1
|
Mancha S, Horan M, Pasachhe O, Keikhosravi A, Eliceiri KW, Matkowskyj KA, Notbohm J, Skala MC, Campagnola PJ. Multiphoton excited polymerized biomimetic models of collagen fiber morphology to study single cell and collective migration dynamics in pancreatic cancer. Acta Biomater 2024; 187:212-226. [PMID: 39182805 PMCID: PMC11446658 DOI: 10.1016/j.actbio.2024.08.026] [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: 04/29/2024] [Revised: 08/02/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
The respective roles of aligned collagen fiber morphology found in the extracellular matrix (ECM) of pancreatic cancer patients and cellular migration dynamics have been gaining attention because of their connection with increased aggressive phenotypes and poor prognosis. To better understand how collagen fiber morphology influences cell-matrix interactions associated with metastasis, we used Second Harmonic Generation (SHG) images from patient biopsies with Pancreatic ductal adenocarcinoma (PDAC) as models to fabricate collagen scaffolds to investigate processes associated with motility. Using the PDAC BxPC-3 metastatic cell line, we investigated single and collective cell dynamics on scaffolds of varying collagen alignment. Collective or clustered cells grown on the scaffolds with the highest collagen fiber alignment had increased E-cadherin expression and larger focal adhesion sites compared to single cells, consistent with metastatic behavior. Analysis of single cell motility revealed that the dynamics were characterized by random walk on all substrates. However, examining collective motility over different time points showed that the migration was super-diffusive and enhanced on highly aligned fibers, whereas it was hindered and sub-diffusive on un-patterned substrates. This was further supported by the more elongated morphology observed in collectively migrating cells on aligned collagen fibers. Overall, this approach allows the decoupling of single and collective cell behavior as a function of collagen alignment and shows the relative importance of collective cell behavior as well as fiber morphology in PDAC metastasis. We suggest these scaffolds can be used for further investigations of PDAC cell biology. STATEMENT OF SIGNIFICANCE: Pancreatic ductal adenocarcinoma (PDAC) has a high mortality rate, where aligned collagen has been associated with poor prognosis. Biomimetic models representing this architecture are needed to understand complex cellular interactions. The SHG image-based models based on stromal collagen from human biopsies afford the measurements of cell morphology, cadherin and focal adhesion expression as well as detailed motility dynamics. Using a metastatic cell line, we decoupled the roles of single cell and collective cell behavior as well as that arising from aligned collagen. Our data suggests that metastatic characteristics are enhanced by increased collagen alignment and that collective cell behavior is more relevant to metastatic processes. These scaffolds provide new insight in this disease and can be a platform for further experiments such as testing drug efficacy.
Collapse
Affiliation(s)
- Sophie Mancha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Meghan Horan
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | - Adib Keikhosravi
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kevin W Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI, USA
| | - Kristina A Matkowskyj
- Department of Pathology & Lab Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jacob Notbohm
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Melissa C Skala
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI, USA.
| | - Paul J Campagnola
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
2
|
Kandasamy A, Yeh YT, Serrano R, Mercola M, Del Álamo JC. Uncertainty-Aware Traction Force Microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.602172. [PMID: 39026786 PMCID: PMC11257441 DOI: 10.1101/2024.07.05.602172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Traction Force Microscopy (TFM) is a versatile tool to quantify cell-exerted forces by imaging and tracking fiduciary markers embedded in elastic substrates. The computations involved in TFM are ill-conditioned, and data smoothing or regularization is required to avoid overfitting the noise in the tracked substrate displacements. Most TFM calculations depend critically on the heuristic selection of regularization (hyper)parameters affecting the balance between overfitting and smoothing. However, TFM methods rarely estimate or account for measurement errors in substrate deformation to adjust the regularization level accordingly. Moreover, there is a lack of tools to quantify how these errors propagate to the recovered traction stresses. These limitations make it difficult to interpret TFM readouts and hinder comparing different experiments. This manuscript presents an uncertainty-aware TFM technique that estimates the variability in the magnitude and direction of the traction stress vector recovered at each point in space and time of each experiment. In this technique, substrate deformation and its uncertainty are quantified using a non-parametric bootstrap PIV method by resampling the microscopy image pixels (PIV-UQ). This information is passed to a hierarchical Bayesian framework that automatically selects its hyperparameters to perform spatially adaptive regularization conditioned on image quality and propagates the uncertainty to the traction stress readouts (TFM-UQ). We validate the performance of PIV-UQ and TFM-UQ using synthetic datasets with prescribed image quality variations and demonstrate the application of PIV-UQ and TFM-UQ to experimental datasets. These studies show that TFM-UQ locally adapts the level of smoothing, outperforming traditional regularization methods. They also illustrate how uncertainty-aware TFM tools can be used to objectively choose key image analysis parameters like PIV-UQ interrogation window size. We anticipate that these tools will allow for decoupling biological heterogeneity from measurement variability and facilitate automating the analysis of large datasets by parameter-free, input data-based regularization.
Collapse
|
3
|
Nejad MR, Ruske LJ, McCord M, Zhang J, Zhang G, Notbohm J, Yeomans JM. Stress-shape misalignment in confluent cell layers. Nat Commun 2024; 15:3628. [PMID: 38684651 PMCID: PMC11059169 DOI: 10.1038/s41467-024-47702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
In tissue formation and repair, the epithelium undergoes complex patterns of motion driven by the active forces produced by each cell. Although the principles governing how the forces evolve in time are not yet clear, it is often assumed that the contractile stresses within the cell layer align with the axis defined by the body of each cell. Here, we simultaneously measured the orientations of the cell shape and the cell-generated contractile stresses, observing correlated, dynamic domains in which the stresses were systematically misaligned with the cell body. We developed a continuum model that decouples the orientations of contractile stress and cell body. The model recovered the spatial and temporal dynamics of the regions of misalignment in the experiments. These findings reveal that the cell controls its contractile forces independently from its shape, suggesting that the physical rules relating cell forces and cell shape are more flexible than previously thought.
Collapse
Affiliation(s)
- Mehrana R Nejad
- The Rudolf Peierls Centre for Theoretical Physics, Department of Physics, University of Oxford, Parks Road, Oxford, OX1 3PU, United Kingdom.
| | - Liam J Ruske
- The Rudolf Peierls Centre for Theoretical Physics, Department of Physics, University of Oxford, Parks Road, Oxford, OX1 3PU, United Kingdom
| | - Molly McCord
- Biophysics Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Jun Zhang
- Biophysics Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Guanming Zhang
- Center for Soft Matter Research, Department of Physics, New York University, New York, NY, 10003, USA
- Simons Center for Computational Physical Chemistry, Department of Chemistry, New York University, New York, NY, 10003, USA
| | - Jacob Notbohm
- Biophysics Program, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA.
| | - Julia M Yeomans
- The Rudolf Peierls Centre for Theoretical Physics, Department of Physics, University of Oxford, Parks Road, Oxford, OX1 3PU, United Kingdom.
| |
Collapse
|
4
|
Micek HM, Yang N, Dutta M, Rosenstock L, Ma Y, Hielsberg C, McCord M, Notbohm J, McGregor S, Kreeger PK. The role of Piezo1 mechanotransduction in high-grade serous ovarian cancer: Insights from an in vitro model of collective detachment. SCIENCE ADVANCES 2024; 10:eadl4463. [PMID: 38669327 PMCID: PMC11051664 DOI: 10.1126/sciadv.adl4463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/22/2024] [Indexed: 04/28/2024]
Abstract
Slowing peritoneal spread in high-grade serous ovarian cancer (HGSOC) would improve patient prognosis and quality of life. HGSOC spreads when single cells and spheroids detach, float through the peritoneal fluid and take over new sites, with spheroids thought to be more aggressive than single cells. Using our in vitro model of spheroid collective detachment, we determine that increased substrate stiffness led to the detachment of more spheroids. We identified a mechanism where Piezo1 activity increased MMP-1/MMP-10, decreased collagen I and fibronectin, and increased spheroid detachment. Piezo1 expression was confirmed in omental masses from patients with stage III/IV HGSOC. Using OV90 and CRISPR-modified PIEZO1-/- OV90 in a mouse xenograft model, we determined that while both genotypes efficiently took over the omentum, loss of Piezo1 significantly decreased ascitic volume, tumor spheroids in the ascites, and the number of macroscopic tumors in the mesentery. These results support that slowing collective detachment may benefit patients and identify Piezo1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Hannah M. Micek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ning Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Mayuri Dutta
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Lauren Rosenstock
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yicheng Ma
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Caitlin Hielsberg
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Molly McCord
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Biophysics Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jacob Notbohm
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Biophysics Program, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Stephanie McGregor
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Pamela K. Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
5
|
Denisin AK, Kim H, Riedel-Kruse IH, Pruitt BL. Field Guide to Traction Force Microscopy. Cell Mol Bioeng 2024; 17:87-106. [PMID: 38737454 PMCID: PMC11082129 DOI: 10.1007/s12195-024-00801-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/26/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Traction force microscopy (TFM) is a widely used technique to measure cell contractility on compliant substrates that mimic the stiffness of human tissues. For every step in a TFM workflow, users make choices which impact the quantitative results, yet many times the rationales and consequences for making these decisions are unclear. We have found few papers which show the complete experimental and mathematical steps of TFM, thus obfuscating the full effects of these decisions on the final output. Methods Therefore, we present this "Field Guide" with the goal to explain the mathematical basis of common TFM methods to practitioners in an accessible way. We specifically focus on how errors propagate in TFM workflows given specific experimental design and analytical choices. Results We cover important assumptions and considerations in TFM substrate manufacturing, substrate mechanical properties, imaging techniques, image processing methods, approaches and parameters used in calculating traction stress, and data-reporting strategies. Conclusions By presenting a conceptual review and analysis of TFM-focused research articles published over the last two decades, we provide researchers in the field with a better understanding of their options to make more informed choices when creating TFM workflows depending on the type of cell being studied. With this review, we aim to empower experimentalists to quantify cell contractility with confidence. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00801-6.
Collapse
Affiliation(s)
| | - Honesty Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305 USA
- Present Address: The Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158 USA
- Department of Molecular and Cellular Biology, and (by courtesy) Departments of Biomedical Engineering, Applied Mathematics, and Physics, University of Arizona, Tucson, AZ 85721 USA
| | - Ingmar H. Riedel-Kruse
- Department of Molecular and Cellular Biology, and (by courtesy) Departments of Biomedical Engineering, Applied Mathematics, and Physics, University of Arizona, Tucson, AZ 85721 USA
| | - Beth L. Pruitt
- Departments of Bioengineering and Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA 93106 USA
| |
Collapse
|
6
|
Karimi A, Aga M, Khan T, D'costa SD, Cardenas-Riumallo S, Zelenitz M, Kelley MJ, Acott TS. Dynamic traction force in trabecular meshwork cells: A 2D culture model for normal and glaucomatous states. Acta Biomater 2024; 175:138-156. [PMID: 38151067 PMCID: PMC10843681 DOI: 10.1016/j.actbio.2023.12.033] [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: 10/10/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
Glaucoma, which is associated with intraocular pressure (IOP) elevation, results in trabecular meshwork (TM) cellular dysfunction, leading to increased rigidity of the extracellular matrix (ECM), larger adhesion forces between the TM cells and ECM, and higher resistance to aqueous humor drainage. TM cells sense the mechanical forces due to IOP dynamic and apply multidimensional forces on the ECM. Recognizing the importance of cellular forces in modulating various cellular activities and development, this study is aimed to develop a 2D in vitro cell culture model to calculate the 3D, depth-dependent, dynamic traction forces, tensile/compressive/shear strain of the normal and glaucomatous human TM cells within a deformable polyacrylamide (PAM) gel substrate. Normal and glaucomatous human TM cells were isolated, cultured, and seeded on top of the PAM gel substrate with embedded FluoSpheres, spanning elastic moduli of 1.5 to 80 kPa. Sixteen-hour post-seeding live confocal microscopy in an incubator was conducted to Z-stack image the 3D displacement map of the FluoSpheres within the PAM gels. Combined with the known PAM gel stiffness, we ascertained the 3D traction forces in the gel. Our results revealed meaningfully larger traction forces in the glaucomatous TM cells compared to the normal TM cells, reaching depths greater than 10-µm in the PAM gel substrate. Stress fibers in TM cells increased with gel rigidity, but diminished when stiffness rose from 20 to 80 kPa. The developed 2D cell culture model aids in understanding how altered mechanical properties in glaucoma impact TM cell behavior and aqueous humor outflow resistance. STATEMENT OF SIGNIFICANCE: Glaucoma, a leading cause of irreversible blindness, is intricately linked to elevated intraocular pressures and their subsequent cellular effects. The trabecular meshwork plays a pivotal role in this mechanism, particularly its interaction with the extracellular matrix. This research unveils an advanced 2D in vitro cell culture model that intricately maps the complex 3D forces exerted by trabecular meshwork cells on the extracellular matrix, offering unparalleled insights into the cellular biomechanics at play in both healthy and glaucomatous eyes. By discerning the changes in these forces across varying substrate stiffness levels, we bridge the gap in understanding between cellular mechanobiology and the onset of glaucoma. The findings stand as a beacon for potential therapeutic avenues, emphasizing the gravity of cellular/extracellular matrix interactions in glaucoma's pathogenesis and setting the stage for targeted interventions in its early stages.
Collapse
Affiliation(s)
- Alireza Karimi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA; Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
| | - Mini Aga
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Taaha Khan
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Siddharth Daniel D'costa
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | | | - Meadow Zelenitz
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Mary J Kelley
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA; Department Integrative Biosciences, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
| | - Ted S Acott
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA; Department Chemical Physiology & Biochemistry, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
7
|
Ouderkirk S, Sedley A, Ong M, Shifflet MR, Harkrider QC, Wright NT, Miller CJ. A Perspective on Developing Modeling and Image Analysis Tools to Investigate Mechanosensing Proteins. Integr Comp Biol 2023; 63:1532-1542. [PMID: 37558388 PMCID: PMC10755202 DOI: 10.1093/icb/icad107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 08/11/2023] Open
Abstract
The shift of funding organizations to prioritize interdisciplinary work points to the need for workflow models that better accommodate interdisciplinary studies. Most scientists are trained in a specific field and are often unaware of the kind of insights that other disciplines could contribute to solving various problems. In this paper, we present a perspective on how we developed an experimental pipeline between a microscopy and image analysis/bioengineering lab. Specifically, we connected microscopy observations about a putative mechanosensing protein, obscurin, to image analysis techniques that quantify cell changes. While the individual methods used are well established (fluorescence microscopy; ImageJ WEKA and mTrack2 programs; MATLAB), there are no existing best practices for how to integrate these techniques into a cohesive, interdisciplinary narrative. Here, we describe a broadly applicable workflow of how microscopists can more easily quantify cell properties (e.g., perimeter, velocity) from microscopy videos of eukaryotic (MDCK) adherent cells. Additionally, we give examples of how these foundational measurements can create more complex, customizable cell mechanics tools and models.
Collapse
Affiliation(s)
- Stephanie Ouderkirk
- Department of Chemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Alex Sedley
- Department of Engineering, James Madison University, Harrisonburg, VA 22807, USA
| | - Mason Ong
- Department of Engineering, James Madison University, Harrisonburg, VA 22807, USA
| | - Mary Ruth Shifflet
- Department of Chemistry, Bridgewater College, Bridgewater, VA 22812, USA
| | - Quinn C Harkrider
- Department of Chemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Nathan T Wright
- Department of Chemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Callie J Miller
- Department of Engineering, James Madison University, Harrisonburg, VA 22807, USA
| |
Collapse
|
8
|
Kechagia Z, Sáez P, Gómez-González M, Canales B, Viswanadha S, Zamarbide M, Andreu I, Koorman T, Beedle AEM, Elosegui-Artola A, Derksen PWB, Trepat X, Arroyo M, Roca-Cusachs P. The laminin-keratin link shields the nucleus from mechanical deformation and signalling. NATURE MATERIALS 2023; 22:1409-1420. [PMID: 37709930 PMCID: PMC10627833 DOI: 10.1038/s41563-023-01657-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/31/2023] [Indexed: 09/16/2023]
Abstract
The mechanical properties of the extracellular matrix dictate tissue behaviour. In epithelial tissues, laminin is a very abundant extracellular matrix component and a key supporting element. Here we show that laminin hinders the mechanoresponses of breast epithelial cells by shielding the nucleus from mechanical deformation. Coating substrates with laminin-111-unlike fibronectin or collagen I-impairs cell response to substrate rigidity and YAP nuclear localization. Blocking the laminin-specific integrin β4 increases nuclear YAP ratios in a rigidity-dependent manner without affecting the cell forces or focal adhesions. By combining mechanical perturbations and mathematical modelling, we show that β4 integrins establish a mechanical linkage between the substrate and keratin cytoskeleton, which stiffens the network and shields the nucleus from actomyosin-mediated mechanical deformation. In turn, this affects the nuclear YAP mechanoresponses, chromatin methylation and cell invasion in three dimensions. Our results demonstrate a mechanism by which tissues can regulate their sensitivity to mechanical signals.
Collapse
Affiliation(s)
- Zanetta Kechagia
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Pablo Sáez
- Laboratori de Càlcul Numèric (LàCaN), Universitat Politècnica de Catalunya, Barcelona, Spain
- Institut de Matemátiques de la UPC-BarcelonaTech (IMTech), Barcelona, Spain
| | - Manuel Gómez-González
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Brenda Canales
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King's College London, London, UK
| | - Srivatsava Viswanadha
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | | | - Ion Andreu
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Thijs Koorman
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Amy E M Beedle
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Physics, King's College London, London, UK
| | - Alberto Elosegui-Artola
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King's College London, London, UK
| | - Patrick W B Derksen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Marino Arroyo
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Laboratori de Càlcul Numèric (LàCaN), Universitat Politècnica de Catalunya, Barcelona, Spain
- Institut de Matemátiques de la UPC-BarcelonaTech (IMTech), Barcelona, Spain
- Centre Internacional de Mètodes Numèrics en Enginyeria (CIMNE), Barcelona, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- University of Barcelona, Barcelona, Spain.
| |
Collapse
|
9
|
Wu MC, Yu HW, Chen YQ, Ou MH, Serrano R, Huang GL, Wang YK, Lin KH, Fan YJ, Wu CC, Del Álamo JC, Chiou A, Chien S, Kuo JC. Early committed polarization of intracellular tension in response to cell shape determines the osteogenic differentiation of mesenchymal stromal cells. Acta Biomater 2023; 163:287-301. [PMID: 36328121 PMCID: PMC11389728 DOI: 10.1016/j.actbio.2022.10.052] [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: 04/15/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2022]
Abstract
Within the heterogeneous tissue architecture, a comprehensive understanding of how cell shapes regulate cytoskeletal mechanics by adjusting focal adhesions (FAs) signals to correlate with the lineage commitment of mesenchymal stromal cells (MSCs) remains obscure. Here, via engineered extracellular matrices, we observed that the development of mature FAs, coupled with a symmetrical pattern of radial fiber bundles, appeared at the right-angle vertices in cells with square shape. While circular cells aligned the transverse fibers parallel to the cell edge, and moved them centripetally in a counter-clockwise direction, symmetrical bundles of radial fibers at the vertices of square cells disrupted the counter-clockwise swirling and bridged the transverse fibers to move centripetally. In square cells, the contractile force, generated by the myosin IIA-enriched transverse fibers, were concentrated and transmitted outwards along the symmetrical bundles of radial fibers, to the extracellular matrix through FAs, and thereby driving FA organization and maturation. The symmetrical radial fiber bundles concentrated the transverse fibers contractility inward to the linkage between the actin cytoskeleton and the nuclear envelope. The tauter cytoskeletal network adjusted the nuclear-actomyosin force balance to cause nuclear deformability and to increase nuclear translocation of the transcription co-activator YAP, which in turn modulated the switch in MSC commitment. Thus, FAs dynamically respond to geometric cues and remodel actin cytoskeletal network to re-distribute intracelluar tension towards the cell nucleus, and thereby controlling YAP mechanotransduction signaling in regulating MSC fate decision. STATEMENT OF SIGNIFICANCE: We decipher how cellular mechanics is self-organized depending on extracellular geometric features to correlate with mesenchymal stromal cell lineage commitment. In response to geometry constrains on cell morphology, symmetrical radial fiber bundles are assembled and clustered depending on the maturation state of focal adhesions and bridge with the transverse fibers, and thereby establishing the dynamic cytoskeletal network. Contractile force, generated by the myosin-IIA-enriched transverse fibers, is transmitted and dynamically drives the retrograde movement of the actin cytoskeletal network, which appropriately adjusts the nuclear-actomyosin force balance and deforms the cell nucleus for YAP mechano-transduction signaling in regulating mesenchymal stromal cell fate decision.
Collapse
Affiliation(s)
- Ming-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Helen Wenshin Yu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Yin-Quan Chen
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Meng-Hsin Ou
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Ricardo Serrano
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA; Department of Mechanical and Aerospace Engineering, University of California at San Diego, La Jolla, CA 92093, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Guan-Lin Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Yang-Kao Wang
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Kung-Hui Lin
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Jui Fan
- School of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Chi-Chang Wu
- Department of Electronic Engineering, National Chin-Yi University of Technology, Taichung 411030, Taiwan
| | - Juan C Del Álamo
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA; Department of Mechanical and Aerospace Engineering, University of California at San Diego, La Jolla, CA 92093, USA; Center for Cardiovascular Biology, University of Washington, School of Medicine, Seattle, WA, 98109, USA; Mechanical Engineering Department, University of Washington, Seattle, WA, 98195, USA
| | - Arthur Chiou
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Shu Chien
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan.
| |
Collapse
|
10
|
Muenkel M, Aparicio-Yuste R, Tal MC, Kraiczy P, Bastounis EE. Spatiotemporal characterization of endothelial cell motility and physical forces during exposure to Borrelia burgdorferi. STAR Protoc 2022; 3:101832. [DOI: 10.1016/j.xpro.2022.101832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
11
|
Alkmin S, Patankar MS, Campagnola PJ. Assessing the roles of collagen fiber morphology and matrix stiffness on ovarian cancer cell migration dynamics using multiphoton fabricated orthogonal image-based models. Acta Biomater 2022; 153:342-354. [PMID: 36152908 PMCID: PMC10324295 DOI: 10.1016/j.actbio.2022.09.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/24/2022] [Accepted: 09/15/2022] [Indexed: 11/01/2022]
Abstract
Ovarian cancer remains the deadliest of the gynecological cancers, where this arises from poor screening and imaging tools that can detect early disease, and also limited understanding of the structural and functional aspects of the tumor microenvironment. To gain insight into the underlying cellular dynamics, we have used multiphoton excited fabrication to create Second Harmonic Generation (SHG) image-based orthogonal models from collagen/GelMA that represent both the collagen matrix morphology and stiffness (∼2-8 kPa) of normal ovarian stroma and high grade serous ovarian cancers (HGSOC). These scaffolds are used to study migration/cytoskeletal dynamics of normal (IOSE) and ovarian cancer (OVCA433) cell lines. We found that the highly aligned fiber morphology of HGSOC promotes aspects of motility (motility coefficient, motility, and focal adhesion expression) through a contact guidance mechanism and that stiffer matrix further promotes these same processes through a mechanosensitive mechanism, where these trends were similar for both normal and cancer cells. However, cell specific differences were found on these orthogonal models relative to those providing only morphology, showing the importance of presenting both morphology and stiffness cues. Moreover, we found increased cadherin expression and decreased cell alignment only for cancer cells on scaffolds of intermediate modulus suggesting different stiffness-dependent mechanotransduction mechanisms are engaged. This overall approach affords decoupling the roles of matrix morphology, stiffness and cell genotype and affords hypothesis testing of the factors giving rise to disease progression and metastasis. Further, more established fabrication techniques cannot simultaneously reproduce both the 3D collagen fiber morphology and stiffness. STATEMENT OF SIGNIFICANCE: Ovarian cancer metastasizes when lesions are small, where cells exfoliate from the surface of the ovary and reattach at distal sites in the peritoneum. The adhesion/migration dynamics are not well understood and there is a need for new 3D in vitro models of the extracellular matrix to study the biology. Here we use multiphoton excited crosslinking to fabricate ECM orthogonal models that represent the collagen morphology and stiffness in human ovarian tissues. These are then used to study ovarian cancer cell migration dynamics and we found that contact guidance and a mechanosensitive response and cell genotype all combine to affect the behavior. These models provide insight into disease etiology and progression not readily possible by other fabrication methods.
Collapse
Affiliation(s)
- Samuel Alkmin
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1550 Engineering Drive, Madison, WI 53706, USA
| | - Manish S Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Paul J Campagnola
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1550 Engineering Drive, Madison, WI 53706, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
12
|
Sivagurunathan S, Vahabikashi A, Yang H, Zhang J, Vazquez K, Rajasundaram D, Politanska Y, Abdala-Valencia H, Notbohm J, Guo M, Adam SA, Goldman RD. Expression of vimentin alters cell mechanics, cell-cell adhesion, and gene expression profiles suggesting the induction of a hybrid EMT in human mammary epithelial cells. Front Cell Dev Biol 2022; 10:929495. [PMID: 36200046 PMCID: PMC9527304 DOI: 10.3389/fcell.2022.929495] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Vimentin is a Type III intermediate filament (VIF) cytoskeletal protein that regulates the mechanical and migratory behavior of cells. Its expression is considered to be a marker for the epithelial to mesenchymal transition (EMT) that takes place in tumor metastasis. However, the molecular mechanisms regulated by the expression of vimentin in the EMT remain largely unexplored. We created MCF7 epithelial cell lines expressing vimentin from a cumate-inducible promoter to address this question. When vimentin expression was induced in these cells, extensive cytoplasmic VIF networks were assembled accompanied by changes in the organization of the endogenous keratin intermediate filament networks and disruption of desmosomes. Significant reductions in intercellular forces by the cells expressing VIFs were measured by quantitative monolayer traction force and stress microscopy. In contrast, laser trapping micro-rheology revealed that the cytoplasm of MCF7 cells expressing VIFs was stiffer than the uninduced cells. Vimentin expression activated transcription of genes involved in pathways responsible for cell migration and locomotion. Importantly, the EMT related transcription factor TWIST1 was upregulated only in wild type vimentin expressing cells and not in cells expressing a mutant non-polymerized form of vimentin, which only formed unit length filaments (ULF). Taken together, our results suggest that vimentin expression induces a hybrid EMT correlated with the upregulation of genes involved in cell migration.
Collapse
Affiliation(s)
- Suganya Sivagurunathan
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Amir Vahabikashi
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Haiqian Yang
- Department of Mechanical Engineering , Massachusetts Institute of Technology , Cambridge , MA, United States
| | - Jun Zhang
- Biophysics Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, United States
| | - Kelly Vazquez
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yuliya Politanska
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hiam Abdala-Valencia
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jacob Notbohm
- Biophysics Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Ming Guo
- Department of Mechanical Engineering , Massachusetts Institute of Technology , Cambridge , MA, United States
| | - Stephen A Adam
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Robert D Goldman
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
13
|
Yuste RA, Muenkel M, Axarlis K, Gómez Benito MJ, Reuss A, Blacker G, Tal MC, Kraiczy P, Bastounis EE. Borrelia burgdorferi modulates the physical forces and immunity signaling in endothelial cells. iScience 2022; 25:104793. [PMID: 35992087 PMCID: PMC9389243 DOI: 10.1016/j.isci.2022.104793] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/09/2022] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
Borrelia burgdorferi (Bb), a vector-borne bacterial pathogen and the causative agent of Lyme disease, can spread to distant tissues in the human host by traveling in and through monolayers of endothelial cells (ECs) lining the vasculature. To examine whether Bb alters the physical forces of ECs to promote its dissemination, we exposed ECs to Bb and observed a sharp and transient increase in EC traction and intercellular forces, followed by a prolonged decrease in EC motility and physical forces. All variables returned to baseline at 24 h after exposure. RNA sequencing analysis revealed an upregulation of innate immune signaling pathways during early but not late Bb exposure. Exposure of ECs to heat-inactivated Bb recapitulated only the early weakening of EC mechanotransduction. The differential responses to live versus heat-inactivated Bb indicate a tight interplay between innate immune signaling and physical forces in host ECs and suggest their active modulation by Bb. Early exposure to Borrelia decreases endothelial cell motility and physical forces Early exposure to Borrelia also upregulates the host’s innate immune signaling pathways Host cell mechanics and signaling return to steady state at late exposure times Exposure to dead bacteria steadily reduces motility and physical forces of host cells
Collapse
|
14
|
Human endothelial cells display a rapid tensional stress increase in response to tumor necrosis factor-α. PLoS One 2022; 17:e0270197. [PMID: 35749538 PMCID: PMC9232152 DOI: 10.1371/journal.pone.0270197] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 06/06/2022] [Indexed: 02/07/2023] Open
Abstract
Endothelial cells form the inner layer of blood vessels, making them the first barrier between the blood and interstitial tissues; thus endothelial cells play a crucial role in inflammation. In the inflammatory response, one important element is the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α). While other pro-inflammatory agents like thrombin and histamine induce acute but transient changes in endothelial cells, which have been well studied biologically as well as mechanically, TNF-α is primarily known for its sustained effects on permeability and leukocyte recruitment. These functions are associated with transcriptional changes that take place on the timescale of hours and days. Here, we investigated the early mechanical action of TNF-α and show that even just 4 min after TNF-α was added onto human umbilical vein endothelial cell monolayers, there was a striking rise in mechanical substrate traction force and internal monolayer tension. These traction forces act primarily at the boundary of the monolayer, as was to be expected. This increased internal monolayer tension may, in addition to TNF-α’s other well-studied biochemical responses, provide a mechanical signal for the cells to prepare to recruit leukocytes.
Collapse
|
15
|
Schürmann H, Abbasi F, Russo A, Hofemeier AD, Brandt M, Roth J, Vogl T, Betz T. Analysis of monocyte cell tractions in 2.5D reveals mesoscale mechanics of podosomes during substrate-indenting cell protrusion. J Cell Sci 2022; 135:275542. [PMID: 35621127 PMCID: PMC9189428 DOI: 10.1242/jcs.259042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 04/27/2022] [Indexed: 12/23/2022] Open
Abstract
Podosomes are mechanosensitive protrusive actin structures that are prominent in myeloid cells, and they have been linked to vascular extravasation. Recent studies have suggested that podosomes are hierarchically organized and have coordinated dynamics on the cell scale, which implies that the local force generation by single podosomes can be different from their global combined action. Complementary to previous studies focusing on individual podosomes, here we investigated the cell-wide force generation of podosome-bearing ER-Hoxb8 monocytes. We found that the occurrence of focal tractions accompanied by a cell-wide substrate indentation cannot be explained by summing the forces of single podosomes. Instead, our findings suggest that superimposed contraction on the cell scale gives rise to a buckling mechanism that can explain the measured cell-scale indentation. Specifically, the actomyosin network contraction causes peripheral in-plane substrate tractions, while the accumulated internal stress results in out-of-plane deformation in the central cell region via a buckling instability, producing the cell-scale indentation. Hence, we propose that contraction of the actomyosin network, which connects the podosomes, leads to a substrate indentation that acts in addition to the protrusion forces of individual podosomes. This article has an associated First Person interview with the first author of the paper. Summary: Using a buckling model, we extend the current description of local podosome protrusion and include a mechanical explanation for protrusion on the cell scale.
Collapse
Affiliation(s)
- Hendrik Schürmann
- Institute of Cell Biology, ZMBE, University of Münster, Von-Esmarch-Straße 56, D-48149 Münster, Germany
| | - Fatemeh Abbasi
- Institute of Cell Biology, ZMBE, University of Münster, Von-Esmarch-Straße 56, D-48149 Münster, Germany.,Third Physical Institute, University of Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| | - Antonella Russo
- Institute of Immunology, University of Münster, Röntgenstraße 21, D-48149 Münster, Germany
| | - Arne D Hofemeier
- Institute of Cell Biology, ZMBE, University of Münster, Von-Esmarch-Straße 56, D-48149 Münster, Germany.,Third Physical Institute, University of Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| | - Matthias Brandt
- Institute of Cell Biology, ZMBE, University of Münster, Von-Esmarch-Straße 56, D-48149 Münster, Germany.,Third Physical Institute, University of Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Röntgenstraße 21, D-48149 Münster, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Münster, Röntgenstraße 21, D-48149 Münster, Germany
| | - Timo Betz
- Institute of Cell Biology, ZMBE, University of Münster, Von-Esmarch-Straße 56, D-48149 Münster, Germany.,Third Physical Institute, University of Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| |
Collapse
|
16
|
Zancla A, Mozetic P, Orsini M, Forte G, Rainer A. A primer to traction force microscopy. J Biol Chem 2022; 298:101867. [PMID: 35351517 PMCID: PMC9092999 DOI: 10.1016/j.jbc.2022.101867] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 12/24/2022] Open
Abstract
Traction force microscopy (TFM) has emerged as a versatile technique for the measurement of single-cell-generated forces. TFM has gained wide use among mechanobiology laboratories, and several variants of the original methodology have been proposed. However, issues related to the experimental setup and, most importantly, data analysis of cell traction datasets may restrain the adoption of TFM by a wider community. In this review, we summarize the state of the art in TFM-related research, with a focus on the analytical methods underlying data analysis. We aim to provide the reader with a friendly compendium underlying the potential of TFM and emphasizing the methodological framework required for a thorough understanding of experimental data. We also compile a list of data analytics tools freely available to the scientific community for the furtherance of knowledge on this powerful technique.
Collapse
Affiliation(s)
- Andrea Zancla
- Department of Engineering, Università degli Studi Roma Tre, Rome, Italy; Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Pamela Mozetic
- Institute of Nanotechnology (NANOTEC), National Research Council, Lecce, Italy; Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Monica Orsini
- Department of Engineering, Università degli Studi Roma Tre, Rome, Italy
| | - Giancarlo Forte
- Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St Anne's University Hospital, Brno, Czechia.
| | - Alberto Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy; Institute of Nanotechnology (NANOTEC), National Research Council, Lecce, Italy.
| |
Collapse
|
17
|
Yeh YT, Skinner DE, Criado-Hidalgo E, Chen NS, Garcia-De Herreros A, El-Sakkary N, Liu L, Zhang S, Kandasamy A, Chien S, Lasheras JC, del Álamo JC, Caffrey CR. Biomechanical interactions of Schistosoma mansoni eggs with vascular endothelial cells facilitate egg extravasation. PLoS Pathog 2022; 18:e1010309. [PMID: 35316298 PMCID: PMC8939816 DOI: 10.1371/journal.ppat.1010309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/26/2022] [Indexed: 12/03/2022] Open
Abstract
The eggs of the parasitic blood fluke, Schistosoma, are the main drivers of the chronic pathologies associated with schistosomiasis, a disease of poverty afflicting approximately 220 million people worldwide. Eggs laid by Schistosoma mansoni in the bloodstream of the host are encapsulated by vascular endothelial cells (VECs), the first step in the migration of the egg from the blood stream into the lumen of the gut and eventual exit from the body. The biomechanics associated with encapsulation and extravasation of the egg are poorly understood. We demonstrate that S. mansoni eggs induce VECs to form two types of membrane extensions during encapsulation; filopodia that probe eggshell surfaces and intercellular nanotubes that presumably facilitate VEC communication. Encapsulation efficiency, the number of filopodia and intercellular nanotubes, and the length of these structures depend on the egg’s vitality and, to a lesser degree, its maturation state. During encapsulation, live eggs induce VEC contractility and membranous structures formation in a Rho/ROCK pathway-dependent manner. Using elastic hydrogels embedded with fluorescent microbeads as substrates to culture VECs, live eggs induce VECs to exert significantly greater contractile forces during encapsulation than dead eggs, which leads to 3D deformations on both the VEC monolayer and the flexible substrate underneath. These significant mechanical deformations cause the VEC monolayer tension to fluctuate with the eventual rupture of VEC junctions, thus facilitating egg transit out of the blood vessel. Overall, our data on the mechanical interplay between host VECs and the schistosome egg improve our understanding of how this parasite manipulates its immediate environment to maintain disease transmission. Schistosomiasis, which infects over 200 million people, is a painful disease of poverty that is caused by inflammatory responses to the Schistosoma blood fluke’s eggs. To continue the parasite’s life cycle, eggs must escape the blood vessels and migrate through tissues of the host to the alimentary canal for exit into the environment. The biomechanical processes that help the immobile eggs to cross the blood vessel’s vascular endothelial cells (VECs) as the first step in this migration are not understood. We found that live but not dead eggs induce VECs to crawl over and encapsulate them. VECs in contact with live eggs make membranous extensions (filopodia) to explore the egg’s surface and also form long intercellular nanotubes to communicate with neighboring cells. VECs stimulate particular (Rho/ROCK) biochemical pathways to increase cell contractility and the forces generated are large enough to eventually break the junctions between cells and allow passage of the eggs into the underlying tissue. Our findings show how schistosome eggs activate and interact with VECs to initiate their escape from the bloodstream.
Collapse
Affiliation(s)
- Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, United States of America
- * E-mail: (YTY); (JCdA); (CRC)
| | - Danielle E. Skinner
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Ernesto Criado-Hidalgo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
| | - Natalie Shee Chen
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Antoni Garcia-De Herreros
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
| | - Nelly El-Sakkary
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Lawrence Liu
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Shun Zhang
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
| | - Adithan Kandasamy
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington, Seattle Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle Washington, United States of America
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Juan C. Lasheras
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
- Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California, United States of America
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, United States of America
- Center for Cardiovascular Biology, University of Washington, Seattle Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle Washington, United States of America
- * E-mail: (YTY); (JCdA); (CRC)
| | - Conor R. Caffrey
- Center for Discovery and Innovation in Parasitic Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, United States of America
- * E-mail: (YTY); (JCdA); (CRC)
| |
Collapse
|
18
|
Li Y, Bai P, Cao H, Li L, Li X, Hou X, Fang J, Li J, Meng Y, Ma L, Tian Y. Imaging dynamic three-dimensional traction stresses. SCIENCE ADVANCES 2022; 8:eabm0984. [PMID: 35294236 PMCID: PMC8926338 DOI: 10.1126/sciadv.abm0984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/24/2022] [Indexed: 06/14/2023]
Abstract
Traction stress between contact objects is ubiquitous and crucial for various physical, biological, and engineering processes such as momentum transfer, tactile perception, and mechanical reliability. Newly developed techniques including electronic skin or traction force microscopy enable traction stress measurement. However, measuring the three-dimensional distribution during a dynamic process remains challenging. Here, we demonstrated a method based on stereo vision to measure three-dimensional traction stress with high spatial and temporal resolution. It showed the ability to image the two-stage adhesion failure of bionic microarrays and display the contribution of elastic resistance and adhesive traction to rolling friction at different contact regions. It also revealed the distributed sucking and sealing effect of the concavity pedal waves that propelled a snail crawling in the horizontal, vertical, and upside-down directions. We expected that the method would advance the understanding of various interfacial phenomena and greatly benefit related applications across physics, biology, and robotics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Liran Ma
- Corresponding author. (Y.T.); (L.M.)
| | - Yu Tian
- Corresponding author. (Y.T.); (L.M.)
| |
Collapse
|
19
|
Kummer D, Steinbacher T, Thölmann S, Schwietzer MF, Hartmann C, Horenkamp S, Demuth S, Peddibhotla SS, Brinkmann F, Kemper B, Schnekenburger J, Brandt M, Betz T, Liashkovich I, Kouzel IU, Shahin V, Corvaia N, Rottner K, Tarbashevich K, Raz E, Greune L, Schmidt MA, Gerke V, Ebnet K. A JAM-A-tetraspanin-αvβ5 integrin complex regulates contact inhibition of locomotion. J Biophys Biochem Cytol 2022; 221:213070. [PMID: 35293964 PMCID: PMC8931538 DOI: 10.1083/jcb.202105147] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 12/16/2021] [Accepted: 01/21/2022] [Indexed: 12/30/2022] Open
Abstract
Contact inhibition of locomotion (CIL) is a process that regulates cell motility upon collision with other cells. Improper regulation of CIL has been implicated in cancer cell dissemination. Here, we identify the cell adhesion molecule JAM-A as a central regulator of CIL in tumor cells. JAM-A is part of a multimolecular signaling complex in which tetraspanins CD9 and CD81 link JAM-A to αvβ5 integrin. JAM-A binds Csk and inhibits the activity of αvβ5 integrin-associated Src. Loss of JAM-A results in increased activities of downstream effectors of Src, including Erk1/2, Abi1, and paxillin, as well as increased activity of Rac1 at cell-cell contact sites. As a consequence, JAM-A-depleted cells show increased motility, have a higher cell-matrix turnover, and fail to halt migration when colliding with other cells. We also find that proper regulation of CIL depends on αvβ5 integrin engagement. Our findings identify a molecular mechanism that regulates CIL in tumor cells and have implications on tumor cell dissemination.
Collapse
Affiliation(s)
- Daniel Kummer
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany,Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| | - Tim Steinbacher
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Sonja Thölmann
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Mariel Flavia Schwietzer
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Christian Hartmann
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Simone Horenkamp
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Sabrina Demuth
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Swetha S.D. Peddibhotla
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Frauke Brinkmann
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Björn Kemper
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Jürgen Schnekenburger
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Matthias Brandt
- Institute-associated Research Group “Mechanics of Cellular Systems”, Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| | - Timo Betz
- Institute-associated Research Group “Mechanics of Cellular Systems”, Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| | - Ivan Liashkovich
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Ivan U. Kouzel
- Sars International Centre for Marine Molecular Biology University of Bergen Thormøhlensgt, Bergen, Norway
| | - Victor Shahin
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Nathalie Corvaia
- Centre d’Immunologie Pierre Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Klemens Rottner
- Divison of Molecular Cell Biology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany,Molecular Cell Biology Group, Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Erez Raz
- Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany,Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, 48419 Münster, Germany
| | - Lilo Greune
- Institute of Infectiology, ZMBE, University of Münster, Münster, Germany
| | | | - Volker Gerke
- Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany,Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, 48419 Münster, Germany
| | - Klaus Ebnet
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany,Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany,Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, 48419 Münster, Germany
| |
Collapse
|
20
|
Vazquez K, Saraswathibhatla A, Notbohm J. Effect of substrate stiffness on friction in collective cell migration. Sci Rep 2022; 12:2474. [PMID: 35169196 PMCID: PMC8847350 DOI: 10.1038/s41598-022-06504-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/25/2022] [Indexed: 11/09/2022] Open
Abstract
In collective cell migration, the motion results from forces produced by each cell and transmitted to the neighboring cells and to the substrate. Because inertia is negligible and the migration occurs over long time scales, the cell layer exhibits viscous behavior, where force and motion are connected by an apparent friction that results from the breaking and forming of adhesive bonds at the cell–cell and cell–substrate interfaces. Most theoretical models for collective migration include an apparent friction to connect force and motion, with many models making predictions that depend on the ratio of cell–cell and cell–substrate friction. However, little is known about factors that affect friction, leaving predictions of many theoretical models untested. Here, we considered how substrate stiffness and the number of adhesions affected friction at the cell–substrate interface. The experimental data were interpreted through prior theoretical models, which led to the same conclusion, that increased substrate stiffness increased the number of cell–substrate adhesions and caused increased cell–substrate friction. In turn, the friction affected the collective migration by altering the curvature at the edge of the cell layer. By revealing underlying factors affecting friction and demonstrating how friction perturbs the collective migration, this work provides experimental evidence supporting prior theoretical models and motivates the study of other ways to alter the collective migration by changing friction.
Collapse
Affiliation(s)
- Kelly Vazquez
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA.,Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Jacob Notbohm
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA. .,Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
21
|
Saraswathibhatla A, Zhang J, Notbohm J. Coordination of contractile tension and cell area changes in an epithelial cell monolayer. Phys Rev E 2022; 105:024404. [PMID: 35291100 DOI: 10.1103/physreve.105.024404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/01/2022] [Indexed: 06/14/2023]
Abstract
During tissue development and repair, cells contract and expand in coordination with their neighbors, giving rise to tissue deformations that occur on length scales far larger than that of a single cell. The biophysical mechanisms by which the contractile forces of each cell cause deformations on multicellular length scales are not fully clear. To investigate this question, we began with the principle of force equilibrium, which dictates a balance of tensile forces between neighboring cells. Based on this principle, we hypothesized that coordinated changes in cell area result from tension transmitted across the cell layer. To test this hypothesis, spatial correlations of both contractile tension and the divergence of cell velocities were measured as readouts of coordinated contractility and collective area changes, respectively. Experiments were designed to alter the spatial correlation of contractile tension using three different methods, including disrupting cell-cell adhesions, modulating the alignment of actomyosin stress fibers between neighboring cells, and changing the size of the cell monolayer. In all experiments, the spatial correlations of both tension and divergence increased or decreased together, in agreement with our hypothesis. To relate our findings to the intracellular mechanism connecting changes in cell area to contractile tension, we disrupted activation of extracellular signal-regulated kinase (ERK), which is known to mediate the intracellular relationship between cell area and contraction. Consistent with prior knowledge, a temporal cross-correlation between cell area and tension revealed that ERK was responsible for a proportional relationship between cell area and contraction. Inhibition of ERK activation reduced the spatial correlations of the divergence of cell velocity but not of tension. Together, our findings suggest that coordination of cell contraction and expansion requires transfer of cell tension over space and ERK-mediated coordination between cell area and contraction in time.
Collapse
Affiliation(s)
| | - Jun Zhang
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Biophysics Program, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Jacob Notbohm
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Biophysics Program, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
22
|
Ghabache E, Cao Y, Miao Y, Groisman A, Devreotes PN, Rappel W. Coupling traction force patterns and actomyosin wave dynamics reveals mechanics of cell motion. Mol Syst Biol 2021; 17:e10505. [PMID: 34898015 PMCID: PMC8666840 DOI: 10.15252/msb.202110505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Motile cells can use and switch between different modes of migration. Here, we use traction force microscopy and fluorescent labeling of actin and myosin to quantify and correlate traction force patterns and cytoskeletal distributions in Dictyostelium discoideum cells that move and switch between keratocyte-like fan-shaped, oscillatory, and amoeboid modes. We find that the wave dynamics of the cytoskeletal components critically determine the traction force pattern, cell morphology, and migration mode. Furthermore, we find that fan-shaped cells can exhibit two different propulsion mechanisms, each with a distinct traction force pattern. Finally, the traction force patterns can be recapitulated using a computational model, which uses the experimentally determined spatiotemporal distributions of actin and myosin forces and a viscous cytoskeletal network. Our results suggest that cell motion can be generated by friction between the flow of this network and the substrate.
Collapse
Affiliation(s)
| | - Yuansheng Cao
- Department of PhysicsUniversity of California, San DiegoLa JollaCAUSA
| | - Yuchuan Miao
- Department of Cell BiologySchool of MedicineJohns Hopkins UniversityBaltimoreMDUSA
| | - Alex Groisman
- Department of PhysicsUniversity of California, San DiegoLa JollaCAUSA
| | - Peter N Devreotes
- Department of Cell BiologySchool of MedicineJohns Hopkins UniversityBaltimoreMDUSA
| | - Wouter‐Jan Rappel
- Department of PhysicsUniversity of California, San DiegoLa JollaCAUSA
| |
Collapse
|
23
|
Sanz-Herrera JA, Barrasa-Fano J, Cóndor M, Van Oosterwyck H. Inverse method based on 3D nonlinear physically constrained minimisation in the framework of traction force microscopy. SOFT MATTER 2021; 17:10210-10222. [PMID: 33165455 DOI: 10.1039/d0sm00789g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Traction force microscopy is a methodology that enables to estimate cellular forces from the measurement of the displacement field of an extracellular matrix (ECM)-mimicking hydrogel that a cell is mechanically interacting with. In this paper, a new inverse and physically-consistent methodology is developed and implemented in the context of 3D nonlinear elasticity. The proposed method searches for a displacement field that approximates the measured one, through the imposition of fulfillment of equilibrium with real and known forces acting in the hydrogel. The overall mathematical formulation leads to a constrained optimisation problem that is treated through a Lagrange operator and that is solved numerically by means of a nonlinear finite element framework. In order to illustrate the potential and enhanced accuracy of the proposed inverse method, it is applied to a total of 5 different real cases of cells cultured in a 3D hydrogel that is considered to behave as a nonlinear elastic material. Different error indicators are defined in order to compare ground truth simulated displacements and tractions to the ones recovered by the new inverse as well as by the forward method. Results indicate that the evaluation of displacement gradients leads to errors, in terms of recovered tractions, that are more than three times lower (on average) for the inverse method compared to the forward method. They highlight the enhanced accuracy of the developed methodology and the importance of appropriate inverse methods that impose physical constraints to traction and stress recovery in the context of traction force microscopy.
Collapse
Affiliation(s)
- J A Sanz-Herrera
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla, Seville, Spain
| | - J Barrasa-Fano
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.
| | - M Cóndor
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.
| | - H Van Oosterwyck
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.
- Prometheus division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Lekka M, Gnanachandran K, Kubiak A, Zieliński T, Zemła J. Traction force microscopy - Measuring the forces exerted by cells. Micron 2021; 150:103138. [PMID: 34416532 DOI: 10.1016/j.micron.2021.103138] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/15/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
Cells generate mechanical forces (traction forces, TFs) while interacting with the extracellular matrix or neighbouring cells. Forces are generated by both cells and extracellular matrix (ECM) and transmitted within the cell-ECM or cell-cell contacts involving focal adhesions or adherens junctions. Within more than two decades, substantial progress has been achieved in techniques that measure TFs. One of the techniques is traction force microscopy (TFM). This review discusses the TFM and its advances in measuring TFs exerted by cells (single cells and multicellular systems) at cell-ECM and cell-cell junctional intracellular interfaces. The answers to how cells sense, adapt and respond to mechanical forces unravel their role in controlling and regulating cell behaviour in normal and pathological conditions.
Collapse
Affiliation(s)
- Małgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland.
| | | | - Andrzej Kubiak
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland
| | - Tomasz Zieliński
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland
| | - Joanna Zemła
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland
| |
Collapse
|
25
|
Bastounis EE, Radhakrishnan P, Prinz CK, Theriot JA. Volume measurement and biophysical characterization of mounds in epithelial monolayers after intracellular bacterial infection. STAR Protoc 2021; 2:100551. [PMID: 34095865 PMCID: PMC8165451 DOI: 10.1016/j.xpro.2021.100551] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Mechanical forces are important in (patho)physiological processes, including how host epithelial cells interact with intracellular bacterial pathogens. As these pathogens disseminate within host epithelial monolayers, large mounds of infected cells are formed due to the forceful action of surrounding uninfected cells, limiting bacterial spread across the basal cell monolayer. Here, we present a protocol for mound volume measurement and biophysical characterization of mound formation. Modifications to this protocol may be necessary for studying different host cell types or pathogenic organisms. For complete details on the use and execution of this protocol, please refer to Bastounis et al. (2021). Protocol allows for formation of mounds of extruded infected cells in cell monolayers Confocal microscopy and image processing to calculate volume of extruded domains Laser wounding protocol for tension estimation built around mounds TFM incorporated to measure traction stresses of infected mounders and surrounders
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.,Interfaculty Institute of Microbiology and Infection Medicine, Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI, EXC 2124), University of Tübingen, Tübingen 72074, Germany
| | - Prathima Radhakrishnan
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.,Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Christopher K Prinz
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
26
|
Feyen DAM, McKeithan WL, Bruyneel AAN, Spiering S, Hörmann L, Ulmer B, Zhang H, Briganti F, Schweizer M, Hegyi B, Liao Z, Pölönen RP, Ginsburg KS, Lam CK, Serrano R, Wahlquist C, Kreymerman A, Vu M, Amatya PL, Behrens CS, Ranjbarvaziri S, Maas RGC, Greenhaw M, Bernstein D, Wu JC, Bers DM, Eschenhagen T, Metallo CM, Mercola M. Metabolic Maturation Media Improve Physiological Function of Human iPSC-Derived Cardiomyocytes. Cell Rep 2021; 32:107925. [PMID: 32697997 PMCID: PMC7437654 DOI: 10.1016/j.celrep.2020.107925] [Citation(s) in RCA: 202] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/15/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have enormous potential for the study of human cardiac disorders. However, their physiological immaturity severely limits their utility as a model system and their adoption for drug discovery. Here, we describe maturation media designed to provide oxidative substrates adapted to the metabolic needs of human iPSC (hiPSC)-CMs. Compared with conventionally cultured hiPSC-CMs, metabolically matured hiPSC-CMs contract with greater force and show an increased reliance on cardiac sodium (Na+) channels and sarcoplasmic reticulum calcium (Ca2+) cycling. The media enhance the function, long-term survival, and sarcomere structures in engineered heart tissues. Use of the maturation media made it possible to reliably model two genetic cardiac diseases: long QT syndrome type 3 due to a mutation in the cardiac Na+ channel SCN5A and dilated cardiomyopathy due to a mutation in the RNA splicing factor RBM20. The maturation media should increase the fidelity of hiPSC-CMs as disease models. Physiological immaturity of iPSC-derived cardiomyocytes limits their fidelity as disease models. Feyen et al. developed a low glucose, high oxidative substrate media that increase maturation of ventricular-like hiPSC-CMs in 2D and 3D cultures relative to standard protocols. Improved characteristics include a low resting Vm, rapid depolarization, and increased Ca2+ dependence and force generation.
Collapse
Affiliation(s)
- Dries A M Feyen
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wesley L McKeithan
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Arne A N Bruyneel
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sean Spiering
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Larissa Hörmann
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bärbel Ulmer
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hui Zhang
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Francesca Briganti
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michaela Schweizer
- Electron Microscopy Unit, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bence Hegyi
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Zhandi Liao
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | | | - Kenneth S Ginsburg
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Chi Keung Lam
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ricardo Serrano
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Christine Wahlquist
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Alexander Kreymerman
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michelle Vu
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Prashila L Amatya
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Charlotta S Behrens
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sara Ranjbarvaziri
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Renee G C Maas
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Matthew Greenhaw
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Daniel Bernstein
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Joseph C Wu
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Mark Mercola
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
27
|
Liu MY, Jiao Y, Liu J, Zhang S, Li W. Hemodynamic Parameters Predict In-stent Thrombosis After Multibranched Endovascular Repair of Complex Abdominal Aortic Aneurysms: A Retrospective Study of Branched Stent-Graft Thrombosis. Front Cardiovasc Med 2021; 8:654412. [PMID: 33969018 PMCID: PMC8102902 DOI: 10.3389/fcvm.2021.654412] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/22/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Branch vessel occlusion is reported in endovascular repair of aortic pathology. This study aimed to evaluate the hemodynamic indicators associated with in-stent thrombosis (IST) of a branched stent-graft (BSG) after endovascular aortic repair (EVAR) of a complex abdominal aortic aneurysm. Methods: A retrospective evaluation was performed based on the computed tomography (CT) scans and clinical data of three patients who underwent multi-branched endovascular repair. Patient-specific 3-dimensional models were reconstructed, and hemodynamic analysis was performed for IST. Hemodynamics-related parameters including time-averaged wall shear stress (TAWSS), oscillatory shear stress index (OSI), and relative residence time (RRT) were compared among the individual patients. Results: The flow velocity, TAWSS, OSI, and RRT were radically changed in the area of the IST. In BSGs, IST tended to occur in the regions of hemodynamic alteration near the bends in the device, where a decreased flow velocity (<0.6 m/s) and TAWSS (<0.8 Pa) and an elevated OSI (>0.2) and RRT (>5 s) were consistently observed. Conclusions: Hemodynamic perturbations in BSGs cause a predisposition to IST, which can be predicted by a series of changes in the flow parameters. Early hemodynamic analysis might be useful for identifying and remediating IST after multibranched endovascular repair.
Collapse
Affiliation(s)
- Ming-Yuan Liu
- Department of Vascular Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Center for Vascular Surgery, Beijing, China
| | - Yang Jiao
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China.,The Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Junjun Liu
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Simeng Zhang
- Department of Vascular Surgery, Changhai Hospital, Shanghai, China.,Department of Pediatric Cardiac Surgery, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, China.,The Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
28
|
Bastounis EE, Serrano-Alcalde F, Radhakrishnan P, Engström P, Gómez-Benito MJ, Oswald MS, Yeh YT, Smith JG, Welch MD, García-Aznar JM, Theriot JA. Mechanical competition triggered by innate immune signaling drives the collective extrusion of bacterially infected epithelial cells. Dev Cell 2021; 56:443-460.e11. [PMID: 33621492 PMCID: PMC7982222 DOI: 10.1016/j.devcel.2021.01.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/02/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
Intracellular pathogens alter their host cells' mechanics to promote dissemination through tissues. Conversely, host cells may respond to the presence of pathogens by altering their mechanics to limit infection. Here, we monitored epithelial cell monolayers infected with intracellular bacterial pathogens, Listeria monocytogenes or Rickettsia parkeri, over days. Under conditions in which these pathogens trigger innate immune signaling through NF-κB and use actin-based motility to spread non-lytically intercellularly, we found that infected cell domains formed three-dimensional mounds. These mounds resulted from uninfected cells moving toward the infection site, collectively squeezing the softer and less contractile infected cells upward and ejecting them from the monolayer. Bacteria in mounds were less able to spread laterally in the monolayer, limiting the growth of the infection focus, while extruded infected cells underwent cell death. Thus, the coordinated forceful action of uninfected cells actively eliminates large domains of infected cells, consistent with this collective cell response representing an innate immunity-driven process.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | | | - Prathima Radhakrishnan
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Patrik Engström
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - María J Gómez-Benito
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza 50009, Spain
| | - Mackenzi S Oswald
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jason G Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Matthew D Welch
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - José M García-Aznar
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza 50009, Spain
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
29
|
Bodor DL, Pönisch W, Endres RG, Paluch EK. Of Cell Shapes and Motion: The Physical Basis of Animal Cell Migration. Dev Cell 2020; 52:550-562. [PMID: 32155438 DOI: 10.1016/j.devcel.2020.02.013] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/10/2020] [Accepted: 02/14/2020] [Indexed: 01/31/2023]
Abstract
Motile cells have developed a variety of migration modes relying on diverse traction-force-generation mechanisms. Before the behavior of intracellular components could be easily imaged, cell movements were mostly classified by different types of cellular shape dynamics. Indeed, even though some types of cells move without any significant change in shape, most cell propulsion mechanisms rely on global or local deformations of the cell surface. In this review, focusing mostly on metazoan cells, we discuss how different types of local and global shape changes underlie distinct migration modes. We then discuss mechanical differences between force-generation mechanisms and finish by speculating on how they may have evolved.
Collapse
Affiliation(s)
- Dani L Bodor
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; Oncode Institute, Hubrecht Institute-KNAW, Utrecht, the Netherlands
| | - Wolfram Pönisch
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Robert G Endres
- Department of Life Sciences and Centre for Integrative Systems Biology and Bioinformatics, Imperial College, London SW7 2AZ, UK
| | - Ewa K Paluch
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK.
| |
Collapse
|
30
|
Karmakar R, Schich C, Kamprad N, Scheller V, Gutierrez E, Groisman A, Rappel WJ, Tarantola M. Novel micropatterning technique reveals dependence of cell-substrate adhesion and migration of social amoebas on parental strain, development, and fluorescent markers. PLoS One 2020; 15:e0236171. [PMID: 32702047 PMCID: PMC7377449 DOI: 10.1371/journal.pone.0236171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Cell-substrate adhesion of the social amoeba Dictyostelium discoideum, a model organism often used for the study of chemotaxis, is non-specific and does not involve focal adhesion complexes. Therefore, micropatterned substrates where adherent Dictyostelium cells are constrained to designated microscopic regions are difficult to make. Here we present a micropatterning technique for Dictyostelium cells that relies on coating the substrate with an ∼1μm thick layer of polyethylene glycol (PEG) gel. We show that, when plated on a substrate with narrow parallel stripes of PEG-gel and glass, Dictyostelium cells nearly exclusive adhere to and migrate along the glass stripes, thus providing a model system to study one-dimensional migration of amoeboid cells. Surprisingly, we find substantial differences in the adhesion to PEG-gel and glass stripes between vegetative and developed cells and between two different axenic laboratory strains of Dictyostelium, AX2 and AX4. Even more surprisingly, we find that the distribution of Dictyostelium cells between PEG-gel and glass stripes is significantly affected by the expression of several fluorescent protein markers of the cytoskeleton. We carry out atomic force microscopy based single cell force spectroscopy measurements that confirm that the force of adhesion to PEG-gel substrate can be significantly different between vegetative and developed cells, AX2 and AX4 cells, and cells with and without fluorescent markers. Thus, the choice of parental background, the degree of development, and the expression of fluorescent protein markers can all have a profound effect on cell-substrate adhesion and should be considered when comparing migration of cells and when designing micropatterned substrates.
Collapse
Affiliation(s)
- Richa Karmakar
- Department of Physics, University of California, San Diego, La Jolla, California, United States of America
| | | | - Nadine Kamprad
- Institute for Dynamics of Complex Systems, Goettingen, Germany.,Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | | | - Edgar Gutierrez
- Department of Physics, University of California, San Diego, La Jolla, California, United States of America
| | - Alex Groisman
- Department of Physics, University of California, San Diego, La Jolla, California, United States of America
| | - Wouter-Jan Rappel
- Department of Physics, University of California, San Diego, La Jolla, California, United States of America
| | - Marco Tarantola
- Institute for Dynamics of Complex Systems, Goettingen, Germany.,Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| |
Collapse
|
31
|
Saraswathibhatla A, Galles EE, Notbohm J. Spatiotemporal force and motion in collective cell migration. Sci Data 2020; 7:197. [PMID: 32581285 PMCID: PMC7314837 DOI: 10.1038/s41597-020-0540-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/15/2020] [Indexed: 11/08/2022] Open
Abstract
Cells move in collective groups in biological processes such as wound healing, morphogenesis, and cancer metastasis. How active cell forces produce the motion in collective cell migration is still unclear. Many theoretical models have been introduced to elucidate the relationship between the cell's active forces and different observations about the collective motion such as collective swirls, oscillations, and rearrangements. Though many models share the common feature of balancing forces in the cell layer, the specific relationships between force and motion vary among the different models, which can lead to different conclusions. Simultaneous experimental measurements of force and motion can aid in testing assumptions and predictions of the theoretical models. Here, we provide time-lapse images of cells in 1 mm circular islands, which are used to compute cell velocities, cell-substrate tractions, and monolayer stresses. Additional data are included from experiments that perturbed cell number density and actomyosin contractility. We expect this data set to be useful to researchers interested in force and motion in collective cell migration.
Collapse
Affiliation(s)
| | - Emmett E Galles
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jacob Notbohm
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
32
|
Hur SS, Jeong JH, Ban MJ, Park JH, Yoon JK, Hwang Y. Traction force microscopy for understanding cellular mechanotransduction. BMB Rep 2020. [PMID: 31964473 PMCID: PMC7061206 DOI: 10.5483/bmbrep.2020.53.2.308] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Under physiological and pathological conditions, mechanical forces generated from cells themselves or transmitted from extracellular matrix (ECM) through focal adhesions (FAs) and adherens junctions (AJs) are known to play a significant role in regulating various cell behaviors. Substantial progresses have been made in the field of mechanobiology towards novel methods to understand how cells are able to sense and adapt to these mechanical forces over the years. To address these issues, this review will discuss recent advancements of traction force microscopy (TFM), intracellular force microscopy (IFM), and monolayer stress microscopy (MSM) to measure multiple aspects of cellular forces exerted by cells at cell-ECM and cell-cell junctional intracellular interfaces. We will also highlight how these methods can elucidate the roles of mechanical forces at interfaces of cell-cell/cell-ECM in regulating various cellular functions. [BMB Reports 2020; 53(2): 74-81].
Collapse
Affiliation(s)
- Sung Sik Hur
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan 31151; Department of Integrated Biomedical Science, Soonchunhyang University, Asan 31538, Korea
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan 31151; Department of Integrated Biomedical Science, Soonchunhyang University, Asan 31538, Korea
| | - Myung Jin Ban
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Jae Hong Park
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Jeong Kyo Yoon
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan 31151; Department of Integrated Biomedical Science, Soonchunhyang University, Asan 31538, Korea
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan 31151; Department of Integrated Biomedical Science, Soonchunhyang University, Asan 31538, Korea
| |
Collapse
|
33
|
Steuwe C, Vaeyens MM, Jorge-Peñas A, Cokelaere C, Hofkens J, Roeffaers MBJ, Van Oosterwyck H. Fast quantitative time lapse displacement imaging of endothelial cell invasion. PLoS One 2020; 15:e0227286. [PMID: 31910228 PMCID: PMC6946139 DOI: 10.1371/journal.pone.0227286] [Citation(s) in RCA: 6] [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: 08/26/2019] [Accepted: 12/16/2019] [Indexed: 11/18/2022] Open
Abstract
In order to unravel rapid mechano-chemical feedback mechanisms in sprouting angiogenesis, we combine selective plane illumination microscopy (SPIM) and tailored image registration algorithms - further referred to as SPIM-based displacement microscopy - with an in vitro model of angiogenesis. SPIM successfully tackles the problem of imaging large volumes while upholding the spatial resolution required for the analysis of matrix displacements at a subcellular level. Applied to in vitro angiogenic sprouts, this unique methodological combination relates subcellular activity - minute to second time scale growing and retracting of protrusions - of a multicellular systems to the surrounding matrix deformations with an exceptional temporal resolution of 1 minute for a stack with multiple sprouts simultaneously or every 4 seconds for a single sprout, which is 20 times faster than with a conventional confocal setup. Our study reveals collective but non-synchronised, non-continuous activity of adjacent sprouting cells along with correlations between matrix deformations and protrusion dynamics.
Collapse
Affiliation(s)
- Christian Steuwe
- Centre for Membrane Separations, Adsorption, Catalysis and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular Systems (MS), KU Leuven, Leuven, Belgium
| | - Marie-Mo Vaeyens
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Alvaro Jorge-Peñas
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Célie Cokelaere
- Centre for Membrane Separations, Adsorption, Catalysis and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular Systems (MS), KU Leuven, Leuven, Belgium
| | - Johan Hofkens
- Molecular Imaging and Photonics, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Maarten B. J. Roeffaers
- Centre for Membrane Separations, Adsorption, Catalysis and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular Systems (MS), KU Leuven, Leuven, Belgium
| | - Hans Van Oosterwyck
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Bastounis EE, Yeh YT, Theriot JA. Subendothelial stiffness alters endothelial cell traction force generation while exerting a minimal effect on the transcriptome. Sci Rep 2019; 9:18209. [PMID: 31796790 PMCID: PMC6890669 DOI: 10.1038/s41598-019-54336-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023] Open
Abstract
Endothelial cells respond to changes in subendothelial stiffness by altering their migration and mechanics, but whether those responses are due to transcriptional reprogramming remains largely unknown. We measured traction force generation and also performed gene expression profiling for two endothelial cell types grown in monolayers on soft or stiff matrices: primary human umbilical vein endothelial cells (HUVEC) and immortalized human microvascular endothelial cells (HMEC-1). Both cell types respond to changes in subendothelial stiffness by increasing the traction stresses they exert on stiffer as compared to softer matrices, and exhibit a range of altered protein phosphorylation or protein conformational changes previously implicated in mechanotransduction. However, the transcriptome has only a minimal role in this conserved biomechanical response. Only few genes were differentially expressed in each cell type in a stiffness-dependent manner, and none were shared between them. In contrast, thousands of genes were differentially regulated in HUVEC as compared to HMEC-1. HUVEC (but not HMEC-1) upregulate expression of TGF-β2 on stiffer matrices, and also respond to application of exogenous TGF-β2 by enhancing their endogenous TGF-β2 expression and their cell-matrix traction stresses. Altogether, these findings provide insights into the relationship between subendothelial stiffness, endothelial mechanics and variation of the endothelial cell transcriptome, and reveal that subendothelial stiffness, while critically altering endothelial cells’ mechanical behavior, minimally affects their transcriptome.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195-1800, USA
| | - Yi-Ting Yeh
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195-1800, USA.
| |
Collapse
|
35
|
Notbohm J, Napiwocki B, deLange W, Stempien A, Saraswathibhatla A, Craven R, Salick M, Ralphe J, Crone W. Two-Dimensional Culture Systems to Enable Mechanics-Based Assays for Stem Cell-Derived Cardiomyocytes. EXPERIMENTAL MECHANICS 2019; 59:1235-1248. [PMID: 31680699 PMCID: PMC6824432 DOI: 10.1007/s11340-019-00473-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 01/09/2019] [Indexed: 06/10/2023]
Abstract
Well-controlled 2D cell culture systems advance basic investigations in cell biology and provide innovative platforms for drug development, toxicity testing, and diagnostic assays. These cell culture systems have become more advanced in order to provide and to quantify the appropriate biomechanical and biochemical cues that mimic the milieu of conditions present in vivo. Here we present an innovative 2D cell culture system to investigate human stem cell-derived cardiomyocytes, the muscle cells of the heart responsible for pumping blood throughout the body. We designed our 2D cell culture platform to control intracellular features to produce adult-like cardiomyocyte organization with connectivity and anisotropic conduction comparable to the native heart, and combined it with optical microscopy to quantify cell-cell and cell-substrate mechanical interactions. We show the measurement of forces and displacements that occur within individual cells, between neighboring cells, and between cells and their surrounding matrix. This system has broad potential to expand our understanding of tissue physiology, with particular advantages for the study of the mechanically active heart. Furthermore, this technique should prove valuable in screening potential drugs for efficacy and testing for toxicity.
Collapse
Affiliation(s)
- J. Notbohm
- Department of Engineering Physics, University of Wisconsin-Madison, Madison WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
| | - B.N. Napiwocki
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
| | - W.J. deLange
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - A. Stempien
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
| | - A. Saraswathibhatla
- Department of Engineering Physics, University of Wisconsin-Madison, Madison WI, USA
| | - R.J. Craven
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
| | - M.R. Salick
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison WI, USA
| | - J.C. Ralphe
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - W.C. Crone
- Department of Engineering Physics, University of Wisconsin-Madison, Madison WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison WI, USA
| |
Collapse
|
36
|
Cao Y, Ghabache E, Rappel WJ. Plasticity of cell migration resulting from mechanochemical coupling. eLife 2019; 8:e48478. [PMID: 31625907 PMCID: PMC6799977 DOI: 10.7554/elife.48478] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/02/2019] [Indexed: 01/07/2023] Open
Abstract
Eukaryotic cells can migrate using different modes, ranging from amoeboid-like, during which actin filled protrusions come and go, to keratocyte-like, characterized by a stable morphology and persistent motion. How cells can switch between these modes is not well understood but waves of signaling events are thought to play an important role in these transitions. Here we present a simple two-component biochemical reaction-diffusion model based on relaxation oscillators and couple this to a model for the mechanics of cell deformations. Different migration modes, including amoeboid-like and keratocyte-like, naturally emerge through transitions determined by interactions between biochemical traveling waves, cell mechanics and morphology. The model predictions are explicitly verified by systematically reducing the protrusive force of the actin network in experiments using Dictyostelium discoideum cells. Our results indicate the importance of coupling signaling events to cell mechanics and morphology and may be applicable in a wide variety of cell motility systems.
Collapse
Affiliation(s)
- Yuansheng Cao
- Department of PhysicsUniversity of California, San DiegoLa JollaUnited States
| | - Elisabeth Ghabache
- Department of PhysicsUniversity of California, San DiegoLa JollaUnited States
| | - Wouter-Jan Rappel
- Department of PhysicsUniversity of California, San DiegoLa JollaUnited States
| |
Collapse
|
37
|
Huang J, Lin F, Xiong C. Mechanical characterization of single cells based on microfluidic techniques. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.07.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
38
|
Feng J, Levine H, Mao X, Sander LM. Cell motility, contact guidance, and durotaxis. SOFT MATTER 2019; 15:4856-4864. [PMID: 31161163 DOI: 10.1039/c8sm02564a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Mechanical properties of the substrate play a vital role in cell motility. In particular, cells have been shown to migrate along aligned fibers in the substrate (contact guidance) and up stiffness gradients (durotaxis). Here we present a simple mechanical model for cell migration coupled to substrate properties, by placing a simulated cell on a lattice mimicking biopolymer gels or hydrogels. In our model cells attach to the substrate via focal adhesions (FAs). As the cells contract, forces are generated at the FAs, determining their maturation and detachment. At the same time, the cell was also allowed to move and rotate to maintain force and torque balance. Our model, in which the cells only have access to information regarding forces acting at the FAs, without a prior knowledge of the substrate stiffness or geometry, is able to reproduce both contact guidance and durotaxis.
Collapse
Affiliation(s)
- Jingchen Feng
- Center for Theoretical Biological Physics, Rice University, 6100 Main Street, Houston, TX 77005-1892, USA.
| | | | | | | |
Collapse
|
39
|
Yeh YT, Wei J, Thorossian S, Nguyen K, Hoffman C, Del Álamo JC, Serrano R, Li YSJ, Wang KC, Chien S. MiR-145 mediates cell morphology-regulated mesenchymal stem cell differentiation to smooth muscle cells. Biomaterials 2019; 204:59-69. [PMID: 30884320 PMCID: PMC6825513 DOI: 10.1016/j.biomaterials.2019.03.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/17/2019] [Accepted: 03/01/2019] [Indexed: 01/14/2023]
Abstract
The use of biochemical signaling to derive smooth muscle cells (SMCs) from mesenchymal stem cells (MSCs) has been explored, but the induction of a fully functional SMC phenotype remains to be a major challenge. Cell morphology has been shown to regulate MSC differentiation into various lineages, including SMCs. We engineered substrates with microgrooves to induce cell elongation to study the mechanism underlying the MSC shape modulation in SMC differentiation. In comparison to those on flat substrates, MSCs cultured on engineered substrates were elongated with increased aspect ratios for both cell body and nucleus, as well as augmented cytoskeletal tensions. Biochemical studies indicated that the microgroove-elongated cells expressed significantly higher levels of SMC markers. MicroRNA analyses showed that up-regulation of miR-145 and the consequent repression of KLF4 in these elongated cells promoted MSC-to-SMC differentiation. Rho/ROCK inhibitions, which impair cytoskeletal tension, attenuated cell and nuclear elongations and disrupted the miR-145/KLF4 regulation for SMC differentiation. Furthermore, cell traction force measurements showed that miR-145 is essential for the functional contractility in the microgroove-induced SMC differentiation. Collectively, our findings demonstrate that, through a Rho-ROCK/miR-145/KLF4 pathway, the elongated cell shape serves as a decisive geometric cue to direct MSC differentiation into functional SMCs.
Collapse
Affiliation(s)
- Yi-Ting Yeh
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, 92093, United States; Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Josh Wei
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Satenick Thorossian
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Katherine Nguyen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Clarissa Hoffman
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Juan C Del Álamo
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, 92093, United States; Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Yi-Shuan Julie Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, 92093, United States
| | - Kuei-Chun Wang
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, 92093, United States.
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, United States; Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, 92093, United States.
| |
Collapse
|
40
|
Serrano R, Aung A, Yeh YT, Varghese S, Lasheras JC, Del Álamo JC. Three-Dimensional Monolayer Stress Microscopy. Biophys J 2019; 117:111-128. [PMID: 31103228 DOI: 10.1016/j.bpj.2019.03.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 03/07/2019] [Accepted: 03/28/2019] [Indexed: 01/06/2023] Open
Abstract
Many biological processes involve the collective generation and transmission of mechanical stresses across cell monolayers. In these processes, the monolayer undergoes lateral deformation and bending because of the tangential and normal components of the cell-generated stresses. Monolayer stress microscopy (MSM) methods have been developed to measure the intracellular stress distribution in cell monolayers. However, current methods assume plane monolayer geometry and neglect the contribution of bending to the intracellular stresses. This work introduces a three-dimensional (3D) MSM method that calculates monolayer stress from measurements of the 3D traction stresses exerted by the cells on a flexible substrate. The calculation is carried out by imposing equilibrium of forces and moments in the monolayer, subject to external loads given by the 3D traction stresses. The equilibrium equations are solved numerically, and the algorithm is validated for synthetic loads with known analytical solutions. We present 3D-MSM measurements of monolayer stress in micropatterned islands of endothelial cells of different sizes and shapes. These data indicate that intracellular stresses caused by lateral deformation emerge collectively over long distances; they increase with the distance from the island edge until they reach a constant value that is independent of island size. On the other hand, bending-induced intracellular stresses are more concentrated spatially and remain confined to within one to two cell lengths of bending sites. The magnitude of these bending stresses is highest at the edges of the cell islands, where they can exceed the intracellular stresses caused by lateral deformations. Our data from nonpatterned monolayers suggests that biomechanical perturbations far away from monolayer edges also cause significant localized alterations in bending tension. The localized effect of bending-induced stresses may be important in processes like cellular extravasation, which are accompanied by significant normal deflections of a cell monolayer (i.e., the endothelium) and require localized changes in monolayer permeability.
Collapse
Affiliation(s)
- Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, San Diego, San Diego, California.
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, San Diego, San Diego, California; Department of Bioengineering, San Diego, San Diego, California; Institute of Engineering in Medicine, University of California, San Diego, San Diego, California
| | - Shyni Varghese
- Department of Biomedical Engineering, Durham, North Carolina; Department of Mechanical Engineering and Material Sciences, Durham, North Carolina; Department of Orthopaedic Surgery, Duke University, Durham, North Carolina
| | - Juan C Lasheras
- Department of Mechanical and Aerospace Engineering, San Diego, San Diego, California; Department of Bioengineering, San Diego, San Diego, California; Institute of Engineering in Medicine, University of California, San Diego, San Diego, California
| | - Juan C Del Álamo
- Department of Mechanical and Aerospace Engineering, San Diego, San Diego, California.
| |
Collapse
|
41
|
Machicoane N, Aliseda A, Volk R, Bourgoin M. A simplified and versatile calibration method for multi-camera optical systems in 3D particle imaging. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2019; 90:035112. [PMID: 30927766 DOI: 10.1063/1.5080743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/28/2019] [Indexed: 06/09/2023]
Abstract
This article describes a stereoscopic multi-camera calibration method that does not require any optical model. It is based on a measure of the light propagation within the measurement volume only instead of modeling its entire path up to the sensors. The calibration uses simple plane by plane transformations which allow us to directly link pixel coordinates to light rays. The appeal of the proposed method relies on the combination of its simplicity of implementation (it is particularly easy to apply in any sophisticated optical imaging setup), its versatility (it can easily handle index-of-refraction gradients, as well as complex optical arrangements), and its accuracy {we show that the proposed method gives better accuracy than commonly used techniques, based on Tsai's simple pinhole camera model [R. Tsai, J. Rob. Autom. 3, 323 (1987)], while its numerical implementation remains extremely simple}. Based on ideas that have been available in the fluid mechanics community, this method is a compact turn-key algorithm that can be implemented with open-source routines.
Collapse
Affiliation(s)
- N Machicoane
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, USA
| | - A Aliseda
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, USA
| | - R Volk
- University of Lyon, ENS de Lyon, Université Claude Bernard, CNRS, Laboratoire de Physique, F-69342 Lyon, France
| | - M Bourgoin
- University of Lyon, ENS de Lyon, Université Claude Bernard, CNRS, Laboratoire de Physique, F-69342 Lyon, France
| |
Collapse
|
42
|
Zhang S, Skinner D, Joshi P, Criado-Hidalgo E, Yeh YT, Lasheras JC, Caffrey CR, del Alamo JC. Quantifying the mechanics of locomotion of the schistosome pathogen with respect to changes in its physical environment. J R Soc Interface 2019; 16:20180675. [PMID: 30958153 PMCID: PMC6364656 DOI: 10.1098/rsif.2018.0675] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/21/2018] [Indexed: 01/13/2023] Open
Abstract
Schistosomiasis is a chronic and morbid disease of poverty affecting approximately 200 million people worldwide. Mature schistosome flatworms wander in the host's hepatic portal and mesenteric venous system where they encounter a range of blood flow conditions and geometrical confinement. However, the mechanisms that support schistosome locomotion and underlie the pathogen's adaptation to its physical environment are largely unknown. By combining microfabrication and traction force microscopy, we developed various in vitro assays to quantify the mechanics of locomotion of adult male Schistosoma mansoni in different physiologically relevant conditions. We show that in unconfined settings, the parasite undergoes two-anchor marching mediated by the coordinated action of its oral and ventral suckers. This mode of locomotion is maintained when the worm faces an external flow, to which it responds by adjusting the strength of its suckers. In geometrically confined conditions, S. mansoni switches to a different crawling modality by generating retrograde peristaltic waves along its body, a mechanism shared with terrestrial and marine worms. However, while the surface of most worms has backward-pointing bristles that rectify peristaltic waves and facilitate forward locomotion, S. mansoni has isotropically oriented tubercles. This requires tight coordination between muscle contraction and substrate friction but gives S. mansoni the ability to reverse its direction of locomotion without turning its body, which is likely advantageous to manoeuvre in narrow-bore vessels. We show that the parasite can also coordinate the action of its suckers with its peristaltic body contractions to increase crawling speed. Throughout this study, we report on a number of biomechanical parameters to quantify the motility of adult schistosomes (e.g. sucker grabbing strength, the rate of detachment under flow, peristaltic wave properties and traction stresses). The new series of in vitro assays make it possible to quantify key phenotypical aspects of S. mansoni motility that could guide the discovery of new drugs to treat schistosomiasis.
Collapse
Affiliation(s)
- Shun Zhang
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, USA
| | - Danielle Skinner
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Prateek Joshi
- School of Engineering, Brown University, Providence, RI, USA
| | - Ernesto Criado-Hidalgo
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, USA
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, USA
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Institute for Engineering in Medicine, University of California San Diego, San Diego, CA, USA
| | - Juan C. Lasheras
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, USA
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Institute for Engineering in Medicine, University of California San Diego, San Diego, CA, USA
| | - Conor R. Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Juan C. del Alamo
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, USA
- Institute for Engineering in Medicine, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
43
|
Cortes DB, Dawes A, Liu J, Nickaeen M, Strychalski W, Maddox AS. Unite to divide - how models and biological experimentation have come together to reveal mechanisms of cytokinesis. J Cell Sci 2018; 131:131/24/jcs203570. [PMID: 30563924 DOI: 10.1242/jcs.203570] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Cytokinesis is the fundamental and ancient cellular process by which one cell physically divides into two. Cytokinesis in animal and fungal cells is achieved by contraction of an actomyosin cytoskeletal ring assembled in the cell cortex, typically at the cell equator. Cytokinesis is essential for the development of fertilized eggs into multicellular organisms and for homeostatic replenishment of cells. Correct execution of cytokinesis is also necessary for genome stability and the evasion of diseases including cancer. Cytokinesis has fascinated scientists for well over a century, but its speed and dynamics make experiments challenging to perform and interpret. The presence of redundant mechanisms is also a challenge to understand cytokinesis, leaving many fundamental questions unresolved. For example, how does a disordered cytoskeletal network transform into a coherent ring? What are the long-distance effects of localized contractility? Here, we provide a general introduction to 'modeling for biologists', and review how agent-based modeling and continuum mechanics modeling have helped to address these questions.
Collapse
Affiliation(s)
- Daniel B Cortes
- Department of Biology, University of North Carolina at Chapel Hill, 407 Fordham Hall, Chapel Hill, NC 27599, USA
| | - Adriana Dawes
- Departments of Mathematics and of Molecular Genetics, The Ohio State University, 100 Math Tower, 231 West 18th Avenue, Columbus, OH 43210, USA
| | - Jian Liu
- National Heart, Lung and Blood Institute, Biochemistry and Biophysics Center, 50 South Drive, NIH, Bethesda, MD 20892, USA
| | - Masoud Nickaeen
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Department of Cell Biology, 263 Farmington Avenue, Farmington, CT 06030-6406, USA
| | - Wanda Strychalski
- Department of Mathematics, Applied Mathematics, and Statistics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Amy Shaub Maddox
- Department of Biology, University of North Carolina at Chapel Hill, 407 Fordham Hall, Chapel Hill, NC 27599, USA
| |
Collapse
|
44
|
Lo Sardo V, Chubukov P, Ferguson W, Kumar A, Teng EL, Duran M, Zhang L, Cost G, Engler AJ, Urnov F, Topol EJ, Torkamani A, Baldwin KK. Unveiling the Role of the Most Impactful Cardiovascular Risk Locus through Haplotype Editing. Cell 2018; 175:1796-1810.e20. [PMID: 30528432 DOI: 10.1016/j.cell.2018.11.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 04/24/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023]
Abstract
The 9p21.3 cardiovascular disease locus is the most influential common genetic risk factor for coronary artery disease (CAD), accounting for ∼10%-15% of disease in non-African populations. The ∼60 kb risk haplotype is human-specific and lacks coding genes, hindering efforts to decipher its function. Here, we produce induced pluripotent stem cells (iPSCs) from risk and non-risk individuals, delete each haplotype using genome editing, and generate vascular smooth muscle cells (VSMCs). Risk VSMCs exhibit globally altered transcriptional networks that intersect with previously identified CAD risk genes and pathways, concomitant with aberrant adhesion, contraction, and proliferation. Unexpectedly, deleting the risk haplotype rescues VSMC stability, while expressing the 9p21.3-associated long non-coding RNA ANRIL induces risk phenotypes in non-risk VSMCs. This study shows that the risk haplotype selectively predisposes VSMCs to adopt a cell state associated with CAD phenotypes, defines new VSMC-based networks of CAD risk genes, and establishes haplotype-edited iPSCs as powerful tools for functionally annotating the human genome.
Collapse
Affiliation(s)
- Valentina Lo Sardo
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Pavel Chubukov
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - William Ferguson
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Aditya Kumar
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093-0412, USA
| | - Evan L Teng
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093-0412, USA
| | - Michael Duran
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Lei Zhang
- Sangamo BioSciences, Inc., Richmond, CA 94804-3517, USA
| | - Gregory Cost
- Sangamo BioSciences, Inc., Richmond, CA 94804-3517, USA
| | - Adam J Engler
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093-0412, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Fyodor Urnov
- Sangamo BioSciences, Inc., Richmond, CA 94804-3517, USA
| | - Eric J Topol
- Scripps Research Translational Institute, The Scripps Research Institute, La Jolla, CA, 92037, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, CA
| | - Ali Torkamani
- Scripps Research Translational Institute, The Scripps Research Institute, La Jolla, CA, 92037, USA; Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kristin K Baldwin
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
45
|
Sharma A, McKeithan WL, Serrano R, Kitani T, Burridge PW, Del Álamo JC, Mercola M, Wu JC. Use of human induced pluripotent stem cell-derived cardiomyocytes to assess drug cardiotoxicity. Nat Protoc 2018; 13:3018-3041. [PMID: 30413796 PMCID: PMC6502639 DOI: 10.1038/s41596-018-0076-8] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cardiotoxicity has historically been a major cause of drug removal from the pharmaceutical market. Several chemotherapeutic compounds have been noted for their propensities to induce dangerous cardiac-specific side effects such as arrhythmias or cardiomyocyte apoptosis. However, improved preclinical screening methodologies have enabled cardiotoxic compounds to be identified earlier in the drug development pipeline. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can be used to screen for drug-induced alterations in cardiac cellular contractility, electrophysiology, and viability. We previously established a novel 'cardiac safety index' (CSI) as a metric that can evaluate potential cardiotoxic drugs via high-throughput screening of hiPSC-CMs. This metric quantitatively examines drug-induced alterations in CM function, using several in vitro readouts, and normalizes the resulting toxicity values to the in vivo maximum drug blood plasma concentration seen in preclinical or clinical pharmacokinetic models. In this ~1-month-long protocol, we describe how to differentiate hiPSCs into hiPSC-CMs and subsequently implement contractility and cytotoxicity assays that can evaluate drug-induced cardiotoxicity in hiPSC-CMs. We also describe how to carry out the calculations needed to generate the CSI metric from these quantitative toxicity measurements.
Collapse
Affiliation(s)
- Arun Sharma
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Wesley L McKeithan
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, CA, USA
| | - Tomoya Kitani
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul W Burridge
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University School of Medicine, Chicago, IL, USA
| | - Juan C Del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, CA, USA
| | - Mark Mercola
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
46
|
Basoli F, Giannitelli SM, Gori M, Mozetic P, Bonfanti A, Trombetta M, Rainer A. Biomechanical Characterization at the Cell Scale: Present and Prospects. Front Physiol 2018; 9:1449. [PMID: 30498449 PMCID: PMC6249385 DOI: 10.3389/fphys.2018.01449] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
The rapidly growing field of mechanobiology demands for robust and reproducible characterization of cell mechanical properties. Recent achievements in understanding the mechanical regulation of cell fate largely rely on technological platforms capable of probing the mechanical response of living cells and their physico–chemical interaction with the microenvironment. Besides the established family of atomic force microscopy (AFM) based methods, other approaches include optical, magnetic, and acoustic tweezers, as well as sensing substrates that take advantage of biomaterials chemistry and microfabrication techniques. In this review, we introduce the available methods with an emphasis on the most recent advances, and we discuss the challenges associated with their implementation.
Collapse
Affiliation(s)
- Francesco Basoli
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | | | - Manuele Gori
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Pamela Mozetic
- Center for Translational Medicine, International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Alessandra Bonfanti
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Marcella Trombetta
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Alberto Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy.,Institute for Photonics and Nanotechnologies, National Research Council, Rome, Italy
| |
Collapse
|
47
|
Bastounis EE, Ortega FE, Serrano R, Theriot JA. A Multi-well Format Polyacrylamide-based Assay for Studying the Effect of Extracellular Matrix Stiffness on the Bacterial Infection of Adherent Cells. J Vis Exp 2018. [PMID: 30035758 PMCID: PMC6124605 DOI: 10.3791/57361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Extracellular matrix stiffness comprises one of the multiple environmental mechanical stimuli that are well known to influence cellular behavior, function, and fate in general. Although increasingly more adherent cell types' responses to matrix stiffness have been characterized, how adherent cells' susceptibility to bacterial infection depends on matrix stiffness is largely unknown, as is the effect of bacterial infection on the biomechanics of host cells. We hypothesize that the susceptibility of host endothelial cells to a bacterial infection depends on the stiffness of the matrix on which these cells reside, and that the infection of the host cells with bacteria will change their biomechanics. To test these two hypotheses, endothelial cells were used as model hosts and Listeria monocytogenes as a model pathogen. By developing a novel multi-well format assay, we show that the effect of matrix stiffness on infection of endothelial cells by L. monocytogenes can be quantitatively assessed through flow cytometry and immunostaining followed by microscopy. In addition, using traction force microscopy, the effect of L. monocytogenes infection on host endothelial cell biomechanics can be studied. The proposed method allows for the analysis of the effect of tissue-relevant mechanics on bacterial infection of adherent cells, which is a critical step towards understanding the biomechanical interactions between cells, their extracellular matrix, and pathogenic bacteria. This method is also applicable to a wide variety of other types of studies on cell biomechanics and response to substrate stiffness where it is important to be able to perform many replicates in parallel in each experiment.
Collapse
Affiliation(s)
| | - Fabian E Ortega
- Department of Biochemistry, Stanford University School of Medicine
| | - Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, University of California San Diego
| | - Julie A Theriot
- Departments of Biochemistry, Microbiology and Immunology and Howard Hughes Medical Institute, Stanford University School of Medicine
| |
Collapse
|
48
|
Kulkarni AH, Ghosh P, Seetharaman A, Kondaiah P, Gundiah N. Traction cytometry: regularization in the Fourier approach and comparisons with finite element method. SOFT MATTER 2018; 14:4687-4695. [PMID: 29740649 DOI: 10.1039/c7sm02214j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Traction forces exerted by adherent cells are quantified using displacements of embedded markers on polyacrylamide substrates due to cell contractility. Fourier Transform Traction Cytometry (FTTC) is widely used to calculate tractions but has inherent limitations due to errors in the displacement fields; these are mitigated through a regularization parameter (γ) in the Reg-FTTC method. An alternate finite element (FE) approach computes tractions on a domain using known boundary conditions. Robust verification and recovery studies are lacking but essential in assessing the accuracy and noise sensitivity of the traction solutions from the different methods. We implemented the L2 regularization method and defined a maximum curvature point in the traction with γ plot as the optimal regularization parameter (γ*) in the Reg-FTTC approach. Traction reconstructions using γ* yield accurate values of low and maximum tractions (Tmax) in the presence of up to 5% noise. Reg-FTTC is hence a clear improvement over the FTTC method but is inadequate to reconstruct low stresses such as those at nascent focal adhesions. FE, implemented using a node-by-node comparison, showed an intermediate reconstruction compared to Reg-FTTC. We performed experiments using mouse embryonic fibroblast (MEF) and compared results between these approaches. Tractions from FTTC and FE showed differences of ∼92% and 22% as compared to Reg-FTTC. Selection of an optimum value of γ for each cell reduced variability in the computed tractions as compared to using a single value of γ for all the MEF cells in this study.
Collapse
Affiliation(s)
- Ankur H Kulkarni
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | | | | | | | | |
Collapse
|
49
|
Three-dimensional simulation of obstacle-mediated chemotaxis. Biomech Model Mechanobiol 2018; 17:1243-1268. [DOI: 10.1007/s10237-018-1023-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 04/25/2018] [Indexed: 01/07/2023]
|
50
|
Faralla C, Bastounis EE, Ortega FE, Light SH, Rizzuto G, Gao L, Marciano DK, Nocadello S, Anderson WF, Robbins JR, Theriot JA, Bakardjiev AI. Listeria monocytogenes InlP interacts with afadin and facilitates basement membrane crossing. PLoS Pathog 2018; 14:e1007094. [PMID: 29847585 PMCID: PMC6044554 DOI: 10.1371/journal.ppat.1007094] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 07/13/2018] [Accepted: 05/11/2018] [Indexed: 12/14/2022] Open
Abstract
During pregnancy, the placenta protects the fetus against the maternal immune response, as well as bacterial and viral pathogens. Bacterial pathogens that have evolved specific mechanisms of breaching this barrier, such as Listeria monocytogenes, present a unique opportunity for learning how the placenta carries out its protective function. We previously identified the L. monocytogenes protein Internalin P (InlP) as a secreted virulence factor critical for placental infection. Here, we show that InlP, but not the highly similar L. monocytogenes internalin Lmo2027, binds to human afadin (encoded by AF-6), a protein associated with cell-cell junctions. A crystal structure of InlP reveals several unique features, including an extended leucine-rich repeat (LRR) domain with a distinctive Ca2+-binding site. Despite afadin's involvement in the formation of cell-cell junctions, MDCK epithelial cells expressing InlP displayed a decrease in the magnitude of the traction stresses they could exert on deformable substrates, similar to the decrease in traction exhibited by AF-6 knock-out MDCK cells. L. monocytogenes ΔinlP mutants were deficient in their ability to form actin-rich protrusions from the basal face of polarized epithelial monolayers, a necessary step in the crossing of such monolayers (transcytosis). A similar phenotype was observed for bacteria expressing an internal in-frame deletion in inlP (inlP ΔLRR5) that specifically disrupts its interaction with afadin. However, afadin deletion in the host cells did not rescue the transcytosis defect. We conclude that secreted InlP targets cytosolic afadin to specifically promote L. monocytogenes transcytosis across the basal face of epithelial monolayers, which may contribute to the crossing of the basement membrane during placental infection.
Collapse
Affiliation(s)
- Cristina Faralla
- Benioff Children’s Hospital, University of California, San Francisco, San Francisco, California, United States of America
- Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, California, United States of America
| | - Effie E. Bastounis
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Fabian E. Ortega
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Samuel H. Light
- Center for Structural Genomics of Infectious Diseases and Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Gabrielle Rizzuto
- Benioff Children’s Hospital, University of California, San Francisco, San Francisco, California, United States of America
- Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| | - Lei Gao
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Denise K. Marciano
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Salvatore Nocadello
- Center for Structural Genomics of Infectious Diseases and Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Wayne F. Anderson
- Center for Structural Genomics of Infectious Diseases and Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Jennifer R. Robbins
- Department of Biology, Xavier University, Cincinnati, Ohio, United States of America
| | - Julie A. Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Anna I. Bakardjiev
- Benioff Children’s Hospital, University of California, San Francisco, San Francisco, California, United States of America
- Program in Microbial Pathogenesis and Host Defense, University of California, San Francisco, San Francisco, California, United States of America
| |
Collapse
|