1
|
Lin YJ, Tan XHM, Wang Y, Chung PS, Zhang X, Wu TH, Wu TY, Deb A, Chiou PY. Label-free optical mapping for large-area biomechanical dynamics of multicellular systems. Biosens Bioelectron 2025; 277:117281. [PMID: 40010021 DOI: 10.1016/j.bios.2025.117281] [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: 11/02/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Mapping cellular activities over large areas is crucial for understanding the collective behaviors of multicellular systems. Biomechanical properties, such as cellular traction forces, serve as critical regulators of physiological states and molecular configurations. However, existing technologies for mapping large-area biomechanical dynamics, which arise from changes in cellular traction forces, are limited by their small field of view and scanning-based nature. To address these limitations, we propose a novel platform that utilizes a vast number of optical diffractive elements to profile large-area biomechanical dynamics. This platform achieves a field of view of 10.6 mm × 10.6 mm, a three-order-of-magnitude improvement over traditional traction force microscopy. Transient mechanical waves generated by monolayer neonatal rat ventricular myocytes were captured with high spatiotemporal resolution (130 fps and 20 μm for temporal and spatial resolution, respectively). Furthermore, its label-free nature allows for long-term observations extended to a week, with minimal disruption to cellular functions. Finally, simultaneous measurements of calcium ion concentrations and biomechanical dynamics are demonstrated.
Collapse
Affiliation(s)
- Yen-Ju Lin
- Electrical and Computer Engineering Department, University of California, 420 Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Xing Haw Marvin Tan
- Mechanical and Aerospace Engineering Department, University of California, 420 Westwood Plaza, Los Angeles, CA, 90095, United States; Bioengineering Department, University of California, 420 Westwood Plaza, Los Angeles, CA, 90095, United States; Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A∗STAR), 1 Fusionopolis Way, #16-16 Connexis, Singapore, 138632, Republic of Singapore
| | - Yijie Wang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E Young Dr S, Los Angeles, CA, 90095, United States
| | - Pei-Shan Chung
- Bioengineering Department, University of California, 420 Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Xiang Zhang
- Mechanical and Aerospace Engineering Department, University of California, 420 Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Ting-Hsiang Wu
- MET Biotechnology LLC, 570 Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Tung-Yu Wu
- CyteSi LLC, 1600 Adams Drive, Suite 221, Menlo Park, CA, 94025, United States
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E Young Dr S, Los Angeles, CA, 90095, United States
| | - Pei-Yu Chiou
- Mechanical and Aerospace Engineering Department, University of California, 420 Westwood Plaza, Los Angeles, CA, 90095, United States; Bioengineering Department, University of California, 420 Westwood Plaza, Los Angeles, CA, 90095, United States.
| |
Collapse
|
2
|
Kim SH, Yasunaga AB, Zhang H, Whitley KD, Li ITS. Quantitative Super-Resolution Imaging of Molecular Tension. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2408280. [PMID: 40245301 DOI: 10.1002/advs.202408280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 02/08/2025] [Indexed: 04/19/2025]
Abstract
DNA-based molecular tension probes have revolutionized the localization of mechanical events in live cells with super-resolution. However, imaging the magnitude of these forces at super-resolution has been challenging. Here, qtPAINT (quantitative tension points accumulation for imaging in nanoscale topography) is introduced as a strategy to image the magnitude of molecular tension with super-resolution accuracy. By leveraging the force-dependent dissociation kinetics of short DNA oligonucleotides on their complementary strands, tension is encoded on individual molecules through their binding kinetics. This method allowed for a quantitative analysis of these kinetics, providing a detailed reconstruction of the force magnitudes acting on each tension probe. The technique integrates a molecular-beacon PAINT imager with a hairpin molecular tension probe, achieving a force quantification range of 9-30 pN and maintaining a spatial resolution of 30-120 nm in low and high-density regions. Additionally, qtPAINT offers a temporal resolution on the order of a minute, enhancing its applicability for studying dynamic cellular processes.
Collapse
Affiliation(s)
- Seong Ho Kim
- Department of Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
- Department of Chemistry and Advanced Materials, College of Natural Sciences, Gangneung-Wonju National University, Gangneung, 25457, Republic of Korea
| | - Adam B Yasunaga
- Department of Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Hongyuan Zhang
- Department of Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Kevin D Whitley
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Isaac T S Li
- Department of Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| |
Collapse
|
3
|
Qu KY, Cheng HY, Qiao L, Jiao JC, Chang SQ, Peng XF, Cui C, Zhang F, Huang NP. Construction of engineered cardiac tissue on a heart-on-a-chip device enables modeling of arrhythmogenic right ventricular cardiomyopathy. Biosens Bioelectron 2025; 281:117478. [PMID: 40245609 DOI: 10.1016/j.bios.2025.117478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/21/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a progressive cardiac disorder characterized by the replacement of the right ventricular myocardium with fibrofatty tissue, with an incidence rate of approximately 1 in 5000. To advance our understanding of its pathology and facilitate drug screening, there is an urgent need for myocardial models that closely replicate human physiological conditions. In this study, we developed an engineered cardiac tissue (ECT) model on a chip using cardiomyocytes differentiated from induced pluripotent stem cells (iPSCs) derived from ARVC patients. The disease ECT model successfully recapitulated key phenotypic features of ARVC, including reduced contractility, arrhythmic events, and abnormal calcium transients. We further assessed the drug responses of the model to isoproterenol and amiodarone, confirming increased sensitivity to isoproterenol in the ARVC model, while amiodarone effectively alleviated the arrhythmic events. In conclusion, our ECT model successfully reproduced ARVC phenotypes, providing a novel platform for drug screening and pathological studies.
Collapse
Affiliation(s)
- Kai-Yun Qu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Hong-Yi Cheng
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Li Qiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jin-Cheng Jiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Shi-Qi Chang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xia-Feng Peng
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Chang Cui
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China.
| | - Feng Zhang
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China.
| | - Ning-Ping Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
4
|
Pastor-Escuredo D, Lombardot B, Savy T, Boyreau A, Doursat R, Goicolea JM, Santos A, Bourgine P, del Álamo JC, Ledesma- Carbayo MJ, Peyriéras N. Unsupervised spatiotemporal classification of deformation patterns of embryonic tissues matches their fate map. iScience 2025; 28:111753. [PMID: 40124490 PMCID: PMC11926744 DOI: 10.1016/j.isci.2025.111753] [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: 09/28/2021] [Revised: 04/30/2022] [Accepted: 01/03/2025] [Indexed: 03/25/2025] Open
Abstract
During morphogenesis, embryonic tissues display fluid-like behavior with fluctuating strain rates. Digital cell lineages reconstructed from 4D images of developing zebrafish embryos are used to infer representative tissue deformation patterns and their association with developmental events. Finite deformation analysis along cell trajectories and unsupervised machine learning are applied to obtain reduced-order models condensing the collective cell motions, delineating tissue domains with distinct 4D biomechanical behavior. This reduced-order kinematic description is reproducible across specimens and matches fate maps of the zebrafish brain in wild-type and nodal pathway mutants (zoeptz57/tz57 ), shedding light into the morphogenetic defects causing these mutants' cyclopia. Furthermore, the inferred kinematic maps also match expression maps of the gene transcription factor goosecoid (gsc). In summary, this work introduces an objective analytical framework to systematically unravel the complex spatiotemporal patterns of embryonic tissue deformations and couple them with cell fate and gene expression maps.
Collapse
Affiliation(s)
- David Pastor-Escuredo
- USR3695/FRE2039 BioEmergences, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
- Biomedical Image Technologies, ETSIT, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Benoît Lombardot
- UPS3611 Complex Systems Institute Paris Ile-de-France (ISC-PIF), CNRS, Paris, France
| | - Thierry Savy
- USR3695/FRE2039 BioEmergences, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
- UPS3611 Complex Systems Institute Paris Ile-de-France (ISC-PIF), CNRS, Paris, France
- Matières et Systèmes Complexes (MSC) UMR7057, CNRS, Université Paris Cité, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - Adeline Boyreau
- USR3695/FRE2039 BioEmergences, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
| | - René Doursat
- USR3695/FRE2039 BioEmergences, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
- UPS3611 Complex Systems Institute Paris Ile-de-France (ISC-PIF), CNRS, Paris, France
| | - Jose M. Goicolea
- Computational Mechanics Group, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Andrés Santos
- Biomedical Image Technologies, ETSIT, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Paul Bourgine
- USR3695/FRE2039 BioEmergences, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
- UPS3611 Complex Systems Institute Paris Ile-de-France (ISC-PIF), CNRS, Paris, France
- Matières et Systèmes Complexes (MSC) UMR7057, CNRS, Université Paris Cité, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - Juan C. del Álamo
- Mechanical and Aerospace Engineering Department, University of California San Diego, La Jolla, CA 92093, USA
- Mechanical Engineering Department, University of Washington, Seattle, WA 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
| | - María J. Ledesma- Carbayo
- Biomedical Image Technologies, ETSIT, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Nadine Peyriéras
- USR3695/FRE2039 BioEmergences, CNRS, Paris-Saclay University, Gif-sur-Yvette, France
- UPS3611 Complex Systems Institute Paris Ile-de-France (ISC-PIF), CNRS, Paris, France
- Matières et Systèmes Complexes (MSC) UMR7057, CNRS, Université Paris Cité, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| |
Collapse
|
5
|
Lou Z, Zhang Y, Liang X, Cao M, Ma Y, Chen PR, Fan X. Deep-Red and Ultrafast Photocatalytic Proximity Labeling Empowered In Situ Dissection of Tumor-Immune Interactions in Primary Tissues. J Am Chem Soc 2025; 147:9716-9726. [PMID: 40036744 DOI: 10.1021/jacs.4c17879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Immunotherapy efficacy in solid tumors varies greatly, influenced by the tumor microenvironment (TME) and the dynamic tumor-immune interactions within it. Decoding these interactions in situ with minimal interference with native tissue architecture and delicate immune responses is critical for understanding tumor progression and optimizing therapeutic strategies. Here, we introduce CAT-Tissue, a novel deep-red photocatalytic proximity labeling method that enables ultrafast, high-resolution profiling of tumor-immune interactions in primary tissues. By leveraging nanobody-Chlorin e6 as the photocatalyst and biotin-aniline as the probe, CAT-Tissue enabled the rapid and comprehensive detection of various tumor-immune interactions in both coculture systems and primary tumor sections. Coupled with bulk RNA-sequencing, CAT-Tissue revealed distinct gene expression patterns between tumor-neighboring and tumor-distal lymphocytes, highlighting the recognition and immune responses of tumor-neighboring CD8+ T cells, which exhibited activated, effector, and exhausted phenotypes. By leveraging a deep-red photocatalytic proximity cell labeling strategy with excellent tissue penetration and biocompatibility, CAT-Tissue offers a nongenetically encoded platform with high sensitivity and spatiotemporal controllability for rapid profiling tumor-immune interactions within complex tissue environments in situ, which may advance our understanding of tumor immunology and guide the development of more effective immunotherapies.
Collapse
Affiliation(s)
- Zhizheng Lou
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yan Zhang
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xuan Liang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Mengrui Cao
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yicong Ma
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Peng R Chen
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xinyuan Fan
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
6
|
Lyall M, Kamdar A, Sykes R, Aekbote BL, Gadegaard N, Berry C. Measuring contractile forces in vascular smooth muscle cells. Vascul Pharmacol 2025; 159:107488. [PMID: 40097082 DOI: 10.1016/j.vph.2025.107488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/06/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
Vascular smooth muscle cell (VSMC) contractility mediates blood vessel tone. Abnormalities in VSMC function and in blood vessel tone can contribute to a variety of cardiovascular diseases. This review examines the role of VSMC contractile force in vascular disease, divided into two primary sections. The first section introducing VSMC mechanical contraction and detailing the molecular mechanisms of VSMC contractility in normal and pathological states. The second section exploring methods of measuring contraction in VSMCs, such as Ca2+ imaging, myography, and traction force microscopy, and highlighting where each method is of best use. Understanding the mechanical properties and contractile profiles of VSMCs offers valuable insights into disease mechanisms. By investigating these aspects, this review describes the potential of VSMC contractile forces as diagnostic markers and therapeutic targets in vascular disease.
Collapse
Affiliation(s)
- Maia Lyall
- School of Cardiovascular and Metabolic Health, University of Glasgow, G12 8TA, UK; Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Anna Kamdar
- School of Cardiovascular and Metabolic Health, University of Glasgow, G12 8TA, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Robert Sykes
- School of Cardiovascular and Metabolic Health, University of Glasgow, G12 8TA, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Badri L Aekbote
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Nikolaj Gadegaard
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, G12 8TA, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK; Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK.
| |
Collapse
|
7
|
Aw WY, Sawhney A, Rathod M, Whitworth CP, Doherty EL, Madden E, Lu J, Westphal K, Stack R, Polacheck WJ. Dysfunctional mechanotransduction regulates the progression of PIK3CA-driven vascular malformations. APL Bioeng 2025; 9:016106. [PMID: 39935869 PMCID: PMC11811908 DOI: 10.1063/5.0234507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/19/2025] [Indexed: 02/13/2025] Open
Abstract
Somatic activating mutations in PIK3CA are common drivers of vascular and lymphatic malformations. Despite common biophysical signatures of tissues susceptible to lesion formation, including compliant extracellular matrix and low rates of perfusion, lesions vary in clinical presentation from localized cystic dilatation to diffuse and infiltrative vascular dysplasia. The mechanisms driving the differences in disease severity and variability in clinical presentation and the role of the biophysical microenvironment in potentiating progression are poorly understood. Here, we investigate the role of hemodynamic forces and the biophysical microenvironment in the pathophysiology of vascular malformations (VMs), and we identify hemodynamic shear stress and defective endothelial cell mechanotransduction as key regulators of lesion progression. We found that constitutive PI3K activation impaired flow-mediated endothelial cell alignment and barrier function. We show that defective shear stress sensing in PIK3CAE542K endothelial cells is associated with reduced myosin light chain phosphorylation, junctional instability, and defective recruitment of vinculin to cell-cell junctions. Using three dimensional (3D) microfluidic models of the vasculature, we demonstrate that PIK3CAE542K microvessels apply reduced traction forces and are unaffected by flow interruption. We further found that draining transmural flow resulted in increased sprouting and invasion responses in PIK3CAE542K microvessels. Mechanistically, constitutive PI3K activation decreased cellular and nuclear elasticity resulting in defective cellular tensional homeostasis in endothelial cells which may underlie vascular dilation, tissue hyperplasia, and hypersprouting in PIK3CA-driven venous and lymphatic malformations. Together, these results suggest that defective nuclear mechanics, impaired cellular mechanotransduction, and maladaptive hemodynamic responses contribute to the development and progression of PIK3CA-driven vascular malformations.
Collapse
Affiliation(s)
- Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Aanya Sawhney
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Mitesh Rathod
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | | | - Elizabeth L. Doherty
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Ethan Madden
- Department of Genetics and Molecular Biology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Jingming Lu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Kaden Westphal
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Ryan Stack
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | | |
Collapse
|
8
|
Moradi Kashkooli F, Mirala F, H H Tehrani M, Alirahimi M, Souri M, Golzaryan A, Kar S, Soltani M. Mechanical Forces in Tumor Growth and Treatment: Perspectives From Biology, Physics, Engineering, and Mathematical Modeling. WIREs Mech Dis 2025; 17:e70000. [PMID: 40170456 DOI: 10.1002/wsbm.70000] [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: 07/21/2023] [Revised: 12/14/2024] [Accepted: 01/23/2025] [Indexed: 04/03/2025]
Abstract
The progression of tumors is influenced by mechanical forces and biological elements, such as hypoxia and angiogenesis. Mechanical factors, including stress, pressure, interstitial fluid pressure, and cellular traction forces, compromise normal tissue architecture, augmenting stiffness and thus promoting tumor growth and invasion. The selective elimination of specific tumor components can reduce growth-induced mechanical stress, thereby improving therapeutic efficacy. Furthermore, stress-relief drugs have the potential in enhancing chemotherapy outcomes. In this setting, computational modeling functions as an essential tool for quantitatively elucidating the mechanical principles underlying tumor formation. These models can precisely replicate the impact of mechanical pressures on solid tumors, offering insight into the regulation of tumor behavior by these forces. Tumor growth produces mechanical forces, including compression, displacement, and deformation, leading to irregular stress patterns, expedited tumor advancement, and reduced treatment efficacy. This review analyzes the impact of mechanical forces on carcinogenesis and solid tumor proliferation, emphasizing the significance of stress alleviation in regulating tumor growth. Furthermore, we investigate the influence of mechanical forces on tumor dissemination and emphasize the promise of integrating computational modeling with force-targeted cancer therapies to improve treatment efficacy by tackling the fundamental mechanics of tumor proliferation.
Collapse
Affiliation(s)
| | - Fatemeh Mirala
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Masoud H H Tehrani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mahvash Alirahimi
- Department of Obstetrics & Gynecology, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Aryan Golzaryan
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Egaila, Kuwait
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
9
|
Dalir L, Tatic-Lucic S, Berdichevsky Y. Cell-generated mechanical forces play a role in epileptogenesis after injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.09.637325. [PMID: 39990400 PMCID: PMC11844397 DOI: 10.1101/2025.02.09.637325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Traumatic brain injury (TBI) is associated with a significantly increased risk of epilepsy. One of the consequences of severe TBI is progressive brain atrophy, which is frequently characterized by organized tissue retraction. Retraction is an active process synchronized by mechanical interactions between surviving cells. This results in unbalanced mechanical forces acting on surviving neurons, potentially activating mechanotransduction and leading to hyperexcitability. This novel mechanism of epileptogenesis was examined in organotypic hippocampal cultures, which develop spontaneous seizure-like activity in vitro. Cell-generated forces in this model resulted in contraction of hippocampal tissue. Artificial imbalances in mechanical forces were introduced by placing cultured slices on surfaces with adhesive and non-adhesive regions. This modeled disbalance in mechanical forces that may occur in the brain after trauma. Portions of the slices that were not stabilized by substrate adhesion underwent increased contraction and compaction, revealing the presence of cell-generated forces capable of shaping tissue geometry. Changes in tissue geometry were followed by excitability changes that were specific to hippocampal sub-region and orientation of contractile forces relative to pyramidal cell apical-basal axis. Results of this study suggest that imbalanced cell-generated forces contribute to development of epilepsy, and that force imbalance may represent a novel mechanism of epileptogenesis after trauma.
Collapse
Affiliation(s)
- Laya Dalir
- Department of Bioengineering, Lehigh University, Bethlehem, PA
| | - Svetlana Tatic-Lucic
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA
| | - Yevgeny Berdichevsky
- Department of Bioengineering, Lehigh University, Bethlehem, PA
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA
| |
Collapse
|
10
|
Pu J, Ma J, Zhai H, Wu S, Wang Y, Putnis CV, Wang L, Zhang W. Atomic force microscopy imaging of plant cell walls. PLANT PHYSIOLOGY 2025; 197:kiae655. [PMID: 39928583 DOI: 10.1093/plphys/kiae655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/18/2024] [Indexed: 02/12/2025]
Abstract
Plant cell walls are highly dynamic, complex structures composed of multiple biopolymers that form a scaffold surrounding the plant cell. A nanoscale understanding of their architecture, mechanical properties, and formation/degradation dynamics is crucial for revealing structure-function relationships, mechanisms of shape formation, and cell development. Although imaging techniques have been extensively used in recent decades to reveal the structural organization and chemical compositions of cell walls, observing the detailed native architecture and identifying the physicochemical properties of plant cell walls remains challenging. Atomic force microscopy (AFM) is a powerful tool for simultaneously characterizing the morphology, nanomechanical properties, single-molecule interactions, and surface potentials of living biological systems. However, studies employing AFM to investigate plant cell walls have been relatively scarce. In this review, we discuss the latest advancements in AFM for in situ imaging of the multidimensional structure of the cell wall, measuring the mechanical properties of plant tissues or single cells, specific single-molecule recognition of cell wall-related enzymes-polysaccharides, and detecting the Kelvin potential of plant cell walls. We emphasize the fundamental challenges of AFM in characterizing plant cell walls and review potential applications for state-of-the-art AFM-based infrared/Raman spectroscopy toward answering open questions in plant biology.
Collapse
Affiliation(s)
- Junbao Pu
- College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
- State Key Laboratory of Plant Diversity and Specialty Crops, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| | - Jie Ma
- College of Environmental Science and Engineering, Guilin University of Technology, Guilin 541006, China
| | - Hang Zhai
- College of Resources and Environment, Interdisciplinary Research Center for Agriculture Green Development in Yangtze River Basin, Southwest University, Chongqing 400716, China
| | - Shanshan Wu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Youmei Wang
- Houji Laboratory in Shanxi Province, College of Agriculture, Shanxi Agricultural University, Taiyuan 030031, China
| | - Christine V Putnis
- Institut für Mineralogie, University of Münster, Münster 48149, Germany
- School of Molecular and Life Sciences, Curtin University, Perth 6845, Australia
| | - Lijun Wang
- College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| | - Wenjun Zhang
- College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
11
|
Ren Y, Yang J, Saito T, Glomb O, Mousavi SI, Naughton B, de Fontnouvelle C, Fujita B, Schlieker C, Yogev S, Zhang Y, Berro J. Genetically encoded mechano-sensors with versatile readouts and compact size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633409. [PMID: 39868242 PMCID: PMC11760715 DOI: 10.1101/2025.01.16.633409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Mechanical forces are critical for virtually all fundamental biological processes, yet quantification of mechanical forces at the molecular scale in vivo remains challenging. Here, we present a new strategy using calibrated coiled-coils as genetically encoded, compact, tunable, and modular mechano-sensors to substantially simplify force measurement in vivo, via diverse readouts (luminescence, fluorescence and analytical biochemistry) and instrumentation readily available in biology labs. We demonstrate the broad applicability and ease-of-use of these coiled-coil mechano-sensors by measuring forces during cytokinesis (formin Cdc12) and endocytosis (epsin Ent1) in yeast, force distributions in nematode axons (β-spectrin UNC-70), and forces transmitted to the nucleus (mini-nesprin-2G) and within focal adhesions (vinculin) in mammalian cells. We report discoveries in intracellular force transmission that have been elusive to existing tools.
Collapse
Affiliation(s)
- Yuan Ren
- Department of Molecular Biophysics and Biochemistry; Yale University, New Haven, CT, USA
- Nanobiology Institute, Yale University; West Haven, CT 06516, USA
| | - Jie Yang
- Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06520, USA
| | - Takumi Saito
- Department of Molecular Biophysics and Biochemistry; Yale University, New Haven, CT, USA
- Nanobiology Institute, Yale University; West Haven, CT 06516, USA
- Graduate School of Biomedical Engineering, Tohoku University, Sendai 980-8579, Japan
| | - Oliver Glomb
- Institut für Klinische Anatomie und Zellanalytik, Medizinische Fakultät, Eberhard Karls Universität Tübingen; Österbergstraße 3, 72074 Tübingen, Germany
| | - Sayed Iman Mousavi
- Department of Molecular Biophysics and Biochemistry; Yale University, New Haven, CT, USA
- Nanobiology Institute, Yale University; West Haven, CT 06516, USA
| | - Brigitte Naughton
- Department of Molecular Biophysics and Biochemistry; Yale University, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06520, USA
| | - Christina de Fontnouvelle
- Department of Molecular Biophysics and Biochemistry; Yale University, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06520, USA
| | - Barbara Fujita
- Department of Molecular Biophysics and Biochemistry; Yale University, New Haven, CT, USA
- Nanobiology Institute, Yale University; West Haven, CT 06516, USA
| | - Christian Schlieker
- Department of Molecular Biophysics and Biochemistry; Yale University, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06520, USA
| | - Shaul Yogev
- Department of Neuroscience, Yale University School of Medicine; New Haven, CT 06520, USA
| | - Yongli Zhang
- Department of Molecular Biophysics and Biochemistry; Yale University, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06520, USA
| | - Julien Berro
- Department of Molecular Biophysics and Biochemistry; Yale University, New Haven, CT, USA
- Nanobiology Institute, Yale University; West Haven, CT 06516, USA
- Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06520, USA
| |
Collapse
|
12
|
Nia HT, Munn LL, Jain RK. Probing the physical hallmarks of cancer. Nat Methods 2025:10.1038/s41592-024-02564-4. [PMID: 39815103 DOI: 10.1038/s41592-024-02564-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/11/2024] [Indexed: 01/18/2025]
Abstract
The physical microenvironment plays a crucial role in tumor development, progression, metastasis and treatment. Recently, we proposed four physical hallmarks of cancer, with distinct origins and consequences, to characterize abnormalities in the physical tumor microenvironment: (1) elevated compressive-tensile solid stresses, (2) elevated interstitial fluid pressure and the resulting interstitial fluid flow, (3) altered material properties (for example, increased tissue stiffness) and (4) altered physical micro-architecture. As this emerging field of physical oncology is being advanced by tumor biologists, cell and developmental biologists, engineers, physicists and oncologists, there is a critical need for model systems and measurement tools to mechanistically probe these physical hallmarks. Here, after briefly defining these physical hallmarks, we discuss the tools and model systems available for probing each hallmark in vitro, ex vivo, in vivo and in clinical settings. We finally review the unmet needs for mechanistic probing of the physical hallmarks of tumors and discuss the challenges and unanswered questions associated with each hallmark.
Collapse
Affiliation(s)
- Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | - Lance L Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Fardian-Melamed N, Skripka A, Ursprung B, Lee C, Darlington TP, Teitelboim A, Qi X, Wang M, Gerton JM, Cohen BE, Chan EM, Schuck PJ. Infrared nanosensors of piconewton to micronewton forces. Nature 2025; 637:70-75. [PMID: 39743607 DOI: 10.1038/s41586-024-08221-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 10/15/2024] [Indexed: 01/04/2025]
Abstract
Mechanical force is an essential feature for many physical and biological processes1-7, and remote measurement of mechanical signals with high sensitivity and spatial resolution is needed for diverse applications, including robotics8, biophysics9,10, energy storage11 and medicine12,13. Nanoscale luminescent force sensors excel at measuring piconewton forces, whereas larger sensors have proven powerful in probing micronewton forces14-16. However, large gaps remain in the force magnitudes that can be probed remotely from subsurface or interfacial sites, and no individual, non-invasive sensor is capable of measuring over the large dynamic range needed to understand many systems14,17. Here we demonstrate Tm3+-doped avalanching-nanoparticle18 force sensors that can be addressed remotely by deeply penetrating near-infrared light and can detect piconewton to micronewton forces with a dynamic range spanning more than four orders of magnitude. Using atomic force microscopy coupled with single-nanoparticle optical spectroscopy, we characterize the mechanical sensitivity of the photon-avalanching process and reveal its exceptional force responsiveness. By manipulating the Tm3+ concentrations and energy transfer within the nanosensors, we demonstrate different optical force-sensing modalities, including mechanobrightening and mechanochromism. The adaptability of these nanoscale optical force sensors, along with their multiscale-sensing capability, enable operation in the dynamic and versatile environments present in real-world, complex structures spanning biological organisms to nanoelectromechanical systems.
Collapse
Affiliation(s)
| | - Artiom Skripka
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Nanomaterials for Bioimaging Group, Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autόnoma de Madrid, Madrid, Spain
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
| | - Benedikt Ursprung
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Changhwan Lee
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Thomas P Darlington
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Ayelet Teitelboim
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Xiao Qi
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Maoji Wang
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT, USA
| | - Jordan M Gerton
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT, USA
| | - Bruce E Cohen
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Division of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - Emory M Chan
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - P James Schuck
- Department of Mechanical Engineering, Columbia University, New York, NY, USA.
| |
Collapse
|
14
|
Kruse K, Berthoz R, Barberi L, Reymann AC, Riveline D. Actomyosin clusters as active units shaping living matter. Curr Biol 2024; 34:R1045-R1058. [PMID: 39437723 DOI: 10.1016/j.cub.2024.08.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Stress generation by the actin cytoskeleton shapes cells and tissues. Despite impressive progress in live imaging and quantitative physical descriptions of cytoskeletal network dynamics, the connection between processes at molecular scales and spatiotemporal patterns at the cellular scale is still unclear. Here, we review studies reporting actomyosin clusters of micrometre size and with lifetimes of several minutes in a large number of organisms, ranging from fission yeast to humans. Such structures have also been found in reconstituted systems in vitro and in theoretical analyses of cytoskeletal dynamics. We propose that tracking these clusters could provide a simple readout for characterising living matter. Spatiotemporal patterns of clusters could serve as determinants of morphogenetic processes that have similar roles in diverse organisms.
Collapse
Affiliation(s)
- Karsten Kruse
- Departments of Theoretical Physics and Biochemistry, University of Geneva, 30 quai Ernest-Ansermet, 1204 Geneva, Switzerland.
| | - Rémi Berthoz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 1 Rue Laurent Fries, 67404 Illkirch CEDEX, France; Université de Strasbourg, IGBMC UMR 7104 - UMR-S 1258, F-67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, F-67400 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, UMR-S 1258, F-67400 Illkirch, France
| | - Luca Barberi
- Departments of Theoretical Physics and Biochemistry, University of Geneva, 30 quai Ernest-Ansermet, 1204 Geneva, Switzerland
| | - Anne-Cécile Reymann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 1 Rue Laurent Fries, 67404 Illkirch CEDEX, France; Université de Strasbourg, IGBMC UMR 7104 - UMR-S 1258, F-67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, F-67400 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, UMR-S 1258, F-67400 Illkirch, France
| | - Daniel Riveline
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 1 Rue Laurent Fries, 67404 Illkirch CEDEX, France; Université de Strasbourg, IGBMC UMR 7104 - UMR-S 1258, F-67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, F-67400 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, UMR-S 1258, F-67400 Illkirch, France.
| |
Collapse
|
15
|
Hart RG, Kota D, Li F, Zhang M, Ramallo D, Price AJ, Otterpohl KL, Smith SJ, Dunn AR, Huising MO, Liu J, Chandrasekar I. Myosin II tension sensors visualize force generation within the actin cytoskeleton in living cells. J Cell Sci 2024; 137:jcs262281. [PMID: 39369303 PMCID: PMC11698044 DOI: 10.1242/jcs.262281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
Nonmuscle myosin II (NMII) generates cytoskeletal forces that drive cell division, embryogenesis, muscle contraction and many other cellular functions. However, at present there is no method that can directly measure the forces generated by myosins in living cells. Here, we describe a Förster resonance energy transfer (FRET)-based tension sensor that can detect myosin-associated force along the filamentous actin network. Fluorescence lifetime imaging microscopy (FLIM)-FRET measurements indicate that the forces generated by NMII isoform B (NMIIB) exhibit significant spatial and temporal heterogeneity as a function of donor lifetime and fluorophore energy exchange. These measurements provide a proxy for inferred forces that vary widely along the actin cytoskeleton. This initial report highlights the potential utility of myosin-based tension sensors in elucidating the roles of cytoskeletal contractility in a wide variety of contexts.
Collapse
Affiliation(s)
- Ryan G. Hart
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
| | - Divya Kota
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701, USA
| | - Fangjia Li
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Mengdi Zhang
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Diego Ramallo
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Andrew J. Price
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Karla L. Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Steve J. Smith
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701, USA
| | - Alexander R. Dunn
- Department of Chemical Biology, Stanford University, Stanford, CA 94305, USA
| | - Mark O. Huising
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
- Department of Physiology and Membrane Biology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Jing Liu
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN 46907
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
16
|
Schrope JH, Horn A, Lazorchak K, Tinnen CW, Stevens JJ, Farooqui M, Li J, Bennin D, Robertson T, Juang TD, Ahmed A, Li C, Huttenlocher A, Beebe D. Confinement by liquid-liquid interface replicates in vivo neutrophil deformations and elicits bleb based migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.14.544898. [PMID: 38106211 PMCID: PMC10723256 DOI: 10.1101/2023.06.14.544898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Leukocytes navigate through interstitial spaces resulting in deformation of both the motile leukocytes and surrounding cells. Creating an in vitro system that models the deformable cellular environment encountered in vivo has been challenging. Here, we engineer microchannels with a liquid-liquid interface that exerts confining pressures (200-3000 Pa) similar to cells in tissues, and, thus, is deformable by cell generated forces. Consequently, the balance between migratory cell-generated and interfacial pressures determines the degree of confinement. Pioneer cells that first contact the interfacial barrier require greater deformation forces to forge a path for migration, and as a result migrate slower than trailing cells. Critically, resistive pressures are tunable by controlling the curvature of the liquid interface, which regulates motility. By granting cells autonomy in determining their confinement, and tuning environmental resistance, interfacial deformations are made to match those of surrounding cells in vivo during interstitial neutrophil migration in a larval zebrafish model. We discover that, in this context, neutrophils employ a bleb-based mechanism of force generation to deform a barrier exerting cell-scale confining pressures.
Collapse
|
17
|
Gandin A, Torresan V, Panciera T, Brusatin G. A Scalable Method to Fabricate 2D Hydrogel Substrates for Mechanobiology Studies with Independent Tuning of Adhesiveness and Stiffness. Methods Protoc 2024; 7:75. [PMID: 39452789 PMCID: PMC11510107 DOI: 10.3390/mps7050075] [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] [Received: 08/18/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
Mechanical signals from the extracellular matrix are crucial in guiding cellular behavior. Two-dimensional hydrogel substrates for cell cultures serve as exceptional tools for mechanobiology studies because they mimic the biomechanical and adhesive characteristics of natural environments. However, the interdisciplinary knowledge required to synthetize and manipulate these biomaterials typically restricts their widespread use in biological laboratories, which may not have the material science expertise or specialized instrumentation. To address this, we propose a scalable method that requires minimal setup to produce 2D hydrogel substrates with independent modulation of the rigidity and adhesiveness within the range typical of natural tissues. In this method, norbornene-terminated 8-arm polyethylene glycol is stoichiometrically functionalized with RGD peptides and crosslinked with a di-cysteine terminated peptide via a thiol-ene click reaction. Since the synthesis process significantly influences the final properties of the hydrogels, we provide a detailed description of the chemical procedure to ensure reproducibility and high throughput results. We demonstrate examples of cell mechanosignaling by monitoring the activation state of the mechanoeffector proteins YAP/TAZ. This method effectively dissects the influence of biophysical and adhesive cues on cell behavior. We believe that our procedure will be easily adopted by other cell biology laboratories, improving its accessibility and practical application.
Collapse
Affiliation(s)
- Alessandro Gandin
- Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131 Padova, PD, Italy; (A.G.); (V.T.)
- Consorzio INSTM, Padova RU, Via Marzolo 9, 35131 Padova, PD, Italy
| | - Veronica Torresan
- Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131 Padova, PD, Italy; (A.G.); (V.T.)
- Consorzio INSTM, Padova RU, Via Marzolo 9, 35131 Padova, PD, Italy
| | - Tito Panciera
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, PD, Italy;
| | - Giovanna Brusatin
- Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131 Padova, PD, Italy; (A.G.); (V.T.)
- Consorzio INSTM, Padova RU, Via Marzolo 9, 35131 Padova, PD, Italy
| |
Collapse
|
18
|
Cleveland DS, Gasvoda KL, Ding A, Alsberg E. Cell Contractile Forces Drive Spatiotemporal Morphing in 4D Bioprinted Living Constructs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.613990. [PMID: 39386675 PMCID: PMC11463471 DOI: 10.1101/2024.09.22.613990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Current 4D materials typically rely on external stimuli such as heat or light to accomplish changes in shape, limiting the biocompatibility of these materials. Here, a composite bioink consisting of oxidized and methacrylated alginate (OMA), methacrylated gelatin (GelMA), and gelatin microspheres is developed to accomplish free-standing 4D bioprinting of cell-laden structures driven by an internal stimulus: cell-contractile forces (CCF). 4D changes in shape are directed by forming bilayer constructs consisting of one cell-free and one cell-laden layer. Human mesenchymal stem cells (hMSCs) are encapsulated to demonstrate the ability to simultaneously induce changes in shape and chondrogenic differentiation. Finally, the capability to pattern each layer of the printed constructs is exhibited to obtain complex geometric changes, including bending around two separate, non-parallel axes. Bioprinting of such 4D constructs mediated by CCF empowers the formation of more complex constructs, contributing to a greater degree of in vitro biomimicry of biological 4D phenomena.
Collapse
|
19
|
Lan M, Liu Y, Liu J, Zhang J, Haider MA, Zhang Y, Zhang Q. Matrix Viscoelasticity Tunes the Mechanobiological Behavior of Chondrocytes. Cell Biochem Funct 2024; 42:e4126. [PMID: 39324844 DOI: 10.1002/cbf.4126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/25/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
In articular cartilage, the pericellular matrix acting as a specialized mechanical microenvironment modulates environmental signals to chondrocytes through mechanotransduction. Matrix viscoelastic alterations during cartilage development and osteoarthritis (OA) degeneration play an important role in regulating chondrocyte fate and cartilage matrix homeostasis. In recent years, scientists are gradually realizing the importance of matrix viscoelasticity in regulating chondrocyte function and phenotype. Notably, this is an emerging field, and this review summarizes the existing literatures to the best of our knowledge. This review provides an overview of the viscoelastic properties of hydrogels and the role of matrix viscoelasticity in directing chondrocyte behavior. In this review, we elaborated the mechanotransuction mechanisms by which cells sense and respond to the viscoelastic environment and also discussed the underlying signaling pathways. Moreover, emerging insights into the role of matrix viscoelasticity in regulating chondrocyte function and cartilage formation shed light into designing cell-instructive biomaterial. We also describe the potential use of viscoelastic biomaterials in cartilage tissue engineering and regenerative medicine. Future perspectives on mechanobiological comprehension of the viscoelastic behaviors involved in tissue homeostasis, cellular responses, and biomaterial design are highlighted. Finally, this review also highlights recent strategies utilizing viscoelastic hydrogels for designing cartilage-on-a-chip.
Collapse
Affiliation(s)
- Minhua Lan
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Yanli Liu
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Junjiang Liu
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Jing Zhang
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Muhammad Adnan Haider
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Yanjun Zhang
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Quanyou Zhang
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
20
|
Sripada SA, Hosseini M, Ramesh S, Wang J, Ritola K, Menegatti S, Daniele MA. Advances and opportunities in process analytical technologies for viral vector manufacturing. Biotechnol Adv 2024; 74:108391. [PMID: 38848795 DOI: 10.1016/j.biotechadv.2024.108391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/14/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Viral vectors are an emerging, exciting class of biologics whose application in vaccines, oncology, and gene therapy has grown exponentially in recent years. Following first regulatory approval, this class of therapeutics has been vigorously pursued to treat monogenic disorders including orphan diseases, entering hundreds of new products into pipelines. Viral vector manufacturing supporting clinical efforts has spurred the introduction of a broad swath of analytical techniques dedicated to assessing the diverse and evolving panel of Critical Quality Attributes (CQAs) of these products. Herein, we provide an overview of the current state of analytics enabling measurement of CQAs such as capsid and vector identities, product titer, transduction efficiency, impurity clearance etc. We highlight orthogonal methods and discuss the advantages and limitations of these techniques while evaluating their adaptation as process analytical technologies. Finally, we identify gaps and propose opportunities in enabling existing technologies for real-time monitoring from hardware, software, and data analysis viewpoints for technology development within viral vector biomanufacturing.
Collapse
Affiliation(s)
- Sobhana A Sripada
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Mahshid Hosseini
- Joint Department of Biomedical Engineering, North Carolina State University, and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Srivatsan Ramesh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Junhyeong Wang
- Joint Department of Biomedical Engineering, North Carolina State University, and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Kimberly Ritola
- North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA; Neuroscience Center, Brain Initiative Neurotools Vector Core, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA; Biomanufacturing Training and Education Center, North Carolina State University, 890 Main Campus Dr, Raleigh, NC 27695, USA.
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University, and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA; Department of Electrical and Computer Engineering, North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA.
| |
Collapse
|
21
|
Aydin H, Ozcelikkale A, Acar A. Exploiting Matrix Stiffness to Overcome Drug Resistance. ACS Biomater Sci Eng 2024; 10:4682-4700. [PMID: 38967485 PMCID: PMC11322920 DOI: 10.1021/acsbiomaterials.4c00445] [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: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Drug resistance is arguably one of the biggest challenges facing cancer research today. Understanding the underlying mechanisms of drug resistance in tumor progression and metastasis are essential in developing better treatment modalities. Given the matrix stiffness affecting the mechanotransduction capabilities of cancer cells, characterization of the related signal transduction pathways can provide a better understanding for developing novel therapeutic strategies. In this review, we aimed to summarize the recent advancements in tumor matrix biology in parallel to therapeutic approaches targeting matrix stiffness and its consequences in cellular processes in tumor progression and metastasis. The cellular processes governed by signal transduction pathways and their aberrant activation may result in activating the epithelial-to-mesenchymal transition, cancer stemness, and autophagy, which can be attributed to drug resistance. Developing therapeutic strategies to target these cellular processes in cancer biology will offer novel therapeutic approaches to tailor better personalized treatment modalities for clinical studies.
Collapse
Affiliation(s)
- Hakan
Berk Aydin
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| | - Altug Ozcelikkale
- Department
of Mechanical Engineering, Middle East Technical
University, 06800, Ankara, Turkey
- Graduate
Program of Biomedical Engineering, Middle
East Technical University, 06800, Ankara, Turkey
| | - Ahmet Acar
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| |
Collapse
|
22
|
Su J, Song Y, Zhu Z, Huang X, Fan J, Qiao J, Mao F. Cell-cell communication: new insights and clinical implications. Signal Transduct Target Ther 2024; 9:196. [PMID: 39107318 PMCID: PMC11382761 DOI: 10.1038/s41392-024-01888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/09/2024] [Accepted: 06/02/2024] [Indexed: 09/11/2024] Open
Abstract
Multicellular organisms are composed of diverse cell types that must coordinate their behaviors through communication. Cell-cell communication (CCC) is essential for growth, development, differentiation, tissue and organ formation, maintenance, and physiological regulation. Cells communicate through direct contact or at a distance using ligand-receptor interactions. So cellular communication encompasses two essential processes: cell signal conduction for generation and intercellular transmission of signals, and cell signal transduction for reception and procession of signals. Deciphering intercellular communication networks is critical for understanding cell differentiation, development, and metabolism. First, we comprehensively review the historical milestones in CCC studies, followed by a detailed description of the mechanisms of signal molecule transmission and the importance of the main signaling pathways they mediate in maintaining biological functions. Then we systematically introduce a series of human diseases caused by abnormalities in cell communication and their progress in clinical applications. Finally, we summarize various methods for monitoring cell interactions, including cell imaging, proximity-based chemical labeling, mechanical force analysis, downstream analysis strategies, and single-cell technologies. These methods aim to illustrate how biological functions depend on these interactions and the complexity of their regulatory signaling pathways to regulate crucial physiological processes, including tissue homeostasis, cell development, and immune responses in diseases. In addition, this review enhances our understanding of the biological processes that occur after cell-cell binding, highlighting its application in discovering new therapeutic targets and biomarkers related to precision medicine. This collective understanding provides a foundation for developing new targeted drugs and personalized treatments.
Collapse
Affiliation(s)
- Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Song
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Zhipeng Zhu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Xinyue Huang
- Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
23
|
Kandasamy A, Yeh YT, Serrano R, Mercola M, Del Álamo JC. Uncertainty-Aware Traction Force Microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.602172. [PMID: 39026786 PMCID: PMC11257441 DOI: 10.1101/2024.07.05.602172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Traction Force Microscopy (TFM) is a versatile tool to quantify cell-exerted forces by imaging and tracking fiduciary markers embedded in elastic substrates. The computations involved in TFM are ill-conditioned, and data smoothing or regularization is required to avoid overfitting the noise in the tracked substrate displacements. Most TFM calculations depend critically on the heuristic selection of regularization (hyper)parameters affecting the balance between overfitting and smoothing. However, TFM methods rarely estimate or account for measurement errors in substrate deformation to adjust the regularization level accordingly. Moreover, there is a lack of tools to quantify how these errors propagate to the recovered traction stresses. These limitations make it difficult to interpret TFM readouts and hinder comparing different experiments. This manuscript presents an uncertainty-aware TFM technique that estimates the variability in the magnitude and direction of the traction stress vector recovered at each point in space and time of each experiment. In this technique, substrate deformation and its uncertainty are quantified using a non-parametric bootstrap PIV method by resampling the microscopy image pixels (PIV-UQ). This information is passed to a hierarchical Bayesian framework that automatically selects its hyperparameters to perform spatially adaptive regularization conditioned on image quality and propagates the uncertainty to the traction stress readouts (TFM-UQ). We validate the performance of PIV-UQ and TFM-UQ using synthetic datasets with prescribed image quality variations and demonstrate the application of PIV-UQ and TFM-UQ to experimental datasets. These studies show that TFM-UQ locally adapts the level of smoothing, outperforming traditional regularization methods. They also illustrate how uncertainty-aware TFM tools can be used to objectively choose key image analysis parameters like PIV-UQ interrogation window size. We anticipate that these tools will allow for decoupling biological heterogeneity from measurement variability and facilitate automating the analysis of large datasets by parameter-free, input data-based regularization.
Collapse
|
24
|
Eliahoo P, Setayesh H, Hoffman T, Wu Y, Li S, Treweek JB. Viscoelasticity in 3D Cell Culture and Regenerative Medicine: Measurement Techniques and Biological Relevance. ACS MATERIALS AU 2024; 4:354-384. [PMID: 39006396 PMCID: PMC11240420 DOI: 10.1021/acsmaterialsau.3c00038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 07/16/2024]
Abstract
The field of mechanobiology is gaining prominence due to recent findings that show cells sense and respond to the mechanical properties of their environment through a process called mechanotransduction. The mechanical properties of cells, cell organelles, and the extracellular matrix are understood to be viscoelastic. Various technologies have been researched and developed for measuring the viscoelasticity of biological materials, which may provide insight into both the cellular mechanisms and the biological functions of mechanotransduction. Here, we explain the concept of viscoelasticity and introduce the major techniques that have been used to measure the viscoelasticity of various soft materials in different length- and timescale frames. The topology of the material undergoing testing, the geometry of the probe, the magnitude of the exerted stress, and the resulting deformation should be carefully considered to choose a proper technique for each application. Lastly, we discuss several applications of viscoelasticity in 3D cell culture and tissue models for regenerative medicine, including organoids, organ-on-a-chip systems, engineered tissue constructs, and tunable viscoelastic hydrogels for 3D bioprinting and cell-based therapies.
Collapse
Affiliation(s)
- Payam Eliahoo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089 United States
| | - Hesam Setayesh
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089 United States
| | - Tyler Hoffman
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095 United States
| | - Yifan Wu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095 United States
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095 United States
| | - Jennifer B Treweek
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089 United States
| |
Collapse
|
25
|
Cheung BCH, Abbed RJ, Wu M, Leggett SE. 3D Traction Force Microscopy in Biological Gels: From Single Cells to Multicellular Spheroids. Annu Rev Biomed Eng 2024; 26:93-118. [PMID: 38316064 DOI: 10.1146/annurev-bioeng-103122-031130] [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: 02/07/2024]
Abstract
Cell traction force plays a critical role in directing cellular functions, such as proliferation, migration, and differentiation. Current understanding of cell traction force is largely derived from 2D measurements where cells are plated on 2D substrates. However, 2D measurements do not recapitulate a vital aspect of living systems; that is, cells actively remodel their surrounding extracellular matrix (ECM), and the remodeled ECM, in return, can have a profound impact on cell phenotype and traction force generation. This reciprocal adaptivity of living systems is encoded in the material properties of biological gels. In this review, we summarize recent progress in measuring cell traction force for cells embedded within 3D biological gels, with an emphasis on cell-ECM cross talk. We also provide perspectives on tools and techniques that could be adapted to measure cell traction force in complex biochemical and biophysical environments.
Collapse
Affiliation(s)
- Brian C H Cheung
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York, USA;
| | - Rana J Abbed
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA;
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York, USA;
| | - Susan E Leggett
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA;
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
26
|
Hu Y, Li H, Zhang C, Feng J, Wang W, Chen W, Yu M, Liu X, Zhang X, Liu Z. DNA-based ForceChrono probes for deciphering single-molecule force dynamics in living cells. Cell 2024; 187:3445-3459.e15. [PMID: 38838668 DOI: 10.1016/j.cell.2024.05.008] [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: 02/13/2024] [Revised: 04/15/2024] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Understanding cellular force transmission dynamics is crucial in mechanobiology. We developed the DNA-based ForceChrono probe to measure force magnitude, duration, and loading rates at the single-molecule level within living cells. The ForceChrono probe circumvents the limitations of in vitro single-molecule force spectroscopy by enabling direct measurements within the dynamic cellular environment. Our findings reveal integrin force loading rates of 0.5-2 pN/s and durations ranging from tens of seconds in nascent adhesions to approximately 100 s in mature focal adhesions. The probe's robust and reversible design allows for continuous monitoring of these dynamic changes as cells undergo morphological transformations. Additionally, by analyzing how mutations, deletions, or pharmacological interventions affect these parameters, we can deduce the functional roles of specific proteins or domains in cellular mechanotransduction. The ForceChrono probe provides detailed insights into the dynamics of mechanical forces, advancing our understanding of cellular mechanics and the molecular mechanisms of mechanotransduction.
Collapse
Affiliation(s)
- Yuru Hu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Hongyun Li
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China.
| | - Chen Zhang
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Jingjing Feng
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Wenxu Wang
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Wei Chen
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Miao Yu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Xinping Liu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Xinghua Zhang
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China.
| | - Zheng Liu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei Province 430072, China.
| |
Collapse
|
27
|
Lin YJ, Tan XHM, Wang Y, Chung PS, Zhang X, Wu TH, Wu TY, Deb A, Chiou PY. Label-Free Optical Mapping for Large-Area Biomechanical Dynamics of Multicellular Systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598186. [PMID: 38915576 PMCID: PMC11195166 DOI: 10.1101/2024.06.12.598186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Mapping cellular activities over large areas is crucial for understanding the collective behaviors of multicellular systems. Biomechanical properties, such as cellular traction force, serve as critical regulators of physiological states and molecular configurations. However, existing technologies for mapping large-area biomechanical dynamics are limited by the small field of view and scanning nature. To address this, we propose a novel platform that utilizes a vast number of optical diffractive elements for mapping large-area biomechanical dynamics. This platform achieves a field-of-view of 10.6 mm X 10.6 mm, a three-orders-of-magnitude improvement over traditional traction force microscopy. Transient mechanical waves generated by monolayer neonatal rat ventricular myocytes were captured with high spatiotemporal resolution (130 fps and 20 µm for temporal and spatial resolution, respectively). Furthermore, its label-free nature allows for long-term observations extended to a week, with minimal disruption of cellular functions. Finally, simultaneous measurements of calcium ions concentrations and biomechanical dynamics are demonstrated.
Collapse
|
28
|
Chen H, Wang S, Cao Y, Lei H. Molecular Force Sensors for Biological Application. Int J Mol Sci 2024; 25:6198. [PMID: 38892386 PMCID: PMC11173168 DOI: 10.3390/ijms25116198] [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: 04/29/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The mechanical forces exerted by cells on their surrounding microenvironment are known as cellular traction forces. These forces play crucial roles in various biological processes, such as tissue development, wound healing and cell functions. However, it is hard for traditional techniques to measure cellular traction forces accurately because their magnitude (from pN to nN) and the length scales over which they occur (from nm to μm) are extremely small. In order to fully understand mechanotransduction, highly sensitive tools for measuring cellular forces are needed. Current powerful techniques for measuring traction forces include traction force microscopy (TFM) and fluorescent molecular force sensors (FMFS). In this review, we elucidate the force imaging principles of TFM and FMFS. Then we highlight the application of FMFS in a variety of biological processes and offer our perspectives and insights into the potential applications of FMFS.
Collapse
Affiliation(s)
- Huiyan Chen
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China; (H.C.); (S.W.)
| | - Shouhan Wang
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China; (H.C.); (S.W.)
| | - Yi Cao
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China; (H.C.); (S.W.)
| | - Hai Lei
- School of Physics, Zhejiang University, Hangzhou 310027, China
- Institute for Advanced Study in Physics, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
29
|
Liew HY, Liew XH, Lin WX, Lee YZ, Ong YS, Ogawa S, Chong LH. Cellular Traction Force Holds the Potential as a Drug Testing Readout for In Vitro Cancer Metastasis. Cell Mol Bioeng 2024; 17:203-217. [PMID: 39050509 PMCID: PMC11263313 DOI: 10.1007/s12195-024-00811-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 06/28/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Metastasis is responsible for 90% of cancer-related deaths worldwide. However, the potential inhibitory effects of metastasis by various anticancer drugs have been left largely unexplored. Existing preclinical models primarily focus on antiproliferative agents on the primary tumor to halt the cancer growth but not in metastasis. Unlike primary tumors, metastasis requires cancer cells to exert sufficient cellular traction force through the actomyosin machinery to migrate away from the primary tumor site. Therefore, we seek to explore the potential of cellular traction force as a novel readout for screening drugs that target cancer metastasis. Methods In vitro models of invasive and non-invasive breast cancer were first established using MDA-MB-231 and MCF-7 cell lines, respectively. Cellular morphology was characterized, revealing spindle-like morphology in MDA-MB-231 and spherical morphology in MCF-7 cells. The baseline cellular traction force was quantified using the Traction force Microscopy technique. Cisplatin, a paradigm antimetastatic drug, and 5-Fluorouracil (5FU), a non-antimetastatic drug, were selected to evaluate the potential of cellular traction force as a drug testing readout for the in vitro cancer metastasis. Results MDA-MB-231 cells exhibited significantly higher baseline cellular traction force compared to MCF-7 cells. Treatment with Cisplatin, an antimetastatic drug, and 5-Fluorouracil (5FU), a non-antimetastatic drug, demonstrated distinct effects on cellular traction force in MDA-MB-231 but not in MCF-7 cells. These findings correlate with the invasive potential observed in the two models. Conclusion Cellular traction force emerges as a promising metric for evaluating drug efficacy in inhibiting cancer metastasis using in vitro models. This approach could enhance the screening and development of novel anti-metastatic therapies, addressing a critical gap in current anticancer drug research.
Collapse
Affiliation(s)
- Hui Yan Liew
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Malaysia
| | - Xiao Hui Liew
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Malaysia
| | - Wei Xuan Lin
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Malaysia
| | - Yee Zhen Lee
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Malaysia
| | - Yong Sze Ong
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Malaysia
| | - Satoshi Ogawa
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Malaysia
| | - Lor Huai Chong
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500 Subang Jaya, Malaysia
| |
Collapse
|
30
|
Yang C, Yin D, Zhang H, Badea I, Yang SM, Zhang W. Cell Migration Assays and Their Application to Wound Healing Assays-A Critical Review. MICROMACHINES 2024; 15:720. [PMID: 38930690 PMCID: PMC11205366 DOI: 10.3390/mi15060720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
In recent years, cell migration assays (CMAs) have emerged as a tool to study the migration of cells along with their physiological responses under various stimuli, including both mechanical and bio-chemical properties. CMAs are a generic system in that they support various biological applications, such as wound healing assays. In this paper, we review the development of the CMA in the context of its application to wound healing assays. As such, the wound healing assay will be used to derive the requirements on CMAs. This paper will provide a comprehensive and critical review of the development of CMAs along with their application to wound healing assays. One salient feature of our methodology in this paper is the application of the so-called design thinking; namely we define the requirements of CMAs first and then take them as a benchmark for various developments of CMAs in the literature. The state-of-the-art CMAs are compared with this benchmark to derive the knowledge and technological gap with CMAs in the literature. We will also discuss future research directions for the CMA together with its application to wound healing assays.
Collapse
Affiliation(s)
- Chun Yang
- School of Mechanical Engineering, Donghua University, Shanghai 200051, China;
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Di Yin
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China; (D.Y.); (H.Z.)
| | - Hongbo Zhang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China; (D.Y.); (H.Z.)
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada;
| | - Shih-Mo Yang
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Wenjun Zhang
- School of Mechanical Engineering, Donghua University, Shanghai 200051, China;
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| |
Collapse
|
31
|
Huang Y, Chen T, Chen X, Chen X, Zhang J, Liu S, Lu M, Chen C, Ding X, Yang C, Huang R, Song Y. Decoding Biomechanical Cues Based on DNA Sensors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310330. [PMID: 38185740 DOI: 10.1002/smll.202310330] [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: 11/11/2023] [Revised: 12/18/2023] [Indexed: 01/09/2024]
Abstract
Biological systems perceive and respond to mechanical forces, generating mechanical cues to regulate life processes. Analyzing biomechanical forces has profound significance for understanding biological functions. Therefore, a series of molecular mechanical techniques have been developed, mainly including single-molecule force spectroscopy, traction force microscopy, and molecular tension sensor systems, which provide indispensable tools for advancing the field of mechanobiology. DNA molecules with a programmable structure and well-defined mechanical characteristics have attached much attention to molecular tension sensors as sensing elements, and are designed for the study of biomechanical forces to present biomechanical information with high sensitivity and resolution. In this work, a comprehensive overview of molecular mechanical technology is presented, with a particular focus on molecular tension sensor systems, specifically those based on DNA. Finally, the future development and challenges of DNA-based molecular tension sensor systems are looked upon.
Collapse
Affiliation(s)
- Yihao Huang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Ting Chen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Xiaodie Chen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Ximing Chen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jialu Zhang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Sinong Liu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Menghao Lu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chong Chen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Xiangyu Ding
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Ruiyun Huang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| |
Collapse
|
32
|
Karimi A, Aga M, Khan T, D'costa SD, Thaware O, White E, Kelley MJ, Gong H, Acott TS. Comparative analysis of traction forces in normal and glaucomatous trabecular meshwork cells within a 3D, active fluid-structure interaction culture environment. Acta Biomater 2024; 180:206-229. [PMID: 38641184 PMCID: PMC11095374 DOI: 10.1016/j.actbio.2024.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
This study presents a 3D in vitro cell culture model, meticulously 3D printed to replicate the conventional aqueous outflow pathway anatomical structure, facilitating the study of trabecular meshwork (TM) cellular responses under glaucomatous conditions. Glaucoma affects TM cell functionality, leading to extracellular matrix (ECM) stiffening, enhanced cell-ECM adhesion, and obstructed aqueous humor outflow. Our model, reconstructed from polyacrylamide gel with elastic moduli of 1.5 and 21.7 kPa, is based on serial block-face scanning electron microscopy images of the outflow pathway. It allows for quantifying 3D, depth-dependent, dynamic traction forces exerted by both normal and glaucomatous TM cells within an active fluid-structure interaction (FSI) environment. In our experimental design, we designed two scenarios: a control group with TM cells observed over 20 hours without flow (static setting), focusing on intrinsic cellular contractile forces, and a second scenario incorporating active FSI to evaluate its impact on traction forces (dynamic setting). Our observations revealed that active FSI results in higher traction forces (normal: 1.83-fold and glaucoma: 2.24-fold) and shear strains (normal: 1.81-fold and glaucoma: 2.41-fold), with stiffer substrates amplifying this effect. Glaucomatous cells consistently exhibited larger forces than normal cells. Increasing gel stiffness led to enhanced stress fiber formation in TM cells, particularly in glaucomatous cells. Exposure to active FSI dramatically altered actin organization in both normal and glaucomatous TM cells, particularly affecting cortical actin stress fiber arrangement. This model while preliminary offers a new method in understanding TM cell biomechanics and ECM stiffening in glaucoma, highlighting the importance of FSI in these processes. STATEMENT OF SIGNIFICANCE: This pioneering project presents an advanced 3D in vitro model, meticulously replicating the human trabecular meshwork's anatomy for glaucoma research. It enables precise quantification of cellular forces in a dynamic fluid-structure interaction, a leap forward from existing 2D models. This advancement promises significant insights into trabecular meshwork cell biomechanics and the stiffening of the extracellular matrix in glaucoma, offering potential pathways for innovative treatments. This research is positioned at the forefront of ocular disease study, with implications that extend to broader biomedical applications.
Collapse
Affiliation(s)
- Alireza Karimi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States; Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, United States.
| | - Mini Aga
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Taaha Khan
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Siddharth Daniel D'costa
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Omkar Thaware
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States; Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, United States
| | - Elizabeth White
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Mary J Kelley
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States; Department Integrative Biosciences, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Haiyan Gong
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA, United States; Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Ted S Acott
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States; Department Chemical Physiology & Biochemistry, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
33
|
Dorta D, Plazaola C, Carrasco J, Alves-Rosa MF, Coronado LM, Correa R, Zambrano M, Gutiérrez-Medina B, Sarmiento-Gómez E, Spadafora C, Gonzalez G. Mechanical Characterization of the Erythrocyte Membrane Using a Capacitor-Based Technique. MICROMACHINES 2024; 15:590. [PMID: 38793163 PMCID: PMC11122917 DOI: 10.3390/mi15050590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024]
Abstract
Pathological processes often change the mechanical properties of cells. Increased rigidity could be a marker of cellular malfunction. Erythrocytes are a type of cell that deforms to squeeze through tiny capillaries; changes in their rigidity can dramatically affect their functionality. Furthermore, differences in the homeostatic elasticity of the cell can be used as a tool for diagnosis and even for choosing the adequate treatment for some illnesses. More accurate types of equipment needed to study biomechanical phenomena at the single-cell level are very costly and thus out of reach for many laboratories around the world. This study presents a simple and low-cost technique to study the rigidity of red blood cells (RBCs) through the application of electric fields in a hand-made microfluidic chamber that uses a capacitor principle. As RBCs are deformed with the application of voltage, cells are observed under a light microscope. From mechanical force vs. deformation data, the elastic constant of the cells is determined. The results obtained with the capacitor-based method were compared with those obtained using optical tweezers, finding good agreement. In addition, P. falciparum-infected erythrocytes were tested with the electric field applicator. Our technique provides a simple means of testing the mechanical properties of individual cells.
Collapse
Affiliation(s)
- Doriana Dorta
- Facultad de Ciencias Naturales, Exactas y Tecnología, Universidad de Panamá, Panama City 06001-01103, Panama;
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama City 1843-01103, Panama; (J.C.); (M.F.A.-R.); (L.M.C.); (R.C.); (C.S.)
| | - Carlos Plazaola
- Facultad de Ingeniería Mecánica, Universidad Tecnológica de Panamá, Panama City 0819-07289, Panama;
| | - Jafeth Carrasco
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama City 1843-01103, Panama; (J.C.); (M.F.A.-R.); (L.M.C.); (R.C.); (C.S.)
| | - Maria F. Alves-Rosa
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama City 1843-01103, Panama; (J.C.); (M.F.A.-R.); (L.M.C.); (R.C.); (C.S.)
| | - Lorena M. Coronado
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama City 1843-01103, Panama; (J.C.); (M.F.A.-R.); (L.M.C.); (R.C.); (C.S.)
| | - Ricardo Correa
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama City 1843-01103, Panama; (J.C.); (M.F.A.-R.); (L.M.C.); (R.C.); (C.S.)
| | - Maytee Zambrano
- Facultad de Ingeniería Eléctrica, Universidad Tecnológica de Panamá, Panama City 0819-07289, Panama;
- Centro de Estudios Multidisciplinarios en Ciencias, Ingeniería y Tecnología (CEMCIT-AIP), Panama City 0819-07289, Panama
| | - Braulio Gutiérrez-Medina
- Advanced Materials Division, Instituto Potosino de Investigación Científica y Tecnológica A. C. (IPICYT), San Luis Potosí 78216, Mexico;
| | - Erick Sarmiento-Gómez
- Departamento de Ingeniería Física, División de Ciencias e Ingenierías, Campus León, Universidad de Guanajuato, Guanajuato 37320, Mexico;
| | - Carmenza Spadafora
- Centro de Biología Celular y Molecular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Ciudad del Saber, Panama City 1843-01103, Panama; (J.C.); (M.F.A.-R.); (L.M.C.); (R.C.); (C.S.)
| | - Guadalupe Gonzalez
- Facultad de Ingeniería Eléctrica, Universidad Tecnológica de Panamá, Panama City 0819-07289, Panama;
- Centro de Estudios Multidisciplinarios en Ciencias, Ingeniería y Tecnología (CEMCIT-AIP), Panama City 0819-07289, Panama
| |
Collapse
|
34
|
Li L, Griebel ME, Uroz M, Bubli SY, Gagnon KA, Trappmann B, Baker BM, Eyckmans J, Chen CS. A Protein-Adsorbent Hydrogel with Tunable Stiffness for Tissue Culture Demonstrates Matrix-Dependent Stiffness Responses. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2309567. [PMID: 38693998 PMCID: PMC11060701 DOI: 10.1002/adfm.202309567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Indexed: 05/03/2024]
Abstract
Although tissue culture plastic has been widely employed for cell culture, the rigidity of plastic is not physiologic. Softer hydrogels used to culture cells have not been widely adopted in part because coupling chemistries are required to covalently capture extracellular matrix (ECM) proteins and support cell adhesion. To create an in vitro system with tunable stiffnesses that readily adsorbs ECM proteins for cell culture, we present a novel hydrophobic hydrogel system via chemically converting hydroxyl residues on the dextran backbone to methacrylate groups, thereby transforming non-protein adhesive, hydrophilic dextran to highly protein adsorbent substrates. Increasing methacrylate functionality increases the hydrophobicity in the resulting hydrogels and enhances ECM protein adsorption without additional chemical reactions. These hydrophobic hydrogels permit facile and tunable modulation of substrate stiffness independent of hydrophobicity or ECM coatings. Using this approach, we show that substrate stiffness and ECM adsorption work together to affect cell morphology and proliferation, but the strengths of these effects vary in different cell types. Furthermore, we reveal that stiffness mediated differentiation of dermal fibroblasts into myofibroblasts is modulated by the substrate ECM. Our material system demonstrates remarkable simplicity and flexibility to tune ECM coatings and substrate stiffness and study their effects on cell function.
Collapse
Affiliation(s)
- Linqing Li
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire, 03824, United States
| | - Megan E Griebel
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
| | - Marina Uroz
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
| | - Saniya Yesmin Bubli
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire, 03824, United States
| | - Keith A Gagnon
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
| | - Britta Trappmann
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, 48149 Germany
| | - Brendon M Baker
- Engineered Microenvironments and Mechanobiology Lab, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109 United States
| | - Jeroen Eyckmans
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
| |
Collapse
|
35
|
Miyasaka S, Izumi K, Okuda S, Miki Y. Numerical assessment of the applicability of geometry-based force inference on homogeneous and heterogeneous cells. PLoS One 2024; 19:e0299016. [PMID: 38625886 PMCID: PMC11020637 DOI: 10.1371/journal.pone.0299016] [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: 06/14/2023] [Accepted: 02/03/2024] [Indexed: 04/18/2024] Open
Abstract
The measurement of cellular forces, which reflect crucial biological attributes, has the potential to replace conventional cell assessment methods, such as morphology, proliferation, and molecular expression analysis, in medical cell diagnosis and cell culture studies. In medical cell evaluations, force inference techniques have gained prominence due to their non-invasiveness and lack of requirement for specialized equipment. Among those techniques, the method proposed by Ishihara et al., which estimates forces in densely packed cells based only on cell geometry, is a promising method. However, its applicability range of this method has not been fully established. In this study, we employed a two-dimensional vertex model to numerically assess the applicability of this method on homogeneous and heterogeneous cells. Our comparisons between the true values from numerical simulations and the estimated values from the inference method revealed a significant correlation between estimation accuracy and cell roundness in systems of homogeneous cell. Moreover, the method demonstrated efficient force estimations in heterogeneous-cell systems. These findings may be useful when the force inference method is employed to evaluate medical cells.
Collapse
Affiliation(s)
| | | | - Satoru Okuda
- WPI-Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | | |
Collapse
|
36
|
Li M. Harnessing atomic force microscopy-based single-cell analysis to advance physical oncology. Microsc Res Tech 2024; 87:631-659. [PMID: 38053519 DOI: 10.1002/jemt.24467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/07/2023]
Abstract
Single-cell analysis is an emerging and promising frontier in the field of life sciences, which is expected to facilitate the exploration of fundamental laws of physiological and pathological processes. Single-cell analysis allows experimental access to cell-to-cell heterogeneity to reveal the distinctive behaviors of individual cells, offering novel opportunities to dissect the complexity of severe human diseases such as cancers. Among the single-cell analysis tools, atomic force microscopy (AFM) is a powerful and versatile one which is able to nondestructively image the fine topographies and quantitatively measure multiple mechanical properties of single living cancer cells in their native states under aqueous conditions with unprecedented spatiotemporal resolution. Over the past few decades, AFM has been widely utilized to detect the structural and mechanical behaviors of individual cancer cells during the process of tumor formation, invasion, and metastasis, yielding numerous unique insights into tumor pathogenesis from the biomechanical perspective and contributing much to the field of cancer mechanobiology. Here, the achievements of AFM-based analysis of single cancer cells to advance physical oncology are comprehensively summarized, and challenges and future perspectives are also discussed. RESEARCH HIGHLIGHTS: Achievements of AFM in characterizing the structural and mechanical behaviors of single cancer cells are summarized, and future directions are discussed. AFM is not only capable of visualizing cellular fine structures, but can also measure multiple cellular mechanical properties as well as cell-generated mechanical forces. There is still plenty of room for harnessing AFM-based single-cell analysis to advance physical oncology.
Collapse
Affiliation(s)
- Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
37
|
Denisin AK, Kim H, Riedel-Kruse IH, Pruitt BL. Field Guide to Traction Force Microscopy. Cell Mol Bioeng 2024; 17:87-106. [PMID: 38737454 PMCID: PMC11082129 DOI: 10.1007/s12195-024-00801-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/26/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Traction force microscopy (TFM) is a widely used technique to measure cell contractility on compliant substrates that mimic the stiffness of human tissues. For every step in a TFM workflow, users make choices which impact the quantitative results, yet many times the rationales and consequences for making these decisions are unclear. We have found few papers which show the complete experimental and mathematical steps of TFM, thus obfuscating the full effects of these decisions on the final output. Methods Therefore, we present this "Field Guide" with the goal to explain the mathematical basis of common TFM methods to practitioners in an accessible way. We specifically focus on how errors propagate in TFM workflows given specific experimental design and analytical choices. Results We cover important assumptions and considerations in TFM substrate manufacturing, substrate mechanical properties, imaging techniques, image processing methods, approaches and parameters used in calculating traction stress, and data-reporting strategies. Conclusions By presenting a conceptual review and analysis of TFM-focused research articles published over the last two decades, we provide researchers in the field with a better understanding of their options to make more informed choices when creating TFM workflows depending on the type of cell being studied. With this review, we aim to empower experimentalists to quantify cell contractility with confidence. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00801-6.
Collapse
Affiliation(s)
| | - Honesty Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305 USA
- Present Address: The Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158 USA
- Department of Molecular and Cellular Biology, and (by courtesy) Departments of Biomedical Engineering, Applied Mathematics, and Physics, University of Arizona, Tucson, AZ 85721 USA
| | - Ingmar H. Riedel-Kruse
- Department of Molecular and Cellular Biology, and (by courtesy) Departments of Biomedical Engineering, Applied Mathematics, and Physics, University of Arizona, Tucson, AZ 85721 USA
| | - Beth L. Pruitt
- Departments of Bioengineering and Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA 93106 USA
| |
Collapse
|
38
|
Jain N, Shashi Bhushan BL, Natarajan M, Mehta R, Saini DK, Chatterjee K. Advanced 3D In Vitro Lung Fibrosis Models: Contemporary Status, Clinical Uptake, and Prospective Outlooks. ACS Biomater Sci Eng 2024; 10:1235-1261. [PMID: 38335198 DOI: 10.1021/acsbiomaterials.3c01499] [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] [Indexed: 02/12/2024]
Abstract
Fibrosis has been characterized as a global health problem and ranks as one of the primary causes of organ dysfunction. Currently, there is no cure for pulmonary fibrosis, and limited therapeutic options are available due to an inadequate understanding of the disease pathogenesis. The absence of advanced in vitro models replicating dynamic temporal changes observed in the tissue with the progression of the disease is a significant impediment in the development of novel antifibrotic treatments, which has motivated research on tissue-mimetic three-dimensional (3D) models. In this review, we summarize emerging trends in preparing advanced lung models to recapitulate biochemical and biomechanical processes associated with lung fibrogenesis. We begin by describing the importance of in vivo studies and highlighting the often poor correlation between preclinical research and clinical outcomes and the limitations of conventional cell culture in accurately simulating the 3D tissue microenvironment. Rapid advancement in biomaterials, biofabrication, biomicrofluidics, and related bioengineering techniques are enabling the preparation of in vitro models to reproduce the epithelium structure and operate as reliable drug screening strategies for precise prediction. Improving and understanding these model systems is necessary to find the cross-talks between growing cells and the stage at which myofibroblasts differentiate. These advanced models allow us to utilize the knowledge and identify, characterize, and hand pick medicines beneficial to the human community. The challenges of the current approaches, along with the opportunities for further research with potential for translation in this field, are presented toward developing novel treatments for pulmonary fibrosis.
Collapse
Affiliation(s)
- Nipun Jain
- Department of Materials Engineering, Indian Institute of Science, C.V Raman Avenue, Bangalore 560012 India
| | - B L Shashi Bhushan
- Department of Pulmonary Medicine, Victoria Hospital, Bangalore Medical College and Research Institute, Bangalore 560002 India
| | - M Natarajan
- Department of Pathology, Victoria Hospital, Bangalore Medical College and Research Institute, Bangalore 560002 India
| | - Ravi Mehta
- Department of Pulmonology and Critical Care, Apollo Hospitals, Jayanagar, Bangalore 560011 India
| | - Deepak Kumar Saini
- Department of Developmental Biology and Genetics, Indian Institute of Science, C.V Raman Avenue, Bangalore 560012 India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, C.V Raman Avenue, Bangalore 560012 India
| |
Collapse
|
39
|
Karimi A, Aga M, Khan T, D'costa SD, Cardenas-Riumallo S, Zelenitz M, Kelley MJ, Acott TS. Dynamic traction force in trabecular meshwork cells: A 2D culture model for normal and glaucomatous states. Acta Biomater 2024; 175:138-156. [PMID: 38151067 PMCID: PMC10843681 DOI: 10.1016/j.actbio.2023.12.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
Glaucoma, which is associated with intraocular pressure (IOP) elevation, results in trabecular meshwork (TM) cellular dysfunction, leading to increased rigidity of the extracellular matrix (ECM), larger adhesion forces between the TM cells and ECM, and higher resistance to aqueous humor drainage. TM cells sense the mechanical forces due to IOP dynamic and apply multidimensional forces on the ECM. Recognizing the importance of cellular forces in modulating various cellular activities and development, this study is aimed to develop a 2D in vitro cell culture model to calculate the 3D, depth-dependent, dynamic traction forces, tensile/compressive/shear strain of the normal and glaucomatous human TM cells within a deformable polyacrylamide (PAM) gel substrate. Normal and glaucomatous human TM cells were isolated, cultured, and seeded on top of the PAM gel substrate with embedded FluoSpheres, spanning elastic moduli of 1.5 to 80 kPa. Sixteen-hour post-seeding live confocal microscopy in an incubator was conducted to Z-stack image the 3D displacement map of the FluoSpheres within the PAM gels. Combined with the known PAM gel stiffness, we ascertained the 3D traction forces in the gel. Our results revealed meaningfully larger traction forces in the glaucomatous TM cells compared to the normal TM cells, reaching depths greater than 10-µm in the PAM gel substrate. Stress fibers in TM cells increased with gel rigidity, but diminished when stiffness rose from 20 to 80 kPa. The developed 2D cell culture model aids in understanding how altered mechanical properties in glaucoma impact TM cell behavior and aqueous humor outflow resistance. STATEMENT OF SIGNIFICANCE: Glaucoma, a leading cause of irreversible blindness, is intricately linked to elevated intraocular pressures and their subsequent cellular effects. The trabecular meshwork plays a pivotal role in this mechanism, particularly its interaction with the extracellular matrix. This research unveils an advanced 2D in vitro cell culture model that intricately maps the complex 3D forces exerted by trabecular meshwork cells on the extracellular matrix, offering unparalleled insights into the cellular biomechanics at play in both healthy and glaucomatous eyes. By discerning the changes in these forces across varying substrate stiffness levels, we bridge the gap in understanding between cellular mechanobiology and the onset of glaucoma. The findings stand as a beacon for potential therapeutic avenues, emphasizing the gravity of cellular/extracellular matrix interactions in glaucoma's pathogenesis and setting the stage for targeted interventions in its early stages.
Collapse
Affiliation(s)
- Alireza Karimi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA; Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
| | - Mini Aga
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Taaha Khan
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Siddharth Daniel D'costa
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | | | - Meadow Zelenitz
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Mary J Kelley
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA; Department Integrative Biosciences, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
| | - Ted S Acott
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA; Department Chemical Physiology & Biochemistry, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
40
|
Tabrizi MA, Bhattacharyya P, Zheng R, You M. Electrochemical DNA-based sensors for measuring cell-generated forces. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569814. [PMID: 38106148 PMCID: PMC10723317 DOI: 10.1101/2023.12.03.569814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Mechanical forces play an important role in cellular communication and signaling. We developed in this study novel electrochemical DNA-based force sensors for measuring cell-generated adhesion forces. Two types of DNA probes, i.e., tension gauge tether and DNA hairpin, were constructed on the surface of a smartphone-based electrochemical device to detect piconewton-scale cellular forces at tunable levels. Upon experiencing cellular tension, the unfolding of DNA probes induces the separation of redox reporters from the surface of the electrode, which results in detectable electrochemical signals. Using integrin-mediated cell adhesion as an example, our results indicated that these electrochemical sensors can be used for highly sensitive, robust, simple, and portable measurement of cell-generated forces.
Collapse
Affiliation(s)
| | | | - Ru Zheng
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | - Mingxu You
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
41
|
Nauryzgaliyeva Z, Goux Corredera I, Garreta E, Montserrat N. Harnessing mechanobiology for kidney organoid research. Front Cell Dev Biol 2023; 11:1273923. [PMID: 38077999 PMCID: PMC10704179 DOI: 10.3389/fcell.2023.1273923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 10/16/2024] Open
Abstract
Recently, organoids have emerged as revolutionizing tools with the unprecedented potential to recreate organ-specific microanatomy in vitro. Upon their derivation from human pluripotent stem cells (hPSCs), organoids reveal the blueprints of human organogenesis, further allowing the faithful recapitulation of their physiology. Nevertheless, along with the evolution of this field, advanced research exposed the organoids' shortcomings, particularly regarding poor reproducibility rates and overall immatureness. To resolve these challenges, many studies have started to underscore the relevance of mechanical cues as a relevant source to induce and externally control hPSCs differentiation. Indeed, established organoid generation protocols from hPSCs have mainly relyed on the biochemical induction of fundamental signalling pathways present during kidney formation in mammals, whereas mechanical cues have largely been unexplored. This review aims to discuss the pertinence of (bio) physical cues within hPSCs-derived organoid cultures, while deciphering their effect on morphogenesis. Moreover, we will explore state-of-the-art mechanobiology techniques as revolutionizing means for understanding the underlying role of mechanical forces in biological processes in organoid model systems.
Collapse
Affiliation(s)
- Zarina Nauryzgaliyeva
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Iphigénie Goux Corredera
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), University of Barcelona, Barcelona, Spain
| | - Elena Garreta
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), University of Barcelona, Barcelona, Spain
| | - Nuria Montserrat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
42
|
Wang H, Miao X, Zhai C, Chen Y, Lin Z, Zhou X, Guo M, Chai Z, Wang R, Shen W, Li H, Hu C. Mechanistic Insights into the Folding Mechanism of Region V in Ice-Binding Protein Secreted by Marinomonas primoryensis Revealed by Single-Molecule Force Spectroscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:16128-16137. [PMID: 37916685 DOI: 10.1021/acs.langmuir.3c02257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
The Gram-negative bacteria Marinomonas primoryensis secrete an ice-binding protein (MpIBP), which is a vital bacterial adhesin facilitating the adaptation and survival of the bacteria in the harsh Antarctic environment. The C-terminal region of MpIBP, known as region V (RV), is the first domain to be exported into the Ca2+-rich extracellular environment and acts as a folding nucleus for the entire adhesin. However, the mechanisms underlying the secretion and folding of RV remain poorly understood. Here, we used optical tweezers (OT) to investigate the secretion and folding mechanisms of RV at the single-molecule level. In the absence of Ca2+, apo-RV remains unstructured, while Ca2+-bound RV folds into a mechanically stable structure. The folding of RV could occur via the formation of an intermediate state. Even though this folding intermediate is "hidden" during the folding process of wild type RV in vitro, it likely forms in vivo and plays an important role in facilitating protein secretion. Additionally, our results revealed that the N-terminal part of the RV can significantly stabilize its C-terminal structure. Our study paves the way for further investigations into the structure and functions of MpIBP that help bacteria survive in challenging environments.
Collapse
Affiliation(s)
- Han Wang
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Xiaopu Miao
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| | - Cong Zhai
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| | - Yulu Chen
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| | - Zuzeng Lin
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| | - Xiaowei Zhou
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| | - Mengdi Guo
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| | - Zhongyan Chai
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| | - Ruifen Wang
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| | - Wanfu Shen
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| | - Hongbin Li
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Chunguang Hu
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
43
|
Li Q, Chen Z, Zhang Y, Ding S, Ding H, Wang L, Xie Z, Fu Y, Wei M, Liu S, Chen J, Wang X, Gu Z. Imaging cellular forces with photonic crystals. Nat Commun 2023; 14:7369. [PMID: 37963911 PMCID: PMC10646022 DOI: 10.1038/s41467-023-43090-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 10/31/2023] [Indexed: 11/16/2023] Open
Abstract
Current techniques for visualizing and quantifying cellular forces have limitations in live cell imaging, throughput, and multi-scale analysis, which impede progress in cell force research and its practical applications. We developed a photonic crystal cellular force microscopy (PCCFM) to image vertical cell forces over a wide field of view (1.3 mm ⨯ 1.0 mm, a 10 ⨯ objective image) at high speed (about 20 frames per second) without references. The photonic crystal hydrogel substrate (PCS) converts micro-nano deformations into perceivable color changes, enabling in situ visualization and quantification of tiny vertical cell forces with high throughput. It enabled long-term, cross-scale monitoring from subcellular focal adhesions to tissue-level cell sheets and aggregates.
Collapse
Affiliation(s)
- Qiwei Li
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Zaozao Chen
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
- Institute of Biomaterials and Medical Devices, Southeast University, 215163, Suzhou, Jiangsu, China
| | - Ying Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Shuang Ding
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Haibo Ding
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Luping Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
- Faculty of Sports Science, Ningbo University, 315211, Ningbo, China
| | - Zhuoying Xie
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Yifu Fu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Mengxiao Wei
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Shengnan Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Jialun Chen
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Xuan Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China.
- Institute of Biomaterials and Medical Devices, Southeast University, 215163, Suzhou, Jiangsu, China.
| |
Collapse
|
44
|
Wang W, Chen W, Wu C, Zhang C, Feng J, Liu P, Hu Y, Li H, Sun F, Jiang K, Zhang X, Liu Z. Hydrogel-based molecular tension fluorescence microscopy for investigating receptor-mediated rigidity sensing. Nat Methods 2023; 20:1780-1789. [PMID: 37798478 DOI: 10.1038/s41592-023-02037-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 09/05/2023] [Indexed: 10/07/2023]
Abstract
Extracellular matrix (ECM) rigidity serves as a crucial mechanical cue impacting diverse biological processes. However, understanding the molecular mechanisms of rigidity sensing has been limited by the spatial resolution and force sensitivity of current cellular force measurement techniques. Here we developed a method to functionalize DNA tension probes on soft hydrogel surfaces in a controllable and reliable manner, enabling molecular tension fluorescence microscopy for rigidity sensing studies. Our findings showed that fibroblasts respond to substrate rigidity by recruiting more force-bearing integrins and modulating integrin sampling frequency of the ECM, rather than simply overloading the existing integrin-ligand bonds, to promote focal adhesion maturation. We also demonstrated that ECM rigidity positively regulates the pN force of T cell receptor-ligand bond and T cell receptor mechanical sampling frequency, promoting T cell activation. Thus, hydrogel-based molecular tension fluorescence microscopy implemented on a standard confocal microscope provides a simple and effective means to explore detailed molecular force information for rigidity-dependent biological processes.
Collapse
Affiliation(s)
- Wenxu Wang
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Wei Chen
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Chaoyang Wu
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Chen Zhang
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Jingjing Feng
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Pengxiang Liu
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Yuru Hu
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Hongyun Li
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Feng Sun
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Kai Jiang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xinghua Zhang
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Zheng Liu
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan, China.
- College of Life Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
45
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
46
|
Tian Q, Yang F, Jiang H, Bhattacharyya P, Xie T, Ali AA, Sun Y, You M. Imaging and detecting intercellular tensile forces in spheroids and embryoid bodies using lipid-modified DNA probes. Front Cell Dev Biol 2023; 11:1220079. [PMID: 37920824 PMCID: PMC10619156 DOI: 10.3389/fcell.2023.1220079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023] Open
Abstract
Cells continuously experience and respond to different physical forces that are used to regulate their physiology and functions. Our ability to measure these mechanical cues is essential for understanding the bases of various mechanosensing and mechanotransduction processes. While multiple strategies have been developed to study mechanical forces within two-dimensional (2D) cell culture monolayers, the force measurement at cell-cell junctions in real three-dimensional (3D) cell models is still pretty rare. Considering that in real biological systems, cells are exposed to forces from 3D directions, measuring these molecular forces in their native environment is thus highly critical for the better understanding of different development and disease processes. We have recently developed a type of DNA-based molecular probe for measuring intercellular tensile forces in 2D cell models. Herein, we will report the further development and first-time usage of these molecular tension probes to visualize and detect mechanical forces within 3D spheroids and embryoid bodies (EBs). These probes can spontaneously anchor onto live cell membranes via the attached lipid moieties. By varying the concentrations of these DNA probes and their incubation time, we have first characterized the kinetics and efficiency of probe penetration and loading onto tumor spheroids and stem cell EBs of different sizes. After optimization, we have further imaged and measured E-cadherin-mediated forces in these 3D spheroids and EBs for the first time. Our results indicated that these DNA-based molecular tension probes can be used to study the spatiotemporal distributions of target mechanotransduction processes. These powerful imaging tools may be potentially applied to fill the gap between ongoing research of biomechanics in 2D systems and that in real 3D cell complexes.
Collapse
Affiliation(s)
- Qian Tian
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, United States
| | - Feiyu Yang
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| | - Han Jiang
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| | - Priyanka Bhattacharyya
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, United States
| | - Tianfa Xie
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| | - Ahsan Ausaf Ali
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, United States
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA, United States
| | - Mingxu You
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
47
|
Luo Y, Sun C, Wei M, Ma H, Wu Y, Chen Z, Dai H, Jian J, Sun B, Zhong C, Li J, Richardson KA, Lin H, Li L. Integrated Flexible Microscale Mechanical Sensors Based on Cascaded Free Spectral Range-Free Cavities. NANO LETTERS 2023; 23:8898-8906. [PMID: 37676244 DOI: 10.1021/acs.nanolett.3c02239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Photonic mechanical sensors offer several advantages over their electronic counterparts, including immunity to electromagnetic interference, increased sensitivity, and measurement accuracy. Exploring flexible mechanical sensors on deformable substrates provides new opportunities for strain-optical coupling operations. Nevertheless, existing flexible photonics strategies often require cumbersome signal collection and analysis with bulky setups, limiting their portability and affordability. To address these challenges, we propose a waveguide-integrated flexible mechanical sensor based on cascaded photonic crystal microcavities with inherent deformation and biaxial tensile state analysis. Leveraging the advanced multiplexing capability of the sensor, for the first time, we successfully demonstrate 2D shape reconstruction and quasi-distributed strain sensing with 110 μm spatial resolution. Our microscale mechanical sensor also exhibits exceptional sensitivity with a detected force level as low as 13.6 μN in real-time measurements. This sensing platform has potential applications in various fields, including biomedical sensing, surgical catheters, aircraft and spacecraft engineering, and robotic photonic skin development.
Collapse
Affiliation(s)
- Ye Luo
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Chunlei Sun
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Maoliang Wei
- State Key Laboratory of Modern Optical Instrumentation, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310027, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310027, China
| | - Hui Ma
- State Key Laboratory of Modern Optical Instrumentation, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310027, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310027, China
| | - Yingchun Wu
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Zequn Chen
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Hao Dai
- State Key Laboratory of Modern Optical Instrumentation, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310027, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310027, China
| | - Jialing Jian
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, China
- State Key Laboratory of Modern Optical Instrumentation, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310027, China
| | - Boshu Sun
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, China
- State Key Laboratory of Modern Optical Instrumentation, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310027, China
| | - Chuyu Zhong
- State Key Laboratory of Modern Optical Instrumentation, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310027, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310027, China
| | - Junying Li
- State Key Laboratory of Modern Optical Instrumentation, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310027, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310027, China
| | - Kathleen A Richardson
- The College of Optics & Photonics, Department of Materials Science & Engineering, University of Central Florida, Orlando, Florida 32816, United States
| | - Hongtao Lin
- State Key Laboratory of Modern Optical Instrumentation, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou 310027, China
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration, Zhejiang University, Hangzhou 310027, China
| | - Lan Li
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| |
Collapse
|
48
|
Cheikh MI, Tchoufag J, Osterfield M, Dean K, Bhaduri S, Zhang C, Mandadapu KK, Doubrovinski K. A comprehensive model of Drosophila epithelium reveals the role of embryo geometry and cell topology in mechanical responses. eLife 2023; 12:e85569. [PMID: 37782009 PMCID: PMC10584372 DOI: 10.7554/elife.85569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 09/29/2023] [Indexed: 10/03/2023] Open
Abstract
In order to understand morphogenesis, it is necessary to know the material properties or forces shaping the living tissue. In spite of this need, very few in vivo measurements are currently available. Here, using the early Drosophila embryo as a model, we describe a novel cantilever-based technique which allows for the simultaneous quantification of applied force and tissue displacement in a living embryo. By analyzing data from a series of experiments in which embryonic epithelium is subjected to developmentally relevant perturbations, we conclude that the response to applied force is adiabatic and is dominated by elastic forces and geometric constraints, or system size effects. Crucially, computational modeling of the experimental data indicated that the apical surface of the epithelium must be softer than the basal surface, a result which we confirmed experimentally. Further, we used the combination of experimental data and comprehensive computational model to estimate the elastic modulus of the apical surface and set a lower bound on the elastic modulus of the basal surface. More generally, our investigations revealed important general features that we believe should be more widely addressed when quantitatively modeling tissue mechanics in any system. Specifically, different compartments of the same cell can have very different mechanical properties; when they do, they can contribute differently to different mechanical stimuli and cannot be merely averaged together. Additionally, tissue geometry can play a substantial role in mechanical response, and cannot be neglected.
Collapse
Affiliation(s)
- Mohamad Ibrahim Cheikh
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Joel Tchoufag
- Department of Chemical and Biomolecular Engineering, University of California, BerkeleyBerkeleyUnited States
- Chemical Sciences Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | - Miriam Osterfield
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Kevin Dean
- Department of Bioinformatics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Swayamdipta Bhaduri
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Chuzhong Zhang
- Department of Material Science and Engineering, University of Texas at ArlingtonArlingtonUnited States
| | - Kranthi Kiran Mandadapu
- Department of Chemical and Biomolecular Engineering, University of California, BerkeleyBerkeleyUnited States
- Chemical Sciences Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | - Konstantin Doubrovinski
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
49
|
White MJ, Singh T, Wang E, Smith Q, Kutys ML. 'Chip'-ing away at morphogenesis - application of organ-on-chip technologies to study tissue morphogenesis. J Cell Sci 2023; 136:jcs261130. [PMID: 37795818 PMCID: PMC10565497 DOI: 10.1242/jcs.261130] [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: 10/06/2023] Open
Abstract
Emergent cell behaviors that drive tissue morphogenesis are the integrated product of instructions from gene regulatory networks, mechanics and signals from the local tissue microenvironment. How these discrete inputs intersect to coordinate diverse morphogenic events is a critical area of interest. Organ-on-chip technology has revolutionized the ability to construct and manipulate miniaturized human tissues with organotypic three-dimensional architectures in vitro. Applications of organ-on-chip platforms have increasingly transitioned from proof-of-concept tissue engineering to discovery biology, furthering our understanding of molecular and mechanical mechanisms that operate across biological scales to orchestrate tissue morphogenesis. Here, we provide the biological framework to harness organ-on-chip systems to study tissue morphogenesis, and we highlight recent examples where organ-on-chips and associated microphysiological systems have enabled new mechanistic insight in diverse morphogenic settings. We further highlight the use of organ-on-chip platforms as emerging test beds for cell and developmental biology.
Collapse
Affiliation(s)
- Matthew J. White
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Tania Singh
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
- UCSF-UC Berkeley Joint Program in Bioengineering, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Matthew L. Kutys
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
- UCSF-UC Berkeley Joint Program in Bioengineering, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
50
|
Wang H, Lu J, Rathod M, Aw WY, Huang SA, Polacheck WJ. A facile fluid pressure system reveals differential cellular response to interstitial pressure gradients and flow. BIOMICROFLUIDICS 2023; 17:054103. [PMID: 37781136 PMCID: PMC10539030 DOI: 10.1063/5.0165119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/09/2023] [Indexed: 10/03/2023]
Abstract
Interstitial fluid pressure gradients and interstitial flow have been shown to drive morphogenic processes that shape tissues and influence progression of diseases including cancer. The advent of porous media microfluidic approaches has enabled investigation of the cellular response to interstitial flow, but questions remain as to the critical biophysical and biochemical signals imparted by interstitial fluid pressure gradients and resulting flow on resident cells and extracellular matrix (ECM). Here, we introduce a low-cost method to maintain physiological interstitial fluid pressures that is built from commonly accessible laboratory equipment, including a laser pointer, camera, Arduino board, and a commercially available linear actuator. We demonstrate that when the system is connected to a microfluidic device containing a 3D porous hydrogel, physiologic pressure is maintained with sub-Pascal resolution and when basic feedback control is directed using an Arduino, constant pressure and pressure gradient can be maintained even as cells remodel and degrade the ECM hydrogel over time. Using this model, we characterized breast cancer cell growth and ECM changes to ECM fibril structure and porosity in response to constant interstitial fluid pressure or constant interstitial flow. We observe increased collagen fibril bundling and the formation of porous structures in the vicinity of cancer cells in response to constant interstitial fluid pressure as compared to constant interstitial flow. Collectively, these results further define interstitial fluid pressure as a driver of key pathogenic responses in cells, and the systems and methods developed here will allow for future mechanistic work investigating mechanotransduction of interstitial fluid pressures and flows.
Collapse
Affiliation(s)
- Hao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Jingming Lu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Mitesh Rathod
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Stephanie A. Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | | |
Collapse
|