1
|
Dong Y, Wang C, Ding X, Ma X, Huang R, Li M, Yang Q. The characterization of cell traction force on nonflat surfaces with different curvature by elastic hydrogel microspheres. Biotechnol Bioeng 2024; 121:3537-3550. [PMID: 38978386 DOI: 10.1002/bit.28802] [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: 03/20/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
It is of great importance to study the detachment/attachment behaviors of cells (cancer cell, immune cell, and epithelial cell), as they are closely related with tumor metastasis, immunoreaction, and tissue development at variety scales. To characterize the detachment/attachment during the interaction between cells and substrate, some researchers proposed using cell traction force (CTF) as the indicator. To date, various strategies have been developed to measure the CTF. However, these methods only realize the measurements of cell passive forces on flat cases. To quantify the active CTF on nonflat surfaces, which can better mimic the in vivo case, we employed elastic hydrogel microspheres as a force sensor. The microspheres were fabricated by microfluidic chips with controllable size and mechanical properties to mimic substrate. Cells were cultured on microsphere and the CTF led to the deformation of microsphere. By detecting the morphology information, the CTF exerted by attached cells can be calculated by the in-house numerical code. Using these microspheres, the CTF of various cells (including tumor cell, immunological cell, and epithelium cell) were successfully obtained on nonflat surfaces with different curvature radii. The proposed method provides a versatile platform to measure the CTF with high precision and to understand the detachment/attachment behaviors during physiology processes.
Collapse
Affiliation(s)
- Yuqing Dong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Cong Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xin Ding
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xingquan Ma
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
- School of Civil Engineering and Architecture, Xi'an University of Technology, Xi'an, P.R. China
| | - Rong Huang
- Department of Burn and Plastic Surgery, Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Moxiao Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
- Nanjing Center for Multifunctional Lightweight Materials and Structures (MLMS), Nanjing University of Aeronautics and Astronautics, Nanjing, P.R. China
| | - Qingzhen Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
2
|
Linke JA, Munn LL, Jain RK. Compressive stresses in cancer: characterization and implications for tumour progression and treatment. Nat Rev Cancer 2024; 24:768-791. [PMID: 39390249 DOI: 10.1038/s41568-024-00745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Beyond their many well-established biological aberrations, solid tumours create an abnormal physical microenvironment that fuels cancer progression and confers treatment resistance. Mechanical forces impact tumours across a range of biological sizes and timescales, from rapid events at the molecular level involved in their sensing and transmission, to slower and larger-scale events, including clonal selection, epigenetic changes, cell invasion, metastasis and immune response. Owing to challenges with studying these dynamic stimuli in biological systems, the mechanistic understanding of the effects and pathways triggered by abnormally elevated mechanical forces remains elusive, despite clear correlations with cancer pathophysiology, aggressiveness and therapeutic resistance. In this Review, we examine the emerging and diverse roles of physical forces in solid tumours and provide a comprehensive framework for understanding solid stress mechanobiology. We first review the physiological importance of mechanical forces, especially compressive stresses, and discuss their defining characteristics, biological context and relative magnitudes. We then explain how abnormal compressive stresses emerge in tumours and describe the experimental challenges in investigating these mechanically induced processes. Finally, we discuss the clinical translation of mechanotherapeutics that alleviate solid stresses and their potential to synergize with chemotherapy, radiotherapy and immunotherapies.
Collapse
Affiliation(s)
- Julia A Linke
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Rodriguez Navas M, Darling EM. Selection of Force Sensors for In Situ Measurement of Neotissue Microenvironments. Tissue Eng Part A 2024. [PMID: 39453885 DOI: 10.1089/ten.tea.2024.0192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024] Open
Abstract
Mechanical forces are a critical stimulus in both native and engineered tissues. Direct measurement of these microenvironmental forces has been challenging, particularly for cell-dense models. To address this, we previously developed hydrogel-based force sensors that are approximately the size of a cell and can be imaged over time to computationally assess the forces exerted by surrounding cells and matrix. The goal of this project was to identify how the physical characteristics of force sensors impact measurements. Sensors were varied in size, elastic modulus, and surface coating before being included in stem cell suspensions that then spontaneously self-assembled into spheroidal neotissues. Using this model of early mesenchymal condensation, we hypothesized that larger, softer sensors would provide greater sensitivity and precision, whereas protein coatings would influence the directionality of applied forces (tensile vs. compressive). These experiments were conducted using a high-content imaging system that allowed analysis of over a thousand sensors to evaluate the various conditions. Results indicated that measurement fidelity was highest for force sensors that had a diameter >20 µm and modulus ∼0.2 kPa. Extremely soft sensors deformed too much, whereas stiffer sensors deformed too little. Collagen and N-cadherin coatings, which replicated cell-matrix or cell-cell binding, respectively, allowed for tensile forces to be exerted on the sensors, with greater forces being observed for N-cadherin sensors in these highly cellular neotissue constructs. Uncoated sensors were universally compressed due to the lack of cell-sensor adhesion. Disruption of the actin cytoskeleton lessened microenvironmental forces, whereas disruption of microtubules had no measurable effect. Potential future applications of the technology include studies of in situ forces in developing tissues as well as a real-time sensor for monitoring the growth of engineered constructs.
Collapse
Affiliation(s)
- Marta Rodriguez Navas
- Institute for Biology, Engineering, and Medicine, Brown University, Providence, Rhode Island, USA
| | - Eric M Darling
- Institute for Biology, Engineering, and Medicine, Brown University, Providence, Rhode Island, USA
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
- School of Engineering, Brown University, Providence, Rhode Island, USA
- Department of Orthopaedics, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
4
|
Grimm N, von Bischopinck M, Zumbusch A, Fuchs M. Long ranged stress correlations in the hard sphere liquid. J Chem Phys 2024; 161:144118. [PMID: 39399963 DOI: 10.1063/5.0225890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024] Open
Abstract
The smooth emergence of shear elasticity is a hallmark of the liquid to glass transition. In a liquid, viscous stresses arise from local structural rearrangements. In the solid, Eshelby has shown that stresses around an inclusion decay as a power law r-D, where D is the dimension of the system. We study glass-forming hard sphere fluids by simulation and observe the emergence of the unscreened power-law Eshelby pattern in the stress correlations of the isotropic liquid state. By a detailed tensorial analysis, we show that the fluctuating force field, viz., the divergence of the stress field, relaxes to zero with time in all states, while the shear stress correlations develop spatial power-law structures inside regions that grow with longitudinal and transverse sound propagation. We observe the predicted exponents r-D and r-D-2. In Brownian systems, shear stresses relax diffusively within these regions, with the diffusion coefficient determined by the shear modulus and the friction coefficient.
Collapse
Affiliation(s)
- Niklas Grimm
- Fachbereich Physik, Universität Konstanz, 78457 Konstanz, Germany
| | | | - Andreas Zumbusch
- Fachbereich Chemie, Universität Konstanz, 78457 Konstanz, Germany
| | - Matthias Fuchs
- Fachbereich Physik, Universität Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
5
|
Fang G, Ho BX, Xu H, Gong C, Qiao Z, Liao Y, Zhu S, Lu H, Nie N, Zhou T, Kim M, Huang C, Soh BS, Chen YC. Compressible Hollow Microlasers in Organoids for High-Throughput and Real-Time Mechanical Screening. ACS NANO 2024. [PMID: 39214618 DOI: 10.1021/acsnano.4c08886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Mechanical stress within organoids is a pivotal indicator in disease modeling and pharmacokinetics, yet current tools lack the ability to rapidly and dynamically screen these mechanics. Here, we introduce biocompatible and compressible hollow microlasers that realize all-optical assessment of cellular stress within organoids. The laser spectroscopy yields identification of cellular deformation at the nanometer scale, corresponding to tens of pascals stress sensitivity. The compressibility enables the investigation of the isotropic component, which is the fundamental mechanics of multicellular models. By integrating with a microwell array, we demonstrate the high-throughput screening of mechanical cues in tumoroids, establishing a platform for mechano-responsive drug screening. Furthermore, we showcase the monitoring and mapping of dynamic contractile stress within human embryonic stem cell-derived cardiac organoids, revealing the internal mechanical inhomogeneity within a single organoid. This method eliminates time-consuming scanning and sample damage, providing insights into organoid mechanobiology.
Collapse
Affiliation(s)
- Guocheng Fang
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Beatrice Xuan Ho
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Hongmei Xu
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Chaoyang Gong
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Zhen Qiao
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Yikai Liao
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Song Zhu
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Hongxu Lu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Biomaterials and Tissue Engineering Research Center, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, China
| | - Ningyuan Nie
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Tian Zhou
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Munho Kim
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Changjin Huang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Boon Seng Soh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117543, Singapore
| | - Yu-Cheng Chen
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
6
|
Jain S, Belkadi H, Michaut A, Sart S, Gros J, Genet M, Baroud CN. Using a micro-device with a deformable ceiling to probe stiffness heterogeneities within 3D cell aggregates. Biofabrication 2024; 16:035010. [PMID: 38447213 DOI: 10.1088/1758-5090/ad30c7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/06/2024] [Indexed: 03/08/2024]
Abstract
Recent advances in the field of mechanobiology have led to the development of methods to characterise single-cell or monolayer mechanical properties and link them to their functional behaviour. However, there remains a strong need to establish this link for three-dimensional (3D) multicellular aggregates, which better mimic tissue function. Here we present a platform to actuate and observe many such aggregates within one deformable micro-device. The platform consists of a single polydimethylsiloxane piece cast on a 3D-printed mould and bonded to a glass slide or coverslip. It consists of a chamber containing cell spheroids, which is adjacent to air cavities that are fluidically independent. Controlling the air pressure in these air cavities leads to a vertical displacement of the chamber's ceiling. The device can be used in static or dynamic modes over time scales of seconds to hours, with displacement amplitudes from a fewµm to several tens of microns. Further, we show how the compression protocols can be used to obtain measurements of stiffness heterogeneities within individual co-culture spheroids, by comparing image correlations of spheroids at different levels of compression with finite element simulations. The labelling of the cells and their cytoskeleton is combined with image correlation methods to relate the structure of the co-culture spheroid with its mechanical properties at different locations. The device is compatible with various microscopy techniques, including confocal microscopy, which can be used to observe the displacements and rearrangements of single cells and neighbourhoods within the aggregate. The complete experimental and imaging platform can now be used to provide multi-scale measurements that link single-cell behaviour with the global mechanical response of the aggregates.
Collapse
Affiliation(s)
- Shreyansh Jain
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, 25-28 Rue du Dr Roux, 75015 Paris, France
- Laboratoire d' Hydrodynamique (LadHyX), CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau, France
| | - Hiba Belkadi
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, 25-28 Rue du Dr Roux, 75015 Paris, France
- Laboratoire d' Hydrodynamique (LadHyX), CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau, France
| | - Arthur Michaut
- Institut Pasteur, Université Paris Cité, Dynamic Regulation of Morphogenesis, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Sébastien Sart
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, 25-28 Rue du Dr Roux, 75015 Paris, France
- Laboratoire d' Hydrodynamique (LadHyX), CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau, France
| | - Jérôme Gros
- Institut Pasteur, Université Paris Cité, Dynamic Regulation of Morphogenesis, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Martin Genet
- Laboratoire de Mécanique des Solides, CNRS, École Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau, France
- Inria, Palaiseau, France
| | - Charles N Baroud
- Institut Pasteur, Université Paris Cité, Physical Microfluidics and Bioengineering, 25-28 Rue du Dr Roux, 75015 Paris, France
- Laboratoire d' Hydrodynamique (LadHyX), CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau, France
| |
Collapse
|
7
|
Campàs O, Noordstra I, Yap AS. Adherens junctions as molecular regulators of emergent tissue mechanics. Nat Rev Mol Cell Biol 2024; 25:252-269. [PMID: 38093099 DOI: 10.1038/s41580-023-00688-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 03/28/2024]
Abstract
Tissue and organ development during embryogenesis relies on the collective and coordinated action of many cells. Recent studies have revealed that tissue material properties, including transitions between fluid and solid tissue states, are controlled in space and time to shape embryonic structures and regulate cell behaviours. Although the collective cellular flows that sculpt tissues are guided by tissue-level physical changes, these ultimately emerge from cellular-level and subcellular-level molecular mechanisms. Adherens junctions are key subcellular structures, built from clusters of classical cadherin receptors. They mediate physical interactions between cells and connect biochemical signalling to the physical characteristics of cell contacts, hence playing a fundamental role in tissue morphogenesis. In this Review, we take advantage of the results of recent, quantitative measurements of tissue mechanics to relate the molecular and cellular characteristics of adherens junctions, including adhesion strength, tension and dynamics, to the emergent physical state of embryonic tissues. We focus on systems in which cell-cell interactions are the primary contributor to morphogenesis, without significant contribution from cell-matrix interactions. We suggest that emergent tissue mechanics is an important direction for future research, bridging cell biology, developmental biology and mechanobiology to provide a holistic understanding of morphogenesis in health and disease.
Collapse
Affiliation(s)
- Otger Campàs
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Center for Systems Biology Dresden, Dresden, Germany.
| | - Ivar Noordstra
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Alpha S Yap
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia.
| |
Collapse
|
8
|
Zheng S, Banerji R, LeBourdais R, Zhang S, DuBois E, O’Shea T, Nia HT. Alteration of mechanical stresses in the murine brain by age and hemorrhagic stroke. PNAS NEXUS 2024; 3:pgae141. [PMID: 38659974 PMCID: PMC11042661 DOI: 10.1093/pnasnexus/pgae141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024]
Abstract
Residual mechanical stresses, also known as solid stresses, emerge during rapid differential growth or remodeling of tissues, as observed in morphogenesis and tumor growth. While residual stresses typically dissipate in most healthy adult organs, as the growth rate decreases, high residual stresses have been reported in mature, healthy brains. However, the origins and consequences of residual mechanical stresses in the brain across health, aging, and disease remain poorly understood. Here, we utilized and validated a previously developed method to map residual mechanical stresses in the brains of mice across three age groups: 5-7 days, 8-12 weeks, and 22 months. We found that residual solid stress rapidly increases from 5-7 days to 8-12 weeks and remains high in mature 22 months mice brains. Three-dimensional mapping revealed unevenly distributed residual stresses from the anterior to posterior coronal brain sections. Since the brain is rich in negatively charged hyaluronic acid, we evaluated the contribution of charged extracellular matrix (ECM) constituents in maintaining solid stress levels. We found that lower ionic strength leads to elevated solid stresses, consistent with its unshielding effect and the subsequent expansion of charged ECM components. Lastly, we demonstrated that hemorrhagic stroke, accompanied by loss of cellular density, resulted in decreased residual stress in the murine brain. Our findings contribute to a better understanding of spatiotemporal alterations of residual solid stresses in healthy and diseased brains, a crucial step toward uncovering the biological and immunological consequences of this understudied mechanical phenotype in the brain.
Collapse
Affiliation(s)
- Siyi Zheng
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Rohin Banerji
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Rob LeBourdais
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Eric DuBois
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Timothy O’Shea
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| |
Collapse
|
9
|
Krüger LJ, Vrugt MT, Bröker S, Wallmeyer B, Betz T, Wittkowski R. Analytical method for reconstructing the stress on a spherical particle from its surface deformation. Biophys J 2024; 123:527-537. [PMID: 38258291 PMCID: PMC10938078 DOI: 10.1016/j.bpj.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/10/2023] [Accepted: 01/17/2024] [Indexed: 01/24/2024] Open
Abstract
The mechanical forces that cells experience from the tissue surrounding them are crucial for their behavior and development. Experimental studies of such mechanical forces require a method for measuring them. A widely used approach in this context is bead deformation analysis, where spherical particles are embedded into the tissue. The deformation of the particles then allows to reconstruct the mechanical stress acting on them. Existing approaches for this reconstruction are either very time-consuming or not sufficiently general. In this article, we present an analytical approach to this problem based on an expansion in solid spherical harmonics that allows us to find the complete stress tensor describing the stress acting on the tissue. Our approach is based on the linear theory of elasticity and uses an ansatz specifically designed for deformed spherical bodies. We clarify the conditions under which this ansatz can be used, making our results useful also for other contexts in which this ansatz is employed. Our method can be applied to arbitrary radial particle deformations and requires a very low computational effort. The usefulness of the method is demonstrated by an application to experimental data.
Collapse
Affiliation(s)
- Lea Johanna Krüger
- Institute of Theoretical Physics, Center for Soft Nanoscience, University of Münster, Münster, Germany
| | - Michael Te Vrugt
- Institute of Theoretical Physics, Center for Soft Nanoscience, University of Münster, Münster, Germany; DAMTP, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK
| | - Stephan Bröker
- Institute of Theoretical Physics, Center for Soft Nanoscience, University of Münster, Münster, Germany
| | - Bernhard Wallmeyer
- Centre for Molecular Biology of Inflammation, Institute of Cell Biology, University of Münster, Münster, Germany
| | - Timo Betz
- Third Institute of Physics - Biophysics, University of Göttingen, Göttingen, Germany
| | - Raphael Wittkowski
- Institute of Theoretical Physics, Center for Soft Nanoscience, University of Münster, Münster, Germany.
| |
Collapse
|
10
|
Yeh YT, Del Álamo JC, Caffrey CR. Biomechanics of parasite migration within hosts. Trends Parasitol 2024; 40:164-175. [PMID: 38172015 DOI: 10.1016/j.pt.2023.12.001] [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: 09/26/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
The dissemination of protozoan and metazoan parasites through host tissues is hindered by cellular barriers, dense extracellular matrices, and fluid forces in the bloodstream. To overcome these diverse biophysical impediments, parasites implement versatile migratory strategies. Parasite-exerted mechanical forces and upregulation of the host's cellular contractile machinery are the motors for these strategies, and these are comparably better characterized for protozoa than for helminths. Using the examples of the protozoans, Toxoplasma gondii and Plasmodium, and the metazoan, Schistosoma mansoni, we highlight how quantitative tools such as traction force and reflection interference contrast microscopies have improved our understanding of how parasites alter host mechanobiology to promote their migration.
Collapse
Affiliation(s)
- Yi-Ting Yeh
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA.
| | - Juan C Del Álamo
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Division of Cardiology, University of Washington, Seattle, WA 98109, USA; Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 93093, USA
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, 9500 Gilman Drive, MC0657, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
11
|
Li M, Sun H, Hou Z, Hao S, Jin L, Wang B. Engineering the Physical Microenvironment into Neural Organoids for Neurogenesis and Neurodevelopment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306451. [PMID: 37771182 DOI: 10.1002/smll.202306451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/04/2023] [Indexed: 09/30/2023]
Abstract
Understanding the signals from the physical microenvironment is critical for deciphering the processes of neurogenesis and neurodevelopment. The discovery of how surrounding physical signals shape human developing neurons is hindered by the bottleneck of conventional cell culture and animal models. Notwithstanding neural organoids provide a promising platform for recapitulating human neurogenesis and neurodevelopment, building neuronal physical microenvironment that accurately mimics the native neurophysical features is largely ignored in current organoid technologies. Here, it is discussed how the physical microenvironment modulates critical events during the periods of neurogenesis and neurodevelopment, such as neural stem cell fates, neural tube closure, neuronal migration, axonal guidance, optic cup formation, and cortical folding. Although animal models are widely used to investigate the impacts of physical factors on neurodevelopment and neuropathy, the important roles of human stem cell-derived neural organoids in this field are particularly highlighted. Considering the great promise of human organoids, building neural organoid microenvironments with mechanical forces, electrophysiological microsystems, and light manipulation will help to fully understand the physical cues in neurodevelopmental processes. Neural organoids combined with cutting-edge techniques, such as advanced atomic force microscopes, microrobots, and structural color biomaterials might promote the development of neural organoid-based research and neuroscience.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Heng Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| | - Zongkun Hou
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
| |
Collapse
|
12
|
Abstract
Recent methodological advances in measurements of geometry and forces in the early embryo and its models are enabling a deeper understanding of the complex interplay of genetics, mechanics and geometry during development.
Collapse
Affiliation(s)
- Zong-Yuan Liu
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Vikas Trivedi
- EMBL Barcelona, Barcelona, Spain
- EMBL Heidelberg, Developmental Biology Unit, Heidelberg, Germany
| | - Idse Heemskerk
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA.
- Center for Cell Plasticity and Organ Design, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Physics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Vian A, Pochitaloff M, Yen ST, Kim S, Pollock J, Liu Y, Sletten EM, Campàs O. In situ quantification of osmotic pressure within living embryonic tissues. Nat Commun 2023; 14:7023. [PMID: 37919265 PMCID: PMC10622550 DOI: 10.1038/s41467-023-42024-9] [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: 02/08/2023] [Accepted: 09/27/2023] [Indexed: 11/04/2023] Open
Abstract
Mechanics is known to play a fundamental role in many cellular and developmental processes. Beyond active forces and material properties, osmotic pressure is believed to control essential cell and tissue characteristics. However, it remains very challenging to perform in situ and in vivo measurements of osmotic pressure. Here we introduce double emulsion droplet sensors that enable local measurements of osmotic pressure intra- and extra-cellularly within 3D multicellular systems, including living tissues. After generating and calibrating the sensors, we measure the osmotic pressure in blastomeres of early zebrafish embryos as well as in the interstitial fluid between the cells of the blastula by monitoring the size of droplets previously inserted in the embryo. Our results show a balance between intracellular and interstitial osmotic pressures, with values of approximately 0.7 MPa, but a large pressure imbalance between the inside and outside of the embryo. The ability to measure osmotic pressure in 3D multicellular systems, including developing embryos and organoids, will help improve our understanding of its role in fundamental biological processes.
Collapse
Affiliation(s)
- Antoine Vian
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Cluster of Excellence Physics of Life, TU Dresden, 01062, Dresden, Germany
| | - Marie Pochitaloff
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Cluster of Excellence Physics of Life, TU Dresden, 01062, Dresden, Germany
| | - Shuo-Ting Yen
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Cluster of Excellence Physics of Life, TU Dresden, 01062, Dresden, Germany
| | - Sangwoo Kim
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Jennifer Pollock
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Yucen Liu
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Ellen M Sletten
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Otger Campàs
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.
- Cluster of Excellence Physics of Life, TU Dresden, 01062, Dresden, Germany.
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Center for Systems Biology Dresden, 01307, Dresden, Germany.
| |
Collapse
|
14
|
Ramos AP, Szalapak A, Ferme LC, Modes CD. From cells to form: A roadmap to study shape emergence in vivo. Biophys J 2023; 122:3587-3599. [PMID: 37243338 PMCID: PMC10541488 DOI: 10.1016/j.bpj.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/25/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Organogenesis arises from the collective arrangement of cells into progressively 3D-shaped tissue. The acquisition of a correctly shaped organ is then the result of a complex interplay between molecular cues, responsible for differentiation and patterning, and the mechanical properties of the system, which generate the necessary forces that drive correct shape emergence. Nowadays, technological advances in the fields of microscopy, molecular biology, and computer science are making it possible to see and record such complex interactions in incredible, unforeseen detail within the global context of the developing embryo. A quantitative and interdisciplinary perspective of developmental biology becomes then necessary for a comprehensive understanding of morphogenesis. Here, we provide a roadmap to quantify the events that lead to morphogenesis from imaging to image analysis, quantification, and modeling, focusing on the discrete cellular and tissue shape changes, as well as their mechanical properties.
Collapse
Affiliation(s)
| | - Alicja Szalapak
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany
| | | | - Carl D Modes
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany; Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
| |
Collapse
|
15
|
Xu KL, Mauck RL, Burdick JA. Modeling development using hydrogels. Development 2023; 150:dev201527. [PMID: 37387575 PMCID: PMC10323241 DOI: 10.1242/dev.201527] [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] [Indexed: 07/01/2023]
Abstract
The development of multicellular complex organisms relies on coordinated signaling from the microenvironment, including both biochemical and mechanical interactions. To better understand developmental biology, increasingly sophisticated in vitro systems are needed to mimic these complex extracellular features. In this Primer, we explore how engineered hydrogels can serve as in vitro culture platforms to present such signals in a controlled manner and include examples of how they have been used to advance our understanding of developmental biology.
Collapse
Affiliation(s)
- Karen L. Xu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert L. Mauck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
16
|
Ryoo H, Underhill GH. Spatially Defined Cell-Secreted Protein Detection Using Granular Hydrogels: μGeLISA. ACS Biomater Sci Eng 2023; 9:2317-2328. [PMID: 37070831 PMCID: PMC11135160 DOI: 10.1021/acsbiomaterials.2c01308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Intercellular communication through secreted proteins is necessary in essential processes such as embryo and limb development, disease progression, and immune responses. There exist many techniques to study bulk solution protein concentrations, but there is a limited set of tools to study the concentrations of cell-secreted proteins in situ within diverse cell platforms while retaining spatial information. In this study, we have developed a microgel system that is able to quantitatively measure the cell-secreted protein concentration within defined three-dimensional culture configurations with single-cell spatial resolution, called μGeLISA (microgel-linked immunosorbent assay). This system, which is based on the surface modification of polyethylene glycol microgels, was able to detect interleukin 6 (IL-6) concentrations of 2.21-21.86 ng/mL. Microgels were also able to detect cell spheroid-secreted IL-6 and distinguish between low- and high-secreting single cells. The system was also adapted to measure the concentration of cell-secreted matrix metalloproteinase-2 (MMP-2). μGeLISA represents a highly versatile system with a straightforward fabrication process that can be adapted toward the detection of secreted proteins within a diverse range of cell culture configurations.
Collapse
Affiliation(s)
- Hyeon Ryoo
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Gregory H Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
17
|
Lee W, Boghdady CM, Lelarge V, Leask RL, McCaffrey L, Moraes C. Ultrasoft edge-labelled hydrogel sensors reveal internal tissue stress patterns in invasive engineered tumors. Biomaterials 2023; 296:122073. [PMID: 36905756 DOI: 10.1016/j.biomaterials.2023.122073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 02/06/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Measuring internal mechanical stresses within 3D tissues can provide important insights into drivers of morphogenesis and disease progression. Cell-sized hydrogel microspheres have recently emerged as a powerful technique to probe tissue mechanobiology, as they can be sufficiently soft as to deform within remodelling tissues, and optically imaged to measure internal stresses. However, measuring stresses at resolutions of ∼10 Pa requires ultrasoft, low-polymer content hydrogel formulations that are challenging to label with sufficiently fluorescent materials to support repeated measurements, particularly in optically dense tissues over 100 μm thick, as required in cancer tumor models. Here, we leverage thermodynamic partitioning of hydrogel components to create "edge-labelled" ultrasoft hydrogel microdroplets, in a single polymerization step. Bright and stable fluorescent nanoparticles preferentially polymerize at the hydrogel droplet interface, and can be used to repeatedly track sensor surfaces over long-term experiments, even when embedded deep in light-scattering tissues. We utilize these edge-labelled microspherical stress gauges (eMSGs) in inducible breast cancer tumor models of invasion, and demonstrate distinctive internal stress patterns that arise from cell-matrix interactions at different stages of breast cancer progression. Our studies demonstrate a long-term macroscale compaction of the tumor during matrix encapsulation, but only a short-term increase in local stress as non-invasive tumors rapidly make small internal reorganizations that reduce the mechanical stress to baseline levels. In contrast, once invasion programs are initiated, internal stress throughout the tumor is negligible. These findings suggest that internal tumor stresses may initially prime the cells to invade, but are lost once invasion occurs. Together, this work demonstrates that mapping internal mechanical stress in tumors may have utility in advancing cancer prognostic strategies, and that eMSGs can have broad utility in understanding dynamic mechanical processes of disease and development.
Collapse
Affiliation(s)
- Wontae Lee
- Department of Chemical Engineering, McGill University, Montréal H3A 0C5 QC, Canada
| | | | - Virginie Lelarge
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal H3A 1A3 QC, Canada
| | - Richard L Leask
- Department of Chemical Engineering, McGill University, Montréal H3A 0C5 QC, Canada; McGill University Health Centre, Montréal H4A 3J1 QC, Canada
| | - Luke McCaffrey
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal H3A 1A3 QC, Canada; Division of Experimental Medicine, McGill University, Montréal H4A 3J1 QC, Canada; Gerald Bronfman Department of Oncology, McGill University, Montréal H4A 3T2, QC, Canada
| | - Christopher Moraes
- Department of Chemical Engineering, McGill University, Montréal H3A 0C5 QC, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal H3A 1A3 QC, Canada; Division of Experimental Medicine, McGill University, Montréal H4A 3J1 QC, Canada; Department of Biological and Biomedical Engineering, McGill University, Montréal H3A 2B4 QC, Canada.
| |
Collapse
|
18
|
Mohagheghian E, Luo J, Yavitt FM, Wei F, Bhala P, Amar K, Rashid F, Wang Y, Liu X, Ji C, Chen J, Arnold DP, Liu Z, Anseth KS, Wang N. Quantifying stiffness and forces of tumor colonies and embryos using a magnetic microrobot. Sci Robot 2023; 8:eadc9800. [PMID: 36696474 PMCID: PMC10098875 DOI: 10.1126/scirobotics.adc9800] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 12/22/2022] [Indexed: 01/27/2023]
Abstract
Stiffness and forces are two fundamental quantities essential to living cells and tissues. However, it has been a challenge to quantify both 3D traction forces and stiffness (or modulus) using the same probe in vivo. Here, we describe an approach that overcomes this challenge by creating a magnetic microrobot probe with controllable functionality. Biocompatible ferromagnetic cobalt-platinum microcrosses were fabricated, and each microcross (about 30 micrometers) was trapped inside an arginine-glycine-apartic acid-conjugated stiff poly(ethylene glycol) (PEG) round microgel (about 50 micrometers) using a microfluidic device. The stiff magnetic microrobot was seeded inside a cell colony and acted as a stiffness probe by rigidly rotating in response to an oscillatory magnetic field. Then, brief episodes of ultraviolet light exposure were applied to dynamically photodegrade and soften the fluorescent nanoparticle-embedded PEG microgel, whose deformation and 3D traction forces were quantified. Using the microrobot probe, we show that malignant tumor-repopulating cell colonies altered their modulus but not traction forces in response to different 3D substrate elasticities. Stiffness and 3D traction forces were measured, and both normal and shear traction force oscillations were observed in zebrafish embryos from blastula to gastrula. Mouse embryos generated larger tensile and compressive traction force oscillations than shear traction force oscillations during blastocyst. The microrobot probe with controllable functionality via magnetic fields could potentially be useful for studying the mechanoregulation of cells, tissues, and embryos.
Collapse
Affiliation(s)
- Erfan Mohagheghian
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Junyu Luo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Laboratory for Cellular Biomechanics and Regenerative Medicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - F. Max Yavitt
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Fuxiang Wei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Laboratory for Cellular Biomechanics and Regenerative Medicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Parth Bhala
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kshitij Amar
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Fazlur Rashid
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yuzheng Wang
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Xingchen Liu
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenyang Ji
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Laboratory for Cellular Biomechanics and Regenerative Medicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Junwei Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Laboratory for Cellular Biomechanics and Regenerative Medicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - David P. Arnold
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Zhen Liu
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Ning Wang
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
19
|
Abstract
The miniaturization of robotic tools and probes enables the fundamental study of mechanical properties of cells and tissues.
Collapse
Affiliation(s)
- José A Plaza
- Instituto de Microelectrónica de Barcelona [IMB-CNM (CSIC)], Campus UAB, 08193, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
20
|
Morales IA, Boghdady CM, Campbell BE, Moraes C. Integrating mechanical sensor readouts into organ-on-a-chip platforms. Front Bioeng Biotechnol 2022; 10:1060895. [PMID: 36588933 PMCID: PMC9800895 DOI: 10.3389/fbioe.2022.1060895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Organs-on-a-chip have emerged as next-generation tissue engineered models to accurately capture realistic human tissue behaviour, thereby addressing many of the challenges associated with using animal models in research. Mechanical features of the culture environment have emerged as being critically important in designing organs-on-a-chip, as they play important roles in both stimulating realistic tissue formation and function, as well as capturing integrative elements of homeostasis, tissue function, and tissue degeneration in response to external insult and injury. Despite the demonstrated impact of incorporating mechanical cues in these models, strategies to measure these mechanical tissue features in microfluidically-compatible formats directly on-chip are relatively limited. In this review, we first describe general microfluidically-compatible Organs-on-a-chip sensing strategies, and categorize these advances based on the specific advantages of incorporating them on-chip. We then consider foundational and recent advances in mechanical analysis techniques spanning cellular to tissue length scales; and discuss their integration into Organs-on-a-chips for more effective drug screening, disease modeling, and characterization of biological dynamics.
Collapse
Affiliation(s)
| | | | | | - Christopher Moraes
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada,Department of Chemical Engineering, McGill University, Montreal, QC, Canada,Department of Biomedical Engineering, McGill University, Montreal, QC, Canada,*Correspondence: Christopher Moraes,
| |
Collapse
|
21
|
Biswas A, Ng BH, Prabhakaran VS, Chan CJ. Squeezing the eggs to grow: The mechanobiology of mammalian folliculogenesis. Front Cell Dev Biol 2022; 10:1038107. [PMID: 36531957 PMCID: PMC9756970 DOI: 10.3389/fcell.2022.1038107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/16/2022] [Indexed: 08/25/2023] Open
Abstract
The formation of functional eggs (oocyte) in ovarian follicles is arguably one of the most important events in early mammalian development since the oocytes provide the bulk genetic and cytoplasmic materials for successful reproduction. While past studies have identified many genes that are critical to normal ovarian development and function, recent studies have highlighted the role of mechanical force in shaping folliculogenesis. In this review, we discuss the underlying mechanobiological principles and the force-generating cellular structures and extracellular matrix that control the various stages of follicle development. We also highlight emerging techniques that allow for the quantification of mechanical interactions and follicular dynamics during development, and propose new directions for future studies in the field. We hope this review will provide a timely and useful framework for future understanding of mechano-signalling pathways in reproductive biology and diseases.
Collapse
Affiliation(s)
- Arikta Biswas
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Boon Heng Ng
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | | | - Chii Jou Chan
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
The continuous evolution of 2D cell-traction forces quantification technology. Innovation (N Y) 2022; 3:100313. [PMID: 36160940 PMCID: PMC9494227 DOI: 10.1016/j.xinn.2022.100313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 09/04/2022] [Indexed: 11/23/2022] Open
Abstract
Cells generate traction forces by interacting with the extracellular matrix (ECM) during migration, contraction, invasion, and spreading. Cell-traction forces (CTFs) are extremely small but have enormous biological effects. It has been discovered that CTFs serve a crucial role in regulating proliferation, differentiation, wound healing, morphogenesis, angiogenesis, inflammation, and tumor genesis by working together with biochemical signals to maintain a coherent framework for these processes. For the study of cell biology, it is essential to understand the possible effect of CTFs on the various cellular functions and the amount of traction forces that can be generated by cells in their various states. Currently, CTF quantification approaches are either confined to detecting numerous scattered places on the surface of cells or are severely limited in temporal and spatial resolution, both of which are critical for living cells. Obtaining a highly accurate and dynamic mapping of the force distribution across living cells in real time via a simple mathematical technique remains a significant difficulty. This perspective provides a brief overview of recent landmark advances in the measurement of two-dimensional (2D) CTFs, as well as unique ideas for future improvement.
Collapse
|
23
|
Zhang Y, Dong Q, An Q, Zhang C, Mohagheghian E, Niu B, Qi F, Wei F, Chen S, Chen X, Wang A, Cao X, Wang N, Chen J. Synthetic Retinoid Kills Drug-Resistant Cancer Stem Cells via Inducing RARγ-Translocation-Mediated Tension Reduction and Chromatin Decondensation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203173. [PMID: 36031407 PMCID: PMC9631059 DOI: 10.1002/advs.202203173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/16/2022] [Indexed: 05/11/2023]
Abstract
A recently developed synthetic retinoid abrogates proliferation and induces apoptosis of drug-resistant malignant-cancer-stem-cell-like cells. However, the underlying mechanisms of how the synthetic retinoid induces cancer-stem-cell-like cell tumor-repopulating cell (TRC) apoptosis are elusive. Here, it is shown that although the retinoid and conventional anticancer drugs cisplatin, all-trans retinoic acid, and tazarotene all inhibit cytoskeletal tension and decondense chromatin prior to inducing TRC apoptosis, half-maximal inhibitory concentration of the retinoid is 20-fold lower than those anticancer drugs. The synthetic retinoid induces retinoic acid receptor gamma (RARγ) translocation from the nucleus to the cytoplasm, leading to reduced RARγ binding to Cdc42 promoter and Cdc42 downregulation, which decreases filamentous-actin (F-actin) and inhibits cytoskeletal tension. Elevating F-actin or upregulating histone 3 lysine 9 trimethylation decreases retinoid-induced DNA damage and apoptosis of TRCs. The combinatorial treatment with a chromatin decondensation molecule and the retinoid inhibits tumor metastasis in mice more effectively than the synthetic retinoid alone. These findings suggest a strategy of lowering cell tension and decondensing chromatin to enhance DNA damage to abrogate metastasis of cancer-stem-cell-like cells with high efficacy.
Collapse
Affiliation(s)
- Yao Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of EducationLaboratory for Cellular Biomechanics and Regenerative MedicineDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Qi Dong
- Key Laboratory of Molecular Biophysics of the Ministry of EducationLaboratory for Cellular Biomechanics and Regenerative MedicineDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Quanlin An
- Institute of Clinical ScienceZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Chumei Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of EducationLaboratory for Cellular Biomechanics and Regenerative MedicineDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Erfan Mohagheghian
- Department of Mechanical Science and EngineeringThe Grainger College of EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Bing Niu
- School of Life SciencesShanghai University99 Shangda RoadShanghai200444China
| | - Feng Qi
- Institute of Clinical ScienceZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Fuxiang Wei
- Key Laboratory of Molecular Biophysics of the Ministry of EducationLaboratory for Cellular Biomechanics and Regenerative MedicineDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Sihan Chen
- Key Laboratory of Molecular Biophysics of the Ministry of EducationLaboratory for Cellular Biomechanics and Regenerative MedicineDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Xinman Chen
- Key Laboratory of Molecular Biophysics of the Ministry of EducationLaboratory for Cellular Biomechanics and Regenerative MedicineDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Anqi Wang
- Key Laboratory of Molecular Biophysics of the Ministry of EducationLaboratory for Cellular Biomechanics and Regenerative MedicineDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Xin Cao
- Institute of Clinical ScienceZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
| | - Ning Wang
- Department of Mechanical Science and EngineeringThe Grainger College of EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Junwei Chen
- Key Laboratory of Molecular Biophysics of the Ministry of EducationLaboratory for Cellular Biomechanics and Regenerative MedicineDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074China
| |
Collapse
|
24
|
Chowdhury F, Huang B, Wang N. Forces in stem cells and cancer stem cells. Cells Dev 2022; 170:203776. [DOI: 10.1016/j.cdev.2022.203776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/26/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
|
25
|
Guo H, Huang S, He B. Evidence for a Role of the Lateral Ectoderm in Drosophila Mesoderm Invagination. Front Cell Dev Biol 2022; 10:867438. [PMID: 35547820 PMCID: PMC9081377 DOI: 10.3389/fcell.2022.867438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/01/2022] [Indexed: 01/09/2023] Open
Abstract
The folding of two-dimensional epithelial sheets into specific three-dimensional structures is a fundamental tissue construction mechanism in animal development. A common mechanism that mediates epithelial folding is apical constriction, the active shrinking of cell apices driven by actomyosin contractions. It remains unclear whether cells outside of the constriction domain also contribute to folding. During Drosophila mesoderm invagination, ventrally localized mesoderm epithelium undergoes apical constriction and subsequently folds into a furrow. While the critical role of apical constriction in ventral furrow formation has been well demonstrated, it remains unclear whether, and if so, how the laterally localized ectodermal tissue adjacent to the mesoderm contributes to furrow invagination. In this study, we combine experimental and computational approaches to test the potential function of the ectoderm in mesoderm invagination. Through laser-mediated, targeted disruption of cell formation prior to gastrulation, we found that the presence of intact lateral ectoderm is important for the effective transition between apical constriction and furrow invagination in the mesoderm. In addition, using a laser-ablation approach widely used for probing tissue tension, we found that the lateral ectodermal tissues exhibit signatures of tissue compression when ablation was performed shortly before the onset of mesoderm invagination. These observations led to the hypothesis that in-plane compression from the surrounding ectoderm facilitates mesoderm invagination by triggering buckling of the mesoderm epithelium. In support of this notion, we show that the dynamics of tissue flow during mesoderm invagination displays characteristic of elastic buckling, and this tissue dynamics can be recapitulated by combining local apical constriction and global compression in a simulated elastic monolayer. We propose that Drosophila mesoderm invagination is achieved through epithelial buckling jointly mediated by apical constriction in the mesoderm and compression from the neighboring ectoderm.
Collapse
Affiliation(s)
| | | | - Bing He
- Department of Biological Sciences, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
26
|
Ding X, Li M, Cheng B, Wei Z, Dong Y, Xu F. Microsphere sensors for characterizing stress fields within three-dimensional extracellular matrix. Acta Biomater 2022; 141:1-13. [PMID: 34979325 DOI: 10.1016/j.actbio.2021.12.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022]
Abstract
Stress in the three-dimensional extracellular matrix is one of the key cues in regulating multiscale biological processes. Thus far, noticeable progress in methods and techniques (e.g., micropipette aspiration, AFM, and molecule probes) has been made to quantify stress in cell microenvironment at different length scales. Among them, the microsphere sensor-based method (MSS-based method) has emerged as an advantageous approach over conventional techniques in quantifying stress in situ and in vivo at cellular and supra-cellular scales. This method is implemented by seven sequential steps, including fabrication, modification, characterization, cell adhesion, imaging, displacement field extraction and stress calculation. Precise control of each step and inter-tunning between steps can provide quantitative characterization of stress field. However, detailed procedural information associated with each step and process has been scattered. This review aims to provide a comprehensive overview of MSS-based method, systematically summarizing the principles and research progresses. Firstly, the basic principles are introduced, and the specific experiment and calculation processes of MSS-based method are presented in detail. Then, recent advances and applications of this method are summarized. Finally, perspectives of the limitations and development trends of MSS-based method are discussed. This specific and comprehensive review would provide a guideline for the widespread application of MSS-based method as an advantageous method for in situ and in vivo stress characterization at cellular and supra-cellular scale within three-dimensional extracellular matrix. STATEMENT OF SIGNIFICANCE: In this review, a method based on a microsphere sensor (MSS-based method) as an advantageous approach over conventional techniques in quantifying stress in situ and in vivo at cellular and supra-cellular scales is introduced and discussed. This technique is implemented by seven sequential steps, including fabrication, modification, characterization, cell junction, imaging, displacement field extraction, and stress calculation. Precise control of each step and inter-tunning between steps can provide quantitative stress field. However, detailed procedural information associated with each step has been scattered. Thus, a comprehensive review collating recent advances and perspective discussions is a necessity to introduce a better option for quantifying the stress field in biological processes at the cellular and supra-cellular scales.
Collapse
Affiliation(s)
- Xin Ding
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Moxiao Li
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China
| | - Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Zhao Wei
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuqing Dong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
27
|
Souchaud A, Boutillon A, Charron G, Asnacios A, Noûs C, David NB, Graner F, Gallet F. Live 3D imaging and mapping of shear stresses within tissues using incompressible elastic beads. Development 2022; 149:274481. [DOI: 10.1242/dev.199765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 12/17/2021] [Indexed: 12/30/2022]
Abstract
ABSTRACT
To investigate the role of mechanical constraints in morphogenesis and development, we have developed a pipeline of techniques based on incompressible elastic sensors. These techniques combine the advantages of incompressible liquid droplets, which have been used as precise in situ shear stress sensors, and of elastic compressible beads, which are easier to tune and to use. Droplets of a polydimethylsiloxane mix, made fluorescent through specific covalent binding to a rhodamin dye, are produced by a microfluidics device. The elastomer rigidity after polymerization is adjusted to the tissue rigidity. Its mechanical properties are carefully calibrated in situ, for a sensor embedded in a cell aggregate submitted to uniaxial compression. The local shear stress tensor is retrieved from the sensor shape, accurately reconstructed through an active contour method. In vitro, within cell aggregates, and in vivo, in the prechordal plate of the zebrafish embryo during gastrulation, our pipeline of techniques demonstrates its efficiency to directly measure the three dimensional shear stress repartition within a tissue.
Collapse
Affiliation(s)
- Alexandre Souchaud
- Matière et Systèmes Complexes, UMR 7057 associée au CNRS et à l'Université de Paris, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - Arthur Boutillon
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, 91128 Palaiseau, France
| | - Gaëlle Charron
- Matière et Systèmes Complexes, UMR 7057 associée au CNRS et à l'Université de Paris, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - Atef Asnacios
- Matière et Systèmes Complexes, UMR 7057 associée au CNRS et à l'Université de Paris, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - Camille Noûs
- Laboratory Cogitamus, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - Nicolas B. David
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, 91128 Palaiseau, France
| | - François Graner
- Matière et Systèmes Complexes, UMR 7057 associée au CNRS et à l'Université de Paris, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - François Gallet
- Matière et Systèmes Complexes, UMR 7057 associée au CNRS et à l'Université de Paris, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| |
Collapse
|
28
|
Abstract
Biological systems display a rich phenomenology of states that resemble the physical states of matter - solid, liquid and gas. These phases result from the interactions between the microscopic constituent components - the cells - that manifest in macroscopic properties such as fluidity, rigidity and resistance to changes in shape and volume. Looked at from such a perspective, phase transitions from a rigid to a flowing state or vice versa define much of what happens in many biological processes especially during early development and diseases such as cancer. Additionally, collectively moving confluent cells can also lead to kinematic phase transitions in biological systems similar to multi-particle systems where the particles can interact and show sub-populations characterised by specific velocities. In this Perspective we discuss the similarities and limitations of the analogy between biological and inert physical systems both from theoretical perspective as well as experimental evidence in biological systems. In understanding such transitions, it is crucial to acknowledge that the macroscopic properties of biological materials and their modifications result from the complex interplay between the microscopic properties of cells including growth or death, neighbour interactions and secretion of matrix, phenomena unique to biological systems. Detecting phase transitions in vivo is technically difficult. We present emerging approaches that address this challenge and may guide our understanding of the organization and macroscopic behaviour of biological tissues.
Collapse
Affiliation(s)
- Pierre-François Lenne
- Aix Marseille Univ, CNRS, UMR 7288, IBDM, Turing Center for Living Systems, Marseille, France.
| | - Vikas Trivedi
- European Molecular Biology Laboratory (EMBL), Barcelona, 08003, Spain.
- EMBL Heidelberg, Developmental Biology Unit, Heidelberg, 69117, Germany.
| |
Collapse
|
29
|
Sun W, Gao X, Lei H, Wang W, Cao Y. Biophysical Approaches for Applying and Measuring Biological Forces. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105254. [PMID: 34923777 PMCID: PMC8844594 DOI: 10.1002/advs.202105254] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Indexed: 05/13/2023]
Abstract
Over the past decades, increasing evidence has indicated that mechanical loads can regulate the morphogenesis, proliferation, migration, and apoptosis of living cells. Investigations of how cells sense mechanical stimuli or the mechanotransduction mechanism is an active field of biomaterials and biophysics. Gaining a further understanding of mechanical regulation and depicting the mechanotransduction network inside cells require advanced experimental techniques and new theories. In this review, the fundamental principles of various experimental approaches that have been developed to characterize various types and magnitudes of forces experienced at the cellular and subcellular levels are summarized. The broad applications of these techniques are introduced with an emphasis on the difficulties in implementing these techniques in special biological systems. The advantages and disadvantages of each technique are discussed, which can guide readers to choose the most suitable technique for their questions. A perspective on future directions in this field is also provided. It is anticipated that technical advancement can be a driving force for the development of mechanobiology.
Collapse
Affiliation(s)
- Wenxu Sun
- School of SciencesNantong UniversityNantong226019P. R. China
| | - Xiang Gao
- Key Laboratory of Intelligent Optical Sensing and IntegrationNational Laboratory of Solid State Microstructureand Department of PhysicsCollaborative Innovation Center of Advanced MicrostructuresNanjing UniversityNanjing210023P. R. China
- Institute of Brain ScienceNanjing UniversityNanjing210023P. R. China
| | - Hai Lei
- Key Laboratory of Intelligent Optical Sensing and IntegrationNational Laboratory of Solid State Microstructureand Department of PhysicsCollaborative Innovation Center of Advanced MicrostructuresNanjing UniversityNanjing210023P. R. China
- Institute of Brain ScienceNanjing UniversityNanjing210023P. R. China
- Chemistry and Biomedicine Innovation CenterNanjing UniversityNanjing210023P. R. China
| | - Wei Wang
- Key Laboratory of Intelligent Optical Sensing and IntegrationNational Laboratory of Solid State Microstructureand Department of PhysicsCollaborative Innovation Center of Advanced MicrostructuresNanjing UniversityNanjing210023P. R. China
- Institute of Brain ScienceNanjing UniversityNanjing210023P. R. China
| | - Yi Cao
- Key Laboratory of Intelligent Optical Sensing and IntegrationNational Laboratory of Solid State Microstructureand Department of PhysicsCollaborative Innovation Center of Advanced MicrostructuresNanjing UniversityNanjing210023P. R. China
- Institute of Brain ScienceNanjing UniversityNanjing210023P. R. China
- MOE Key Laboratory of High Performance Polymer Materials and TechnologyDepartment of Polymer Science & EngineeringCollege of Chemistry & Chemical EngineeringNanjing UniversityNanjing210023P. R. China
- Chemistry and Biomedicine Innovation CenterNanjing UniversityNanjing210023P. R. China
| |
Collapse
|
30
|
Blumberg JW, Schwarz US. Comparison of direct and inverse methods for 2.5D traction force microscopy. PLoS One 2022; 17:e0262773. [PMID: 35051243 PMCID: PMC8775276 DOI: 10.1371/journal.pone.0262773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 01/04/2022] [Indexed: 11/18/2022] Open
Abstract
Essential cellular processes such as cell adhesion, migration and division strongly depend on mechanical forces. The standard method to measure cell forces is traction force microscopy (TFM) on soft elastic substrates with embedded marker beads. While in 2D TFM one only reconstructs tangential forces, in 2.5D TFM one also considers normal forces. Here we present a systematic comparison between two fundamentally different approaches to 2.5D TFM, which in particular require different methods to deal with noise in the displacement data. In the direct method, one calculates strain and stress tensors directly from the displacement data, which in principle requires a divergence correction. In the inverse method, one minimizes the difference between estimated and measured displacements, which requires some kind of regularization. By calculating the required Green's functions in Fourier space from Boussinesq-Cerruti potential functions, we first derive a new variant of 2.5D Fourier Transform Traction Cytometry (FTTC). To simulate realistic traction patterns, we make use of an analytical solution for Hertz-like adhesion patches. We find that FTTC works best if only tangential forces are reconstructed, that 2.5D FTTC is more precise for small noise, but that the performance of the direct method approaches the one of 2.5D FTTC for larger noise, before both fail for very large noise. Moreover we find that a divergence correction is not really needed for the direct method and that it profits more from increased resolution than the inverse method.
Collapse
Affiliation(s)
- Johannes W. Blumberg
- Heidelberg University, Institute for Theoretical Physics and Bioquant, Heidelberg, Germany
| | - Ulrich S. Schwarz
- Heidelberg University, Institute for Theoretical Physics and Bioquant, Heidelberg, Germany
| |
Collapse
|
31
|
Brewer G, Fortier AM, Park M, Moraes C. The case for cancer-associated fibroblasts: essential elements in cancer drug discovery? FUTURE DRUG DISCOVERY 2022; 4:FDD71. [PMID: 35600290 PMCID: PMC9112234 DOI: 10.4155/fdd-2021-0004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
Although cancer-associated fibroblasts (CAFs) have gained increased attention for supporting cancer progression, current CAF-targeted therapeutic options are limited and failing in clinical trials. As the largest component of the tumor microenvironment (TME), CAFs alter the biochemical and physical structure of the TME, modulating cancer progression. Here, we review the role of CAFs in altering drug response, modifying the TME mechanics and the current models for studying CAFs. To provide new perspectives, we highlight key considerations of CAF activity and discuss emerging technologies that can better address CAFs; and therefore, increase the likelihood of therapeutic efficacy. We argue that CAFs are crucial components of the cancer drug discovery pipeline and incorporating these cells will improve drug discovery success rates.
Collapse
Affiliation(s)
- Gabrielle Brewer
- Rosalind & Morris Goodman Cancer Research Centre, McGill University, 1160 Avenues des Pins, Montréal, QC, H3A 0G4, Canada
- Department of Biochemistry, McGill University, 3649 Promenade Sir-William-Osler, Montréal, QC, H3A 0G4, Canada
| | - Anne-Marie Fortier
- Rosalind & Morris Goodman Cancer Research Centre, McGill University, 1160 Avenues des Pins, Montréal, QC, H3A 0G4, Canada
| | - Morag Park
- Rosalind & Morris Goodman Cancer Research Centre, McGill University, 1160 Avenues des Pins, Montréal, QC, H3A 0G4, Canada
- Department of Biochemistry, McGill University, 3649 Promenade Sir-William-Osler, Montréal, QC, H3A 0G4, Canada
- Department of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC, H3A 0G4, Canada
- Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Montréal, QC, H3A 0G4, Canada
- Department of Pathology, McGill University, 3775 rue University, Montréal, QC, H3A 0G4, Canada
| | - Christopher Moraes
- Rosalind & Morris Goodman Cancer Research Centre, McGill University, 1160 Avenues des Pins, Montréal, QC, H3A 0G4, Canada
- Department of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC, H3A 0G4, Canada
- Department of Chemical Engineering, McGill University, 3610 rue University, Montréal, QC, H3A 0G4, Canada
- Department of Biomedical Engineering, McGill University, 3775 rue University, Montréal, QC, H3A 0G4, Canada
| |
Collapse
|
32
|
Veenvliet JV, Lenne PF, Turner DA, Nachman I, Trivedi V. Sculpting with stem cells: how models of embryo development take shape. Development 2021; 148:dev192914. [PMID: 34908102 PMCID: PMC8722391 DOI: 10.1242/dev.192914] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During embryogenesis, organisms acquire their shape given boundary conditions that impose geometrical, mechanical and biochemical constraints. A detailed integrative understanding how these morphogenetic information modules pattern and shape the mammalian embryo is still lacking, mostly owing to the inaccessibility of the embryo in vivo for direct observation and manipulation. These impediments are circumvented by the developmental engineering of embryo-like structures (stembryos) from pluripotent stem cells that are easy to access, track, manipulate and scale. Here, we explain how unlocking distinct levels of embryo-like architecture through controlled modulations of the cellular environment enables the identification of minimal sets of mechanical and biochemical inputs necessary to pattern and shape the mammalian embryo. We detail how this can be complemented with precise measurements and manipulations of tissue biochemistry, mechanics and geometry across spatial and temporal scales to provide insights into the mechanochemical feedback loops governing embryo morphogenesis. Finally, we discuss how, even in the absence of active manipulations, stembryos display intrinsic phenotypic variability that can be leveraged to define the constraints that ensure reproducible morphogenesis in vivo.
Collapse
Affiliation(s)
- Jesse V. Veenvliet
- Stembryogenesis Lab, Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307 Dresden, Germany
| | - Pierre-François Lenne
- Aix Marseille University, CNRS, IBDM, Turing Center for Living Systems, 13288, Marseille, France
| | - David A. Turner
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, University of Liverpool, Liverpool, L7 8TX, UK
| | - Iftach Nachman
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Vikas Trivedi
- European Molecular Biology Laboratories (EMBL), Barcelona, 08003, Spain
- EMBL Heidelberg, Developmental Biology Unit, 69117, Heidelberg, Germany
| |
Collapse
|
33
|
Boghdady CM, Kalashnikov N, Mok S, McCaffrey L, Moraes C. Revisiting tissue tensegrity: Biomaterial-based approaches to measure forces across length scales. APL Bioeng 2021; 5:041501. [PMID: 34632250 PMCID: PMC8487350 DOI: 10.1063/5.0046093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022] Open
Abstract
Cell-generated forces play a foundational role in tissue dynamics and homeostasis and are critically important in several biological processes, including cell migration, wound healing, morphogenesis, and cancer metastasis. Quantifying such forces in vivo is technically challenging and requires novel strategies that capture mechanical information across molecular, cellular, and tissue length scales, while allowing these studies to be performed in physiologically realistic biological models. Advanced biomaterials can be designed to non-destructively measure these stresses in vitro, and here, we review mechanical characterizations and force-sensing biomaterial-based technologies to provide insight into the mechanical nature of tissue processes. We specifically and uniquely focus on the use of these techniques to identify characteristics of cell and tissue "tensegrity:" the hierarchical and modular interplay between tension and compression that provide biological tissues with remarkable mechanical properties and behaviors. Based on these observed patterns, we highlight and discuss the emerging role of tensegrity at multiple length scales in tissue dynamics from homeostasis, to morphogenesis, to pathological dysfunction.
Collapse
Affiliation(s)
| | - Nikita Kalashnikov
- Department of Chemical Engineering, McGill University, Montréal, Québec H3A 0C5, Canada
| | - Stephanie Mok
- Department of Chemical Engineering, McGill University, Montréal, Québec H3A 0C5, Canada
| | | | | |
Collapse
|
34
|
Fang G, Lu H, Rodriguez de la Fuente L, Law AMK, Lin G, Jin D, Gallego‐Ortega D. Mammary Tumor Organoid Culture in Non-Adhesive Alginate for Luminal Mechanics and High-Throughput Drug Screening. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102418. [PMID: 34494727 PMCID: PMC8564453 DOI: 10.1002/advs.202102418] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/25/2021] [Indexed: 05/14/2023]
Abstract
Mammary tumor organoids have become a promising in vitro model for drug screening and personalized medicine. However, the dependency on the basement membrane extract (BME) as the growth matrices limits their comprehensive application. In this work, mouse mammary tumor organoids are established by encapsulating tumor pieces in non-adhesive alginate. High-throughput generation of organoids in alginate microbeads is achieved utilizing microfluidic droplet technology. Tumor pieces within the alginate microbeads developed both luminal- and solid-like structures and displayed a high similarity to the original fresh tumor in cellular phenotypes and lineages. The mechanical forces of the luminal organoids in the alginate capsules are analyzed with the theory of the thick-wall pressure vessel (TWPV) model. The luminal pressure of the organoids increase with the lumen growth and can reach 2 kPa after two weeks' culture. Finally, the mammary tumor organoids are treated with doxorubicin and latrunculin A to evaluate their application as a drug screening platform. It is found that the drug response is related to the luminal size and pressures of organoids. This high-throughput culture for mammary tumor organoids may present a promising tool for preclinical drug target validation and personalized medicine.
Collapse
Affiliation(s)
- Guocheng Fang
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
| | - Hongxu Lu
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
| | - Laura Rodriguez de la Fuente
- St. Vincent's Clinical SchoolFaculty of MedicineUniversity of New South Wales SydneyDarlinghurstNew South Wales2010Australia
- Garvan Institute of Medical Research384 Victoria StreetDarlinghurstNew South Wales2010Australia
| | - Andrew M. K. Law
- St. Vincent's Clinical SchoolFaculty of MedicineUniversity of New South Wales SydneyDarlinghurstNew South Wales2010Australia
- Garvan Institute of Medical Research384 Victoria StreetDarlinghurstNew South Wales2010Australia
| | - Gungun Lin
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
| | - Dayong Jin
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
- UTS‐SUSTech Joint Research Centre for Biomedical Materials and DevicesDepartment of Biomedical EngineeringSouthern University of Science and TechnologyShenzhenGuangdong518055China
| | - David Gallego‐Ortega
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
- St. Vincent's Clinical SchoolFaculty of MedicineUniversity of New South Wales SydneyDarlinghurstNew South Wales2010Australia
- Garvan Institute of Medical Research384 Victoria StreetDarlinghurstNew South Wales2010Australia
- School of Biomedical EngineeringFaculty of EngineeringUniversity of Technology SydneyBroadway UltimoSydneyNew South Wales2007Australia
| |
Collapse
|
35
|
Vorselen D, Barger SR, Wang Y, Cai W, Theriot JA, Gauthier NC, Krendel M. Phagocytic 'teeth' and myosin-II 'jaw' power target constriction during phagocytosis. eLife 2021; 10:e68627. [PMID: 34708690 PMCID: PMC8585483 DOI: 10.7554/elife.68627] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 10/27/2021] [Indexed: 12/16/2022] Open
Abstract
Phagocytosis requires rapid actin reorganization and spatially controlled force generation to ingest targets ranging from pathogens to apoptotic cells. How actomyosin activity directs membrane extensions to engulf such diverse targets remains unclear. Here, we combine lattice light-sheet microscopy (LLSM) with microparticle traction force microscopy (MP-TFM) to quantify actin dynamics and subcellular forces during macrophage phagocytosis. We show that spatially localized forces leading to target constriction are prominent during phagocytosis of antibody-opsonized targets. This constriction is largely driven by Arp2/3-mediated assembly of discrete actin protrusions containing myosin 1e and 1f ('teeth') that appear to be interconnected in a ring-like organization. Contractile myosin-II activity contributes to late-stage phagocytic force generation and progression, supporting a specific role in phagocytic cup closure. Observations of partial target eating attempts and sudden target release via a popping mechanism suggest that constriction may be critical for resolving complex in vivo target encounters. Overall, our findings present a phagocytic cup shaping mechanism that is distinct from cytoskeletal remodeling in 2D cell motility and may contribute to mechanosensing and phagocytic plasticity.
Collapse
Affiliation(s)
- Daan Vorselen
- Department of Biology and Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Sarah R Barger
- Department of Cell and Developmental Biology, State University of New York Upstate Medical UniversitySyracuseUnited States
| | - Yifan Wang
- Department of Mechanical Engineering, Stanford UniversityStanfordUnited States
| | - Wei Cai
- Department of Mechanical Engineering, Stanford UniversityStanfordUnited States
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| | | | - Mira Krendel
- Department of Cell and Developmental Biology, State University of New York Upstate Medical UniversitySyracuseUnited States
| |
Collapse
|
36
|
Bredov DV, Volodyaev IV, Luchinskaya NN. Spatio-Temporal Dynamics of Embryonic Tissue Deformations during Gastrulation in Xenopus laevis: Morphometric Analysis. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421050027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Mechanical mapping of mammalian follicle development using Brillouin microscopy. Commun Biol 2021; 4:1133. [PMID: 34580426 PMCID: PMC8476509 DOI: 10.1038/s42003-021-02662-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 09/08/2021] [Indexed: 02/08/2023] Open
Abstract
In early mammalian development, the maturation of follicles containing the immature oocytes is an important biological process as the functional oocytes provide the bulk genetic and cytoplasmic materials for successful reproduction. Despite recent work demonstrating the regulatory role of mechanical stress in oocyte growth, quantitative studies of ovarian mechanical properties remain lacking both in vivo and ex vivo. In this work, we quantify the material properties of ooplasm, follicles and connective tissues in intact mouse ovaries at distinct stages of follicle development using Brillouin microscopy, a non-invasive tool to probe mechanics in three-dimensional (3D) tissues. We find that the ovarian cortex and its interior stroma have distinct material properties associated with extracellular matrix deposition, and that intra-follicular mechanical compartments emerge during follicle maturation. Our work provides an alternative approach to study the role of mechanics in follicle morphogenesis and might pave the way for future understanding of mechanotransduction in reproductive biology, with potential implications for infertility diagnosis and treatment.
Collapse
|
38
|
Sensitive detection of cell-derived force and collagen matrix tension in microtissues undergoing large-scale densification. Proc Natl Acad Sci U S A 2021; 118:2106061118. [PMID: 34470821 DOI: 10.1073/pnas.2106061118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mechanical forces generated by cells and the tension of the extracellular matrix (ECM) play a decisive role in establishment, homeostasis maintenance, and repair of tissue morphology. However, the dynamic change of cell-derived force during large-scale remodeling of soft tissue is still unknown, mainly because the current techniques of force detection usually produce a nonnegligible and interfering feedback force on the cells during measurement. Here, we developed a method to fabricate highly stretchable polymer-based microstrings on which a microtissue of fibroblasts in collagen was cultured and allowed to contract to mimic the densification of soft tissue. Taking advantage of the low-spring constant and large deflection range of the microstrings, we detected a strain-induced contraction force as low as 5.2 µN without disturbing the irreversible densification. Meanwhile, the microtissues displayed extreme sensitivity to the mechanical boundary within a narrow range of tensile stress. More importantly, results indicated that the cell-derived force did not solely increase with increased ECM stiffness as previous studies suggested. Indeed, the cell-derived force and collagen tension exchanged dramatically in dominating the microtissue strain during the densification, and the proportion of cell-derived force decreased linearly as the microtissue densified, with stiffness increasing to ∼500 Pa. Thus, this study provides insights into the biomechanical cross-talk between the cells and ECM of extremely soft tissue during large-extent densification, which may be important to guide the construction of life-like tissue by applying appropriate mechanical boundary conditions.
Collapse
|
39
|
Hu Y, Ma VP, Ma R, Chen W, Duan Y, Glazier R, Petrich BG, Li R, Salaita K. DNA‐Based Microparticle Tension Sensors (μTS) for Measuring Cell Mechanics in Non‐planar Geometries and for High‐Throughput Quantification. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yuesong Hu
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | | | - Rong Ma
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | - Wenchun Chen
- Aflac Cancer and Blood Disorders Center Children's Healthcare of Atlanta Department of Pediatrics Emory University Atlanta GA 30322 USA
| | - Yuxin Duan
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | - Roxanne Glazier
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| | - Brian G. Petrich
- Aflac Cancer and Blood Disorders Center Children's Healthcare of Atlanta Department of Pediatrics Emory University Atlanta GA 30322 USA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center Children's Healthcare of Atlanta Department of Pediatrics Emory University Atlanta GA 30322 USA
| | - Khalid Salaita
- Department of Chemistry Emory University Atlanta GA 30322 USA
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| |
Collapse
|
40
|
He C, Wei X, Liang T, Liu M, Jiang D, Zhuang L, Wang P. Quantifying the Compressive Force of 3D Cardiac Tissues via Calculating the Volumetric Deformation of Built-In Elastic Gelatin Microspheres. Adv Healthc Mater 2021; 10:e2001716. [PMID: 34197053 DOI: 10.1002/adhm.202001716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/30/2020] [Indexed: 01/28/2023]
Abstract
Quantifying cardiac contractile force is of paramount important in studying mechanical heart failure and screening therapeutic drugs. However, most existing methods can only measure the in-plane component of twitch force of cardiomyocytes, such that mismatching the centripetal compressive stress of heart beating in physiology. Here, a non-destructive method is developed for quantifying the compressive stress and mapping the distribution of the local stress within the 3D cardiac tissues. In detail, elastic gelatin microspheres labeled with fluorescence beads are fabricated by microfluidic chips with high throughput, and they serve as built-in pressure sensors which are wrapped by cardiomyocytes in 3D tissues. The deformation of microspheres and the displacements of fluorescent beads induced by the contraction of cardiomyocytes are demonstrated to characterize the amount and distribution of the centripetal compressive stress. Further, the method shows a potent capability to locally quantify contractile force variation of 3D cardiac tissues, which is induced by agonist (norepinephrine) and inhibitor (blebbistatin). On the whole, the method significantly improves the 3D measurement of mechanical force in vitro and provides a solution for locally quantifying the compressive stress within engineered cardiac tissues.
Collapse
Affiliation(s)
- Chuanjiang He
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
- State Key Laboratory of Transducer Technology Chinese Academy of Sciences Shanghai 200050 China
| | - Xinwei Wei
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Tao Liang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Mengxue Liu
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Deming Jiang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Liujing Zhuang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Ping Wang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
- State Key Laboratory of Transducer Technology Chinese Academy of Sciences Shanghai 200050 China
| |
Collapse
|
41
|
Cuvelier M, Pešek J, Papantoniou I, Ramon H, Smeets B. Distribution and propagation of mechanical stress in simulated structurally heterogeneous tissue spheroids. SOFT MATTER 2021; 17:6603-6615. [PMID: 34142683 DOI: 10.1039/d0sm02033h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The mechanical microenvironment of cells has been associated with phenotypic changes that cells undergo in three-dimensional spheroid culture formats. Radial asymmetry in mechanical stress - with compression in the core and tension at the periphery - has been analyzed by representing tissue spheroids as homogeneous visco-elastic droplets under surface tension. However, the influence of the granular microstructure of tissue spheroids in the distribution of mechanical stress in tissue spheroids has not been accounted for in a generic manner. Here, we quantify the distribution and propagation of mechanical forces in structurally heterogeneous multicellular assemblies. For this, we perform numerical simulations of a deformable cell model, which represents cells as elastic, contractile shells surrounding a liquid incompressible cytoplasm, interacting by means of non-specific adhesion. Using this model, we show how cell-scale properties such as cortical stiffness, active tension and cell-cell adhesive tension influence the distribution of mechanical stress in simulated tissue spheroids. Next, we characterize the transition at the tissue-scale from a homogeneous liquid droplet to a heterogeneous packed granular assembly.
Collapse
Affiliation(s)
- Maxim Cuvelier
- MeBioS, KU Leuven, Heverlee, Belgium. and Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Jiří Pešek
- MeBioS, KU Leuven, Heverlee, Belgium. and Team MAMBA, Inria de Paris, Paris, France
| | - Ioannis Papantoniou
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium and Institute of Chemical Engineering Sciences (ICEHT), Foundation for Research and Technology - Hellas (FORTH), Patras, Greece and Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|
42
|
Hu Y, Ma VPY, Ma R, Chen W, Duan Y, Glazier R, Petrich BG, Li R, Salaita K. DNA-Based Microparticle Tension Sensors (μTS) for Measuring Cell Mechanics in Non-planar Geometries and for High-Throughput Quantification. Angew Chem Int Ed Engl 2021; 60:18044-18050. [PMID: 33979471 DOI: 10.1002/anie.202102206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/22/2021] [Indexed: 11/07/2022]
Abstract
Mechanotransduction, the interplay between physical and chemical signaling, plays vital roles in many biological processes. The state-of-the-art techniques to quantify cell forces employ deformable polymer films or molecular probes tethered to glass substrates. However, the applications of these assays in fundamental and clinical research are restricted by the planar geometry and low throughput of microscopy readout. Herein, we develop a DNA-based microparticle tension sensor, which features a spherical surface and thus allows for investigation of mechanotransduction at curved interfaces. The micron-scale of μTS enables flow cytometry readout, which is rapid and high throughput. We applied the method to map and measure T-cell receptor forces and platelet integrin forces at 12 and 56 pN thresholds. Furthermore, we quantified the inhibition efficiency of two anti-platelet drugs providing a proof-of-concept demonstration of μTS to screen drugs that modulate cellular mechanics.
Collapse
Affiliation(s)
- Yuesong Hu
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | | | - Rong Ma
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Wenchun Chen
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Yuxin Duan
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Roxanne Glazier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Brian G Petrich
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
43
|
Berg IC, Mohagheghian E, Habing K, Wang N, Underhill GH. Microtissue Geometry and Cell-Generated Forces Drive Patterning of Liver Progenitor Cell Differentiation in 3D. Adv Healthc Mater 2021; 10:e2100223. [PMID: 33890430 PMCID: PMC8222189 DOI: 10.1002/adhm.202100223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/27/2021] [Indexed: 01/13/2023]
Abstract
3D microenvironments provide a unique opportunity to investigate the impact of intrinsic mechanical signaling on progenitor cell differentiation. Using a hydrogel-based microwell platform, arrays of 3D, multicellular microtissues in constrained geometries, including toroids and cylinders are produced. These generated distinct mechanical profiles to investigate the impact of geometry and stress on early liver progenitor cell fate using a model liver development system. Image segmentation allows the tracking of individual cell fate and the characterization of distinct patterning of hepatocytic makers to the outer shell of the microtissues, and the exclusion from the inner diameter surface of the toroids. Biliary markers are distributed throughout the interior regions of micropatterned tissues and are increased in toroidal tissues when compared with those in cylindrical tissues. Finite element models of predicted stress distributions, combined with mechanical measurements, demonstrates that intercellular tension correlates with increased hepatocytic fate, while compression correlates with decreased hepatocytic and increased biliary fate. This system, which integrates microfabrication, imaging, mechanical modeling, and quantitative analysis, demonstrates how microtissue geometry can drive patterning of mechanical stresses that regulate cell differentiation trajectories. This approach may serve as a platform for further investigation of signaling mechanisms in the liver and other developmental systems.
Collapse
Affiliation(s)
- Ian C. Berg
- University of Illinois at Urbana-Champaign Department of Bioengineering, 1102 Everitt Lab, MC-278, 1406 W. Green Street, Urbana, IL 61801, USA
| | - Erfan Mohagheghian
- University of Illinois at Urbana-Champaign Department of Mechanical Science and Engineering, Mechanical Engineering Building, 1206 W. Green St. MC 244, Urbana, IL, 61801, USA
| | - Krista Habing
- University of Illinois at Urbana-Champaign Department of Bioengineering, 1102 Everitt Lab, MC-278, 1406 W. Green Street, Urbana, IL 61801, USA
| | - Ning Wang
- University of Illinois at Urbana-Champaign Department of Mechanical Science and Engineering, Mechanical Engineering Building, 1206 W. Green St. MC 244, Urbana, IL, 61801, USA
| | - Gregory H. Underhill
- University of Illinois at Urbana-Champaign Department of Bioengineering, 1102 Everitt Lab, MC-278, 1406 W. Green Street, Urbana, IL 61801, USA
| |
Collapse
|
44
|
Chowdhury F, Huang B, Wang N. Cytoskeletal prestress: The cellular hallmark in mechanobiology and mechanomedicine. Cytoskeleton (Hoboken) 2021; 78:249-276. [PMID: 33754478 PMCID: PMC8518377 DOI: 10.1002/cm.21658] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
Increasing evidence demonstrates that mechanical forces, in addition to soluble molecules, impact cell and tissue functions in physiology and diseases. How living cells integrate mechanical signals to perform appropriate biological functions is an area of intense investigation. Here, we review the evidence of the central role of cytoskeletal prestress in mechanotransduction and mechanobiology. Elevating cytoskeletal prestress increases cell stiffness and reinforces cell stiffening, facilitates long-range cytoplasmic mechanotransduction via integrins, enables direct chromatin stretching and rapid gene expression, spurs embryonic development and stem cell differentiation, and boosts immune cell activation and killing of tumor cells whereas lowering cytoskeletal prestress maintains embryonic stem cell pluripotency, promotes tumorigenesis and metastasis of stem cell-like malignant tumor-repopulating cells, and elevates drug delivery efficiency of soft-tumor-cell-derived microparticles. The overwhelming evidence suggests that the cytoskeletal prestress is the governing principle and the cellular hallmark in mechanobiology. The application of mechanobiology to medicine (mechanomedicine) is rapidly emerging and may help advance human health and improve diagnostics, treatment, and therapeutics of diseases.
Collapse
Affiliation(s)
- Farhan Chowdhury
- Department of Mechanical Engineering and Energy ProcessesSouthern Illinois University CarbondaleCarbondaleIllinoisUSA
| | - Bo Huang
- Department of Immunology, Institute of Basic Medical Sciences & State Key Laboratory of Medical Molecular BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ning Wang
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
45
|
Tuning the response of fluid filled hydrogel core-shell structures. J Mech Behav Biomed Mater 2021; 120:104605. [PMID: 34023588 DOI: 10.1016/j.jmbbm.2021.104605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 02/05/2023]
Abstract
Hydrogels are hydrophilic polymer networks that swell upon submersion in water. Thanks to their bio-compatibility, compliance, and ability to undergo large deformations, hydrogels can be used in a wide variety of applications such as in situ sensors for measuring cell-generated forces and drug delivery vehicles. In this work we investigate the equilibrium mechanical responses that can be achieved with hydrogel-based shells filled with a liquid core. Two types of gel shell geometries are considered - a cylinder and a spherical shell. Each shell is filled with either water or oil and subjected to compressive loading. We illustrate the influence of the shell geometry and the core composition on the mechanical response of the structure. We find that all core-shell structures stiffen under increasing compressive loading due to the load-induced expulsion of water molecules from the hydrogel shell. Furthermore, we show that cylindrical core-shell configurations are stiffer then their spherical equivalents. Interestingly, we demonstrate that the compression of a core-shell structure with an aqueous core leads to the transportation of water molecules from the core into the hydrogel. These results will guide the design of novel core-shell structures with tunable properties and mechanical responses.
Collapse
|
46
|
Fan Q, Zheng Y, Wang X, Xie R, Ding Y, Wang B, Yu X, Lu Y, Liu L, Li Y, Li M, Zhao Y, Jiao Y, Ye F. Dynamically Re‐Organized Collagen Fiber Bundles Transmit Mechanical Signals and Induce Strongly Correlated Cell Migration and Self‐Organization. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202016084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Qihui Fan
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
| | - Yu Zheng
- Department of Physics Arizona State University Tempe AZ 85287 USA
| | - Xiaochen Wang
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
- Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang 325001 China
- School of Physical Sciences University of Chinese Academy of Sciences Beijing 100049 China
| | - Ruipei Xie
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
- School of Physical Sciences University of Chinese Academy of Sciences Beijing 100049 China
| | - Yu Ding
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
- School of Physical Sciences University of Chinese Academy of Sciences Beijing 100049 China
| | - Boyi Wang
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
- School of Physical Sciences University of Chinese Academy of Sciences Beijing 100049 China
| | - Xiaoyu Yu
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
- School of Physical Sciences University of Chinese Academy of Sciences Beijing 100049 China
| | - Ying Lu
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
- School of Physical Sciences University of Chinese Academy of Sciences Beijing 100049 China
- Songshan Lake Materials Laboratory Dongguan Guangdong 523808 China
| | - Liyu Liu
- College of Physics Chongqing University Chongqing 401331 China
| | - Yunliang Li
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
| | - Ming Li
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
- School of Physical Sciences University of Chinese Academy of Sciences Beijing 100049 China
- Songshan Lake Materials Laboratory Dongguan Guangdong 523808 China
| | - Yuanjin Zhao
- Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang 325001 China
- Department of Rheumatology and Immunology The Affiliated Drum Tower Hospital of Nanjing University Medical School Nanjing 210008 China
| | - Yang Jiao
- Department of Physics Arizona State University Tempe AZ 85287 USA
- Materials Science and Engineering Arizona State University Tempe AZ 85287 USA
| | - Fangfu Ye
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics Institute of Physics Chinese Academy of Sciences Beijing 100190 China
- Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang 325001 China
- School of Physical Sciences University of Chinese Academy of Sciences Beijing 100049 China
- Songshan Lake Materials Laboratory Dongguan Guangdong 523808 China
| |
Collapse
|
47
|
Lee D, Greer SE, Kuss MA, An Y, Dudley AT. 3D printed alginate bead generator for high-throughput cell culture. Biomed Microdevices 2021; 23:22. [PMID: 33821331 DOI: 10.1007/s10544-021-00561-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2021] [Indexed: 01/02/2023]
Abstract
Alginate hydrogel beads are a common platform for generating 3D cell cultures in biomedical research. Simple methods for bead generation using a manual pipettor or syringe are low-throughput and produce beads showing high variability in size and shape. To address these challenges, we designed a 3D printed bead generator that uses an airflow to cleave beads from a stream of hydrogel solution. The performance of the proposed alginate bead generator was evaluated by changing the volume flow rates of alginate (QAlg) and air (QA), the diameter of device nozzle (d) and the concentration of alginate gel solution (C). We identified that the diameter of beads (D = 0.9 -2.8 mm) can be precisely controlled by changing QA and d. Also the bead generation frequency (f) can be tuned by changing QAlg. Finally, we demonstrated that viability and biological function (pericellular matrix deposition) of chondrocytes were not adversely affected by high f using this bead generator. Because 3D printing is becoming a more accessible technique, our unique design will allow greater access to average biomedical research laboratories, STEM education and industries in cost- and time-effective manner.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sydney E Greer
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mitchell A Kuss
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yang An
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Andrew T Dudley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
48
|
Schwartz AB, Campos OA, Criado-Hidalgo E, Chien S, del Álamo JC, Lasheras JC, Yeh YT. Elucidating the Biomechanics of Leukocyte Transendothelial Migration by Quantitative Imaging. Front Cell Dev Biol 2021; 9:635263. [PMID: 33855018 PMCID: PMC8039384 DOI: 10.3389/fcell.2021.635263] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/09/2021] [Indexed: 01/13/2023] Open
Abstract
Leukocyte transendothelial migration is crucial for innate immunity and inflammation. Upon tissue damage or infection, leukocytes exit blood vessels by adhering to and probing vascular endothelial cells (VECs), breaching endothelial cell-cell junctions, and transmigrating across the endothelium. Transendothelial migration is a critical rate-limiting step in this process. Thus, leukocytes must quickly identify the most efficient route through VEC monolayers to facilitate a prompt innate immune response. Biomechanics play a decisive role in transendothelial migration, which involves intimate physical contact and force transmission between the leukocytes and the VECs. While quantifying these forces is still challenging, recent advances in imaging, microfabrication, and computation now make it possible to study how cellular forces regulate VEC monolayer integrity, enable efficient pathfinding, and drive leukocyte transmigration. Here we review these recent advances, paying particular attention to leukocyte adhesion to the VEC monolayer, leukocyte probing of endothelial barrier gaps, and transmigration itself. To offer a practical perspective, we will discuss the current views on how biomechanics govern these processes and the force microscopy technologies that have enabled their quantitative analysis, thus contributing to an improved understanding of leukocyte migration in inflammatory diseases.
Collapse
Affiliation(s)
- Amy B. Schwartz
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Obed A. Campos
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Ernesto Criado-Hidalgo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States
| | - Juan C. Lasheras
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
49
|
Fan Q, Zheng Y, Wang X, Xie R, Ding Y, Wang B, Yu X, Lu Y, Liu L, Li Y, Li M, Zhao Y, Jiao Y, Ye F. Dynamically Re-Organized Collagen Fiber Bundles Transmit Mechanical Signals and Induce Strongly Correlated Cell Migration and Self-Organization. Angew Chem Int Ed Engl 2021; 60:11858-11867. [PMID: 33533087 DOI: 10.1002/anie.202016084] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/14/2021] [Indexed: 01/23/2023]
Abstract
Correlated cell migration in fibrous extracellular matrix (ECM) is important in many biological processes. During migration, cells can remodel the ECM, leading to the formation of mesoscale structures such as fiber bundles. However, how such mesoscale structures regulate correlated single-cells migration remains to be elucidated. Here, using a quasi-3D in vitro model, we investigate how collagen fiber bundles are dynamically re-organized and guide cell migration. By combining laser ablation technique with 3D tracking and active-particle simulations, we definitively show that only the re-organized fiber bundles that carry significant tensile forces can guide strongly correlated cell migration, providing for the first time a direct experimental evidence supporting that matrix-transmitted long-range forces can regulate cell migration and self-organization. This force regulation mechanism can provide new insights for studies on cellular dynamics, fabrication or selection of biomedical materials in tissue repairing, and many other biomedical applications.
Collapse
Affiliation(s)
- Qihui Fan
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yu Zheng
- Department of Physics, Arizona State University, Tempe, AZ, 85287, USA
| | - Xiaochen Wang
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.,Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China.,School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruipei Xie
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.,School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yu Ding
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.,School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Boyi Wang
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.,School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoyu Yu
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.,School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Lu
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.,School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.,Songshan Lake Materials Laboratory, Dongguan, Guangdong, 523808, China
| | - Liyu Liu
- College of Physics, Chongqing University, Chongqing, 401331, China
| | - Yunliang Li
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ming Li
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.,School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.,Songshan Lake Materials Laboratory, Dongguan, Guangdong, 523808, China
| | - Yuanjin Zhao
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China.,Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yang Jiao
- Department of Physics, Arizona State University, Tempe, AZ, 85287, USA.,Materials Science and Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Fangfu Ye
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter and Biological Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China.,Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China.,School of Physical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.,Songshan Lake Materials Laboratory, Dongguan, Guangdong, 523808, China
| |
Collapse
|
50
|
Roffay C, Chan CJ, Guirao B, Hiiragi T, Graner F. Inferring cell junction tension and pressure from cell geometry. Development 2021; 148:148/18/dev192773. [PMID: 33712442 DOI: 10.1242/dev.192773] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recognizing the crucial role of mechanical regulation and forces in tissue development and homeostasis has stirred a demand for in situ measurement of forces and stresses. Among emerging techniques, the use of cell geometry to infer cell junction tensions, cell pressures and tissue stress has gained popularity owing to the development of computational analyses. This approach is non-destructive and fast, and statistically validated based on comparisons with other techniques. However, its qualitative and quantitative limitations, in theory as well as in practice, should be examined with care. In this Primer, we summarize the underlying principles and assumptions behind stress inference, discuss its validity criteria and provide guidance to help beginners make the appropriate choice of its variants. We extend our discussion from two-dimensional stress inference to three dimensional, using the early mouse embryo as an example, and list a few possible extensions. We hope to make stress inference more accessible to the scientific community and trigger a broader interest in using this technique to study mechanics in development.
Collapse
Affiliation(s)
- Chloé Roffay
- Matière et Systèmes Complexes, Université de Paris - Diderot, CNRS UMR7057, 10 rue Alice Domon et Léonie Duquet, F-75205 Paris Cedex 13, France.,Polarity, Division and Morphogenesis Team, Genetics and Developmental Biology Unit, (CNRS UMR3215/Inserm U934), Institut Curie, F-75248 Paris Cedex 05, France
| | - Chii J Chan
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Boris Guirao
- Polarity, Division and Morphogenesis Team, Genetics and Developmental Biology Unit, (CNRS UMR3215/Inserm U934), Institut Curie, F-75248 Paris Cedex 05, France
| | - Takashi Hiiragi
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan
| | - François Graner
- Matière et Systèmes Complexes, Université de Paris - Diderot, CNRS UMR7057, 10 rue Alice Domon et Léonie Duquet, F-75205 Paris Cedex 13, France
| |
Collapse
|