cancer cells to communicate with each other, but allows stromal cells to communicate with cancer cells through matrix remodeling. Here, we review the mechanisms of intercellular force transmission, the subsequent matrix remodeling, and the implications of this mechanical communication on cancer progression.
Collapse
Affiliation(s)
- Samantha C. Schwager
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, Nashville, TN 37235 USA
| | - Paul V. Taufalele
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, Nashville, TN 37235 USA
| | - Cynthia A. Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, Nashville, TN 37235 USA
| |
Collapse
|