1
|
Mierke CT. Softness or Stiffness What Contributes to Cancer and Cancer Metastasis? Cells 2025; 14:584. [PMID: 40277910 PMCID: PMC12026216 DOI: 10.3390/cells14080584] [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: 03/11/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Beyond the genomic and proteomic analysis of bulk and single cancer cells, a new focus of cancer research is emerging that is based on the mechanical analysis of cancer cells. Therefore, several biophysical techniques have been developed and adapted. The characterization of cancer cells, like human cancer cell lines, started with their mechanical characterization at mostly a single timepoint. A universal hypothesis has been proposed that cancer cells need to be softer to migrate and invade tissues and subsequently metastasize in targeted organs. Thus, the softness of cancer cells has been suggested to serve as a universal physical marker for the malignancy of cancer types. However, it has turned out that there exists the opposite phenomenon, namely that stiffer cancer cells are more migratory and invasive and therefore lead to more metastases. These contradictory results question the universality of the role of softness of cancer cells in the malignant progression of cancers. Another problem is that the various biophysical techniques used can affect the mechanical properties of cancer cells, making it even more difficult to compare the results of different studies. Apart from the instrumentation, the culture and measurement conditions of the cancer cells can influence the mechanical measurements. The review highlights the main advances of the mechanical characterization of cancer cells, discusses the strength and weaknesses of the approaches, and questions whether the passive mechanical characterization of cancer cells is still state-of-the art. Besides the cell models, conditions and biophysical setups, the role of the microenvironment on the mechanical characteristics of cancer cells is presented and debated. Finally, combinatorial approaches to determine the malignant potential of tumors, such as the involvement of the ECM, the cells in a homogeneous or heterogeneous association, or biological multi-omics analyses, together with the dynamic-mechanical analysis of cancer cells, are highlighted as new frontiers of research.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Sciences, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
2
|
Tikhonova TN, Barkovaya AV, Efremov YM, Mamed-Nabizade VV, Kolmogorov VS, Timashev PS, Sysoev NN, Fadeev VV, Gorelkin PV, Adler-Abramovich L, Erofeev AS, Shirshin EA. Non-Invasive Nanometer Resolution Assessment of Cell-Soft Hydrogel System Mechanical Properties by Scanning Ion Conductance Microscopy. Int J Mol Sci 2024; 25:13479. [PMID: 39769241 PMCID: PMC11678895 DOI: 10.3390/ijms252413479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Biomimetic hydrogels have garnered increased interest due to their considerable potential for use in various fields, such as tissue engineering, 3D cell cultivation, and drug delivery. The primary challenge for applying hydrogels in tissue engineering is accurately evaluating their mechanical characteristics. In this context, we propose a method using scanning ion conductance microscopy (SICM) to determine the rigidity of living human breast cancer cells MCF-7 cells grown on a soft, self-assembled Fmoc-FF peptide hydrogel. Moreover, it is demonstrated that the map of Young's modulus distribution obtained by the SICM method allows for determining the core location. The Young's modules for MCF-7 cells decrease with the substrate stiffening, with values of 1050 Pa, 835 Pa, and 600 Pa measured on a Petri dish, Fmoc-FF hydrogel, and Fmoc-FF/chitosan hydrogel, respectively. A comparative analysis of the SICM results and the data obtained by atomic force microscopy was in good agreement, allowing for the use of a composite cell-substrate model (CoCS) to evaluate the 'soft substrate effect'. Using the CoCS model allowed us to conclude that the MCF-7 softening was due to the cells' mechanical properties variations due to cytoskeletal changes. This research provides immediate insights into changes in cell mechanical properties resulting from different soft scaffold substrates.
Collapse
Affiliation(s)
- Tatiana N. Tikhonova
- Department of Physics, M.V. Lomonosov Moscow State University, 1/2 Leninskie Gory, 119991 Moscow, Russia; (A.V.B.); (N.N.S.); (V.V.F.)
| | - Anastasia V. Barkovaya
- Department of Physics, M.V. Lomonosov Moscow State University, 1/2 Leninskie Gory, 119991 Moscow, Russia; (A.V.B.); (N.N.S.); (V.V.F.)
| | - Yuri M. Efremov
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (Y.M.E.); (P.S.T.)
| | - Vugara V. Mamed-Nabizade
- Laboratory of Biophysics, National University of Science and Technology MISIS, 4 Leninskiy prospekt, 119049 Moscow, Russia; (V.V.M.-N.); (V.S.K.); (P.V.G.); (A.S.E.)
| | - Vasilii S. Kolmogorov
- Laboratory of Biophysics, National University of Science and Technology MISIS, 4 Leninskiy prospekt, 119049 Moscow, Russia; (V.V.M.-N.); (V.S.K.); (P.V.G.); (A.S.E.)
- Department of Chemistry, M.V. Lomonosov Moscow State University, 1/2 Leninskie Gory, 119991 Moscow, Russia
| | - Peter S. Timashev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia; (Y.M.E.); (P.S.T.)
- Department of Chemistry, M.V. Lomonosov Moscow State University, 1/2 Leninskie Gory, 119991 Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| | - Nikolay N. Sysoev
- Department of Physics, M.V. Lomonosov Moscow State University, 1/2 Leninskie Gory, 119991 Moscow, Russia; (A.V.B.); (N.N.S.); (V.V.F.)
| | - Victor V. Fadeev
- Department of Physics, M.V. Lomonosov Moscow State University, 1/2 Leninskie Gory, 119991 Moscow, Russia; (A.V.B.); (N.N.S.); (V.V.F.)
| | - Petr V. Gorelkin
- Laboratory of Biophysics, National University of Science and Technology MISIS, 4 Leninskiy prospekt, 119049 Moscow, Russia; (V.V.M.-N.); (V.S.K.); (P.V.G.); (A.S.E.)
| | - Lihi Adler-Abramovich
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for the Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Alexander S. Erofeev
- Laboratory of Biophysics, National University of Science and Technology MISIS, 4 Leninskiy prospekt, 119049 Moscow, Russia; (V.V.M.-N.); (V.S.K.); (P.V.G.); (A.S.E.)
| | - Evgeny A. Shirshin
- Department of Physics, M.V. Lomonosov Moscow State University, 1/2 Leninskie Gory, 119991 Moscow, Russia; (A.V.B.); (N.N.S.); (V.V.F.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| |
Collapse
|
3
|
Gupta S, Bhattacharyya S. Footprints of scanning probe microscopy on halide perovskites. Chem Commun (Camb) 2024; 60:11685-11701. [PMID: 39295277 DOI: 10.1039/d4cc03658a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Scanning probe microscopy (SPM) and advanced atomic force microscopy (AFM++) have become pivotal for nanoscale elucidation of the structural, optoelectronic and photovoltaic properties of halide perovskite single crystals and polycrystalline films, both under ex situ and in situ conditions. These techniques reveal detailed information about film topography, compositional mapping, charge distribution, near-field electrical behaviors, cation-lattice interactions, ion dynamics, piezoelectric characteristics, mechanical durability, thermal conductivity, and magnetic properties of doped perovskite lattices. This article outlines the advancements in SPM techniques that deepen our understanding of the optoelectronic and photovoltaic performances of halide perovskites.
Collapse
Affiliation(s)
- Shresth Gupta
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur-741246, India.
| | - Sayan Bhattacharyya
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur-741246, India.
| |
Collapse
|
4
|
Montes AR, Barroso A, Wang W, O'Connell GD, Tepole AB, Mofrad MRK. Integrin mechanosensing relies on a pivot-clip mechanism to reinforce cell adhesion. Biophys J 2024; 123:2443-2454. [PMID: 38872310 PMCID: PMC11630637 DOI: 10.1016/j.bpj.2024.06.008] [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: 12/13/2023] [Revised: 05/01/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024] Open
Abstract
Cells intricately sense mechanical forces from their surroundings, driving biophysical and biochemical activities. This mechanosensing phenomenon occurs at the cell-matrix interface, where mechanical forces resulting from cellular motion, such as migration or matrix stretching, are exchanged through surface receptors, primarily integrins, and their corresponding matrix ligands. A pivotal player in this interaction is the α5β1 integrin and fibronectin (FN) bond, known for its role in establishing cell adhesion sites for migration. However, upregulation of the α5β1-FN bond is associated with uncontrolled cell metastasis. This bond operates through catch bond dynamics, wherein the bond lifetime paradoxically increases with greater force. The mechanism sustaining the characteristic catch bond dynamics of α5β1-FN remains unclear. Leveraging molecular dynamics simulations, our approach unveils a pivot-clip mechanism. Two key binding sites on FN, namely the synergy site and the RGD (Arg-Gly-Asp) motif, act as active points for structural changes in α5β1 integrin. Conformational adaptations at these sites are induced by a series of hydrogen bond formations and breaks at the synergy site. We disrupt these adaptations through a double mutation on FN, known to reduce cell adhesion. A whole-cell finite-element model is employed to elucidate how the synergy site may promote dynamic α5β1-FN binding, resisting cell contraction. In summary, our study integrates molecular- and cellular-level modeling to propose that FN's synergy site reinforces cell adhesion through enhanced binding dynamics and a mechanosensitive pivot-clip mechanism. This work sheds light on the interplay between mechanical forces and cell-matrix interactions, contributing to our understanding of cellular behaviors in physiological and pathological contexts.
Collapse
Affiliation(s)
- Andre R Montes
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, Berkeley, California
| | - Anahi Barroso
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, Berkeley, California
| | - Wei Wang
- Berkeley City College, Berkeley, California; Berkeley Biomechanics Laboratory, Department of Mechanical Engineering, University of California, Berkeley, Berkeley, California
| | - Grace D O'Connell
- Berkeley Biomechanics Laboratory, Department of Mechanical Engineering, University of California, Berkeley, Berkeley, California
| | - Adrian B Tepole
- Tepole Mechanics and Mechanobiology Laboratory, School of Mechanical Engineering, Purdue University, West Lafayette, Indiana.
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, Berkeley, California; Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, California.
| |
Collapse
|
5
|
Linke P, Munding N, Kimmle E, Kaufmann S, Hayashi K, Nakahata M, Takashima Y, Sano M, Bastmeyer M, Holstein T, Dietrich S, Müller‐Tidow C, Harada A, Ho AD, Tanaka M. Reversible Host-Guest Crosslinks in Supramolecular Hydrogels for On-Demand Mechanical Stimulation of Human Mesenchymal Stem Cells. Adv Healthc Mater 2024; 13:e2302607. [PMID: 38118064 PMCID: PMC11481031 DOI: 10.1002/adhm.202302607] [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: 08/09/2023] [Revised: 12/12/2023] [Indexed: 12/22/2023]
Abstract
Stem cells are regulated not only by biochemical signals but also by biophysical properties of extracellular matrix (ECM). The ECM is constantly monitored and remodeled because the fate of stem cells can be misdirected when the mechanical interaction between cells and ECM is imbalanced. A well-defined ECM model for bone marrow-derived human mesenchymal stem cells (hMSCs) based on supramolecular hydrogels containing reversible host-guest crosslinks is fabricated. The stiffness (Young's modulus E) of the hydrogels can be switched reversibly by altering the concentration of non-cytotoxic, free guest molecules dissolved in the culture medium. Fine-adjustment of substrate stiffness enables the authors to determine the critical stiffness level E* at which hMSCs turn the mechano-sensory machinery on or off. Next, the substrate stiffness across E* is switched and the dynamic adaptation characteristics such as morphology, traction force, and YAP/TAZ signaling of hMSCs are monitored. These data demonstrate the instantaneous switching of traction force, which is followed by YAP/TAZ signaling and morphological adaptation. Periodical switching of the substrate stiffness across E* proves that frequent applications of mechanical stimuli drastically suppress hMSC proliferation. Mechanical stimulation across E* level using dynamic hydrogels is a promising strategy for the on-demand control of hMSC transcription and proliferation.
Collapse
Affiliation(s)
- Philipp Linke
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
| | - Natalie Munding
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
| | - Esther Kimmle
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
| | - Stefan Kaufmann
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
| | - Kentaro Hayashi
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
| | - Masaki Nakahata
- Department of Macromolecular ScienceGraduate School of ScienceOsaka UniversityOsaka560‐0043Japan
| | - Yoshinori Takashima
- Department of Macromolecular ScienceGraduate School of ScienceOsaka UniversityOsaka560‐0043Japan
| | - Masaki Sano
- Institute of Natural SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Martin Bastmeyer
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
- Cell and NeurobiologyZoological InstituteKarlsruhe Institute of Technology76131KarlsruheGermany
- Institute for Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP)Karlsruhe Institute of Technology76334Eggenstein‐LeopoldshafenGermany
| | - Thomas Holstein
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
- Molecular Genetics and EvolutionCentre for Organismal StudiesHeidelberg University69221HeidelbergGermany
| | - Sascha Dietrich
- Department of Internal Medicine VHematology, Oncology, RheumatologyUniversity Hospital Heidelberg69120HeidelbergGermany
- Department of Haematology, Oncology, and Clinical ImmunologyUniversitätsklinikum Düsseldorf40225DüsseldorfGermany
| | - Carsten Müller‐Tidow
- Department of Internal Medicine VHematology, Oncology, RheumatologyUniversity Hospital Heidelberg69120HeidelbergGermany
| | - Akira Harada
- The Institute of Scientific and Industrial ResearchOsaka University8‐1 MihogaokaIbarakiOsaka567‐0047Japan
| | - Anthony D. Ho
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
- Department of Internal Medicine VHematology, Oncology, RheumatologyUniversity Hospital Heidelberg69120HeidelbergGermany
- Molecular Medicine Partnership Unit HeidelbergEMBL and Heidelberg University69120HeidelbergGermany
| | - Motomu Tanaka
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
| |
Collapse
|
6
|
Deir S, Mozhdehbakhsh Mofrad Y, Mashayekhan S, Shamloo A, Mansoori-Kermani A. Step-by-step fabrication of heart-on-chip systems as models for cardiac disease modeling and drug screening. Talanta 2024; 266:124901. [PMID: 37459786 DOI: 10.1016/j.talanta.2023.124901] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/23/2023] [Accepted: 07/01/2023] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases are caused by hereditary factors, environmental conditions, and medication-related issues. On the other hand, the cardiotoxicity of drugs should be thoroughly examined before entering the market. In this regard, heart-on-chip (HOC) systems have been developed as a more efficient and cost-effective solution than traditional methods, such as 2D cell culture and animal models. HOCs must replicate the biology, physiology, and pathology of human heart tissue to be considered a reliable platform for heart disease modeling and drug testing. Therefore, many efforts have been made to find the best methods to fabricate different parts of HOCs and to improve the bio-mimicry of the systems in the last decade. Beating HOCs with different platforms have been developed and techniques, such as fabricating pumpless HOCs, have been used to make HOCs more user-friendly systems. Recent HOC platforms have the ability to simultaneously induce and record electrophysiological stimuli. Additionally, systems including both heart and cancer tissue have been developed to investigate tissue-tissue interactions' effect on cardiac tissue response to cancer drugs. In this review, all steps needed to be considered to fabricate a HOC were introduced, including the choice of cellular resources, biomaterials, fabrication techniques, biomarkers, and corresponding biosensors. Moreover, the current HOCs used for modeling cardiac diseases and testing the drugs are discussed. We finally introduced some suggestions for fabricating relatively more user-friendly HOCs and facilitating the commercialization process.
Collapse
Affiliation(s)
- Sara Deir
- School of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Yasaman Mozhdehbakhsh Mofrad
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- School of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | - Amir Shamloo
- Nano-Bioengineering Lab, School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran.
| | | |
Collapse
|
7
|
Eroles M, Lopez-Alonso J, Ortega A, Boudier T, Gharzeddine K, Lafont F, Franz CM, Millet A, Valotteau C, Rico F. Coupled mechanical mapping and interference contrast microscopy reveal viscoelastic and adhesion hallmarks of monocyte differentiation into macrophages. NANOSCALE 2023. [PMID: 37378568 DOI: 10.1039/d3nr00757j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Monocytes activated by pro-inflammatory signals adhere to the vascular endothelium and migrate from the bloodstream to the tissue ultimately differentiating into macrophages. Cell mechanics and adhesion play a crucial role in macrophage functions during this inflammatory process. However, how monocytes change their adhesion and mechanical properties upon differentiation into macrophages is still not well understood. In this work, we used various tools to quantify the morphology, adhesion, and viscoelasticity of monocytes and differentiatted macrophages. Combination of atomic force microscopy (AFM) high resolution viscoelastic mapping with interference contrast microscopy (ICM) at the single-cell level revealed viscoelasticity and adhesion hallmarks during monocyte differentiation into macrophages. Quantitative holographic tomography imaging revealed a dramatic increase in cell volume and surface area during monocyte differentiation and the emergence of round and spread macrophage subpopulations. AFM viscoelastic mapping showed important stiffening (increase of the apparent Young's modulus, E0) and solidification (decrease of cell fluidity, β) on differentiated cells that correlated with increased adhesion area. These changes were enhanced in macrophages with a spread phenotype. Remarkably, when adhesion was perturbed, differentiated macrophages remained stiffer and more solid-like than monocytes, suggesting a permanent reorganization of the cytoskeleton. We speculate that the stiffer and more solid-like microvilli and lamellipodia might help macrophages to minimize energy dissipation during mechanosensitive activities. Thus, our results revealed viscoelastic and adhesion hallmarks of monocyte differentiation that may be important for biological function.
Collapse
Affiliation(s)
- Mar Eroles
- Aix-Marseille University, INSERM, CNRS, LAI, Turing Centre for Living Systems, Marseille, France.
| | - Javier Lopez-Alonso
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Alexandre Ortega
- Aix-Marseille University, INSERM, CNRS, LAI, Turing Centre for Living Systems, Marseille, France.
| | | | - Khaldoun Gharzeddine
- Univ.Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Team Mechanobiology, Immunity and Cancer, La Tronche, France
- Department of Hepatogastroenterology, Centre Hospitalier Universitaire de Grenoble Alpes, La Tronche, France
| | - Frank Lafont
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Clemens M Franz
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Arnaud Millet
- Univ.Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Team Mechanobiology, Immunity and Cancer, La Tronche, France
- Department of Hepatogastroenterology, Centre Hospitalier Universitaire de Grenoble Alpes, La Tronche, France
| | - Claire Valotteau
- Aix-Marseille University, INSERM, CNRS, LAI, Turing Centre for Living Systems, Marseille, France.
| | - Felix Rico
- Aix-Marseille University, INSERM, CNRS, LAI, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
8
|
Dow LP, Parmar T, Marchetti MC, Pruitt BL. Engineering tools for quantifying and manipulating forces in epithelia. BIOPHYSICS REVIEWS 2023; 4:021303. [PMID: 38510344 PMCID: PMC10903508 DOI: 10.1063/5.0142537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/20/2023] [Indexed: 03/22/2024]
Abstract
The integrity of epithelia is maintained within dynamic mechanical environments during tissue development and homeostasis. Understanding how epithelial cells mechanosignal and respond collectively or individually is critical to providing insight into developmental and (patho)physiological processes. Yet, inferring or mimicking mechanical forces and downstream mechanical signaling as they occur in epithelia presents unique challenges. A variety of in vitro approaches have been used to dissect the role of mechanics in regulating epithelia organization. Here, we review approaches and results from research into how epithelial cells communicate through mechanical cues to maintain tissue organization and integrity. We summarize the unique advantages and disadvantages of various reduced-order model systems to guide researchers in choosing appropriate experimental systems. These model systems include 3D, 2D, and 1D micromanipulation methods, single cell studies, and noninvasive force inference and measurement techniques. We also highlight a number of in silico biophysical models that are informed by in vitro and in vivo observations. Together, a combination of theoretical and experimental models will aid future experiment designs and provide predictive insight into mechanically driven behaviors of epithelial dynamics.
Collapse
Affiliation(s)
| | - Toshi Parmar
- Department of Physics, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | | | | |
Collapse
|
9
|
Melo-Fonseca F, Carvalho O, Gasik M, Miranda G, Silva FS. Mechanical stimulation devices for mechanobiology studies: a market, literature, and patents review. Biodes Manuf 2023. [DOI: 10.1007/s42242-023-00232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
AbstractSignificant advancements in various research and technological fields have contributed to remarkable findings on the physiological dynamics of the human body. To more closely mimic the complex physiological environment, research has moved from two-dimensional (2D) culture systems to more sophisticated three-dimensional (3D) dynamic cultures. Unlike bioreactors or microfluidic-based culture models, cells are typically seeded on polymeric substrates or incorporated into 3D constructs which are mechanically stimulated to investigate cell response to mechanical stresses, such as tensile or compressive. This review focuses on the working principles of mechanical stimulation devices currently available on the market or custom-built by research groups or protected by patents and highlights the main features still open to improvement. These are the features which could be focused on to perform, in the future, more reliable and accurate mechanobiology studies.
Graphic abstract
Collapse
|
10
|
Li W, Li J, Wu Y, Zhou T. A Novel Method in Identifying Pyroptosis and Apoptosis Based on the Double Resonator Piezoelectric Cytometry Technology. BIOSENSORS 2023; 13:356. [PMID: 36979568 PMCID: PMC10046136 DOI: 10.3390/bios13030356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 06/18/2023]
Abstract
In this study, a double resonator piezoelectric cytometry (DRPC) technology based on quartz crystal microbalance (QCM) was first employed to identify HeLa cell pyroptosis and apoptosis by monitoring cells' mechanical properties in a real-time and non-invasive manner. AT and BT cut quartz crystals with the same frequency and surface conditions were used concurrently to quantify the cells-exerted surface stress (ΔS). It is the first time that cells-exerted surface stress (ΔS) and cell viscoelasticity have been monitored simultaneously during pyroptosis and apoptosis. The results showed that HeLa pyroptotic cells exerted a tensile stress on quartz crystal along with an increase in the elastic modulus (G'), viscous modulus (G″), and a decrease of the loss tangent (G″/G'), whereas apoptotic cells exerted increasing compressive stress on quartz crystal along with a decrease in G', G″ and an increase in G″/G'. Furthermore, engineered GSDMD-/--DEVD- HeLa cells were used to investigate drug-induced disturbance and testify the mechanical responses during the processes of pyroptosis and non-pyroptosis. These findings demonstrated that the DRPC technology can serve as a precise cytomechanical sensor capable of identifying pyroptosis and apoptosis, providing a novel method in cell death detection and paving the road for pyroptosis and apoptosis related drug evaluation and screening.
Collapse
Affiliation(s)
- Wenwei Li
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
- Hunan Provincial Engineering Technology Research Center for Cell Mechanics and Function Analysis, Changsha 410128, China
| | - Jing Li
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yanyang Wu
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Tiean Zhou
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
- Hunan Provincial Engineering Technology Research Center for Cell Mechanics and Function Analysis, Changsha 410128, China
| |
Collapse
|
11
|
Light-driven biological actuators to probe the rheology of 3D microtissues. Nat Commun 2023; 14:717. [PMID: 36759504 PMCID: PMC9911700 DOI: 10.1038/s41467-023-36371-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
The mechanical properties of biological tissues are key to their physical integrity and function. Although external loading or biochemical treatments allow the estimation of these properties globally, it remains difficult to assess how such external stimuli compare with cell-generated contractions. Here we engineer microtissues composed of optogenetically-modified fibroblasts encapsulated within collagen. Using light to control the activity of RhoA, a major regulator of cellular contractility, we induce local contractions within microtissues, while monitoring microtissue stress and strain. We investigate the regulation of these local contractions and their spatio-temporal distribution. We demonstrate the potential of our technique for quantifying tissue elasticity and strain propagation, before examining the possibility of using light to create and map local anisotropies in mechanically heterogeneous microtissues. Altogether, our results open an avenue to guide the formation of tissues while non-destructively charting their rheology in real time, using their own constituting cells as internal actuators.
Collapse
|
12
|
Xing H, Huang Y, Kunkemoeller BH, Dahl PJ, Muraleetharan O, Malvankar NS, Murrell MP, Kyriakides TR. Dysregulation of TSP2-Rac1-WAVE2 axis in diabetic cells leads to cytoskeletal disorganization, increased cell stiffness, and dysfunction. Sci Rep 2022; 12:22474. [PMID: 36577792 PMCID: PMC9797577 DOI: 10.1038/s41598-022-26337-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Abstract
Fibroblasts are a major cell population that perform critical functions in the wound healing process. In response to injury, they proliferate and migrate into the wound space, engaging in extracellular matrix (ECM) production, remodeling, and contraction. However, there is limited knowledge of how fibroblast functions are altered in diabetes. To address this gap, several state-of-the-art microscopy techniques were employed to investigate morphology, migration, ECM production, 2D traction, 3D contraction, and cell stiffness. Analysis of cell-derived matrix (CDM) revealed that diabetic fibroblasts produce thickened and less porous ECM that hindered migration of normal fibroblasts. In addition, diabetic fibroblasts were found to lose spindle-like shape, migrate slower, generate less traction force, exert limited 3D contractility, and have increased cell stiffness. These changes were due, in part, to a decreased level of active Rac1 and a lack of co-localization between F-actin and Waskott-Aldrich syndrome protein family verprolin homologous protein 2 (WAVE2). Interestingly, deletion of thrombospondin-2 (TSP2) in diabetic fibroblasts rescued these phenotypes and restored normal levels of active Rac1 and WAVE2-F-actin co-localization. These results provide a comprehensive view of the extent of diabetic fibroblast dysfunction, highlighting the regulatory role of the TSP2-Rac1-WAVE2-actin axis, and describing a new function of TSP2 in regulating cytoskeleton organization.
Collapse
Affiliation(s)
- Hao Xing
- Department of Biomedical Engineering, Yale University, New Haven, USA.,Vascular Biology and Therapeutics Program, Yale University, New Haven, USA
| | - Yaqing Huang
- Department of Pathology, Yale University, New Haven, USA.,Vascular Biology and Therapeutics Program, Yale University, New Haven, USA
| | - Britta H Kunkemoeller
- Department of Pathology, Yale University, New Haven, USA.,Vascular Biology and Therapeutics Program, Yale University, New Haven, USA
| | - Peter J Dahl
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, USA.,Microbial Sciences Institute, Yale University, New Haven, USA
| | | | - Nikhil S Malvankar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, USA.,Microbial Sciences Institute, Yale University, New Haven, USA
| | - Michael P Murrell
- Department of Biomedical Engineering, Yale University, New Haven, USA.,Department of Physics, Yale University, New Haven, USA.,Systems Biology Institute, Yale University, New Haven, USA
| | - Themis R Kyriakides
- Department of Biomedical Engineering, Yale University, New Haven, USA. .,Department of Pathology, Yale University, New Haven, USA. .,Vascular Biology and Therapeutics Program, Yale University, New Haven, USA.
| |
Collapse
|
13
|
Molter CW, Muszynski EF, Tao Y, Trivedi T, Clouvel A, Ehrlicher AJ. Prostate cancer cells of increasing metastatic potential exhibit diverse contractile forces, cell stiffness, and motility in a microenvironment stiffness-dependent manner. Front Cell Dev Biol 2022; 10:932510. [PMID: 36200037 PMCID: PMC9527313 DOI: 10.3389/fcell.2022.932510] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
During metastasis, all cancer types must migrate through crowded multicellular environments. Simultaneously, cancers appear to change their biophysical properties. Indeed, cell softening and increased contractility are emerging as seemingly ubiquitous biomarkers of metastatic progression which may facilitate metastasis. Cell stiffness and contractility are also influenced by the microenvironment. Stiffer matrices resembling the tumor microenvironment cause metastatic cells to contract more strongly, further promoting contractile tumorigenic phenotypes. Prostate cancer (PCa), however, appears to deviate from these common cancer biophysics trends; aggressive metastatic PCa cells appear stiffer, rather than softer, to their lowly metastatic PCa counterparts. Although metastatic PCa cells have been reported to be more contractile than healthy cells, how cell contractility changes with increasing PCa metastatic potential has remained unknown. Here, we characterize the biophysical changes of PCa cells of various metastatic potential as a function of microenvironment stiffness. Using a panel of progressively increasing metastatic potential cell lines (22RV1, LNCaP, DU145, and PC3), we quantified their contractility using traction force microscopy (TFM), and measured their cortical stiffness using optical magnetic twisting cytometry (OMTC) and their motility using time-lapse microscopy. We found that PCa contractility, cell stiffness, and motility do not universally scale with metastatic potential. Rather, PCa cells of various metastatic efficiencies exhibit unique biophysical responses that are differentially influenced by substrate stiffness. Despite this biophysical diversity, this work concludes that mechanical microenvironment is a key determinant in the biophysical response of PCa with variable metastatic potentials. The mechanics-oriented focus and methodology of the study is unique and complementary to conventional biochemical and genetic strategies typically used to understand this disease, and thus may usher in new perspectives and approaches.
Collapse
Affiliation(s)
- Clayton W. Molter
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Eliana F. Muszynski
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Neuroscience, McGill University, Montreal, QC, Canada
| | - Yuanyuan Tao
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Electrical and Computer Engineering, McGill University, Montreal, QC, Canada
| | - Tanisha Trivedi
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Anna Clouvel
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Allen J. Ehrlicher
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| |
Collapse
|
14
|
Deviri D, Safran SA. Balance of osmotic pressures determines the nuclear-to-cytoplasmic volume ratio of the cell. Proc Natl Acad Sci U S A 2022; 119:e2118301119. [PMID: 35580183 PMCID: PMC9173802 DOI: 10.1073/pnas.2118301119] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 04/05/2022] [Indexed: 01/06/2023] Open
Abstract
The volume of the cell nucleus varies across cell types and species and is commonly thought to be determined by the size of the genome and degree of chromatin compaction. However, this notion has been challenged over the years by much experimental evidence. Here, we consider the physical condition of mechanical force balance as a determining condition of the nuclear volume and use quantitative, order-of-magnitude analysis to estimate the forces from different sources of nuclear and cytoplasmic pressure. Our estimates suggest that the dominant pressure within the nucleus and cytoplasm of nonstriated muscle cells originates from the osmotic pressure of proteins and RNA molecules that are localized to the nucleus or cytoplasm by out-of-equilibrium, active nucleocytoplasmic transport rather than from chromatin or its associated ions. This motivates us to formulate a physical model for the ratio of the cell and nuclear volumes in which osmotic pressures of localized proteins determine the relative volumes. In accordance with unexplained observations that are a century old, our model predicts that the ratio of the cell and nuclear volumes is a constant, robust to a wide variety of biochemical and biophysical manipulations, and is changed only if gene expression or nucleocytoplasmic transport is modulated.
Collapse
Affiliation(s)
- Dan Deviri
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovet 76100, Israel
| | - Samuel A. Safran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovet 76100, Israel
| |
Collapse
|
15
|
Baumann J, Sachs L, Otto O, Schoen I, Nestler P, Zaninetti C, Kenny M, Kranz R, von Eysmondt H, Rodriguez J, Schäffer TE, Nagy Z, Greinacher A, Palankar R, Bender M. Reduced platelet forces underlie impaired hemostasis in mouse models of MYH9-related disease. SCIENCE ADVANCES 2022; 8:eabn2627. [PMID: 35584211 PMCID: PMC9116608 DOI: 10.1126/sciadv.abn2627] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
MYH9-related disease patients with mutations in the contractile protein nonmuscle myosin heavy chain IIA display, among others, macrothrombocytopenia and a mild-to-moderate bleeding tendency. In this study, we used three mouse lines, each with one point mutation in the Myh9 gene at positions 702, 1424, or 1841, to investigate mechanisms underlying the increased bleeding risk. Agonist-induced activation of Myh9 mutant platelets was comparable to controls. However, myosin light chain phosphorylation after activation was reduced in mutant platelets, which displayed altered biophysical characteristics and generated lower adhesion, interaction, and traction forces. Treatment with tranexamic acid restored clot retraction in the presence of tPA and reduced bleeding. We verified our findings from the mutant mice with platelets from patients with the respective mutation. These data suggest that reduced platelet forces lead to an increased bleeding tendency in patients with MYH9-related disease, and treatment with tranexamic acid can improve the hemostatic function.
Collapse
Affiliation(s)
- Juliane Baumann
- Institute of Experimental Biomedicine—Chair I, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Laura Sachs
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Oliver Otto
- Zentrum für Innovationskompetenz—Humorale Immunreaktionen bei Kardiovaskulären Erkrankungen, University Greifswald, Greifswald, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e. V., Standort Greifswald, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Ingmar Schoen
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Peter Nestler
- Zentrum für Innovationskompetenz—Humorale Immunreaktionen bei Kardiovaskulären Erkrankungen, University Greifswald, Greifswald, Germany
| | - Carlo Zaninetti
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
- University of Pavia, Pavia, Italy
| | - Martin Kenny
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ruth Kranz
- Institute of Experimental Biomedicine—Chair I, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | | | - Johanna Rodriguez
- Institute of Applied Physics, University of Tübingen, Tübingen, Germany
| | | | - Zoltan Nagy
- Institute of Experimental Biomedicine—Chair I, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Andreas Greinacher
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Raghavendra Palankar
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
- Corresponding author. (M.B.); (R.P.)
| | - Markus Bender
- Institute of Experimental Biomedicine—Chair I, University Hospital and Rudolf Virchow Center, Würzburg, Germany
- Corresponding author. (M.B.); (R.P.)
| |
Collapse
|
16
|
Ahmed S, Johnson RT, Solanki R, Afewerki T, Wostear F, Warren DT. Using Polyacrylamide Hydrogels to Model Physiological Aortic Stiffness Reveals that Microtubules Are Critical Regulators of Isolated Smooth Muscle Cell Morphology and Contractility. Front Pharmacol 2022; 13:836710. [PMID: 35153800 PMCID: PMC8830533 DOI: 10.3389/fphar.2022.836710] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/12/2022] [Indexed: 12/04/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the medial layer of the aortic wall and normally exist in a quiescent, contractile phenotype where actomyosin-derived contractile forces maintain vascular tone. However, VSMCs are not terminally differentiated and can dedifferentiate into a proliferative, synthetic phenotype. Actomyosin force generation is essential for the function of both phenotypes. Whilst much is already known about the mechanisms of VSMC actomyosin force generation, existing assays are either low throughput and time consuming, or qualitative and inconsistent. In this study, we use polyacrylamide hydrogels, tuned to mimic the physiological stiffness of the aortic wall, in a VSMC contractility assay. Isolated VSMC area decreases following stimulation with the contractile agonists angiotensin II or carbachol. Importantly, the angiotensin II induced reduction in cell area correlated with increased traction stress generation. Inhibition of actomyosin activity using blebbistatin or Y-27632 prevented angiotensin II mediated changes in VSMC morphology, suggesting that changes in VSMC morphology and actomyosin activity are core components of the contractile response. Furthermore, we show that microtubule stability is an essential regulator of isolated VSMC contractility. Treatment with either colchicine or paclitaxel uncoupled the morphological and/or traction stress responses of angiotensin II stimulated VSMCs. Our findings support the tensegrity model of cellular mechanics and we demonstrate that microtubules act to balance actomyosin-derived traction stress generation and regulate the morphological responses of VSMCs.
Collapse
|
17
|
Boghdady CM, Kalashnikov N, Mok S, McCaffrey L, Moraes C. Revisiting tissue tensegrity: Biomaterial-based approaches to measure forces across length scales. APL Bioeng 2021; 5:041501. [PMID: 34632250 PMCID: PMC8487350 DOI: 10.1063/5.0046093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022] Open
Abstract
Cell-generated forces play a foundational role in tissue dynamics and homeostasis and are critically important in several biological processes, including cell migration, wound healing, morphogenesis, and cancer metastasis. Quantifying such forces in vivo is technically challenging and requires novel strategies that capture mechanical information across molecular, cellular, and tissue length scales, while allowing these studies to be performed in physiologically realistic biological models. Advanced biomaterials can be designed to non-destructively measure these stresses in vitro, and here, we review mechanical characterizations and force-sensing biomaterial-based technologies to provide insight into the mechanical nature of tissue processes. We specifically and uniquely focus on the use of these techniques to identify characteristics of cell and tissue "tensegrity:" the hierarchical and modular interplay between tension and compression that provide biological tissues with remarkable mechanical properties and behaviors. Based on these observed patterns, we highlight and discuss the emerging role of tensegrity at multiple length scales in tissue dynamics from homeostasis, to morphogenesis, to pathological dysfunction.
Collapse
Affiliation(s)
| | - Nikita Kalashnikov
- Department of Chemical Engineering, McGill University, Montréal, Québec H3A 0C5, Canada
| | - Stephanie Mok
- Department of Chemical Engineering, McGill University, Montréal, Québec H3A 0C5, Canada
| | | | | |
Collapse
|
18
|
Li J, Cao R, Wang Q, Shi H, Wu Y, Sun K, Liu X, Jiang H. Cadherin-11 promotes the mechanical strength of engineered elastic cartilage by enhancing extracellular matrix synthesis and microstructure. J Tissue Eng Regen Med 2021; 16:188-199. [PMID: 34837334 DOI: 10.1002/term.3271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/03/2021] [Accepted: 11/20/2021] [Indexed: 12/15/2022]
Abstract
Limitations of current treatments for auricular cartilage defects have prompted the field of auricular cartilage tissue engineering. To date, inducing the formation of cartilaginous constructs with biochemical and biomechanical properties of native tissue is the final aim. Through hematoxylin-eosin and immunohistochemistry staining, Cadherin-11(CDH11) was confirmed highly expressed in the auricular cartilage tissue and chondrocytes. In vitro, by knockdown and overexpression of CDH11 in chondrocytes, CDH11 was demonstrated to promote the expression of collagen type II (COL2A), elastin (ELN), aggrecan (ACAN), and cartilage oligomeric matrix protein (COMP). In addition, the CDH11 overexpressed chondrocytes promoted neo-cartilage formation and its biomechanical property by increasing the key transcription factor of chondrogenesis SOX9 expression and cartilage extracellular matrix (ECM) production. The young's modulus and yield stress of the neo-cartilage in CDH11 overexpression group were about 1.7 times (p = 0.0152) and 2 times (p = 0.0428) higher than those in control group, respectively. Then, the immunohistochemistry staining, qRT-PCR and western blot examination results showed that the expression of COL2A and ELN were significantly increased. Notably, the electron microscopy results showed that the collagen and elastic fibers of the neo-cartilage in CDH11-OV group arranged in bunches and were more uniform and compact compared to the control group. Furthermore, CDH11 promoted elastic fiber assembly by increasing lysyl oxidase (LOX), fibrillin-1 (FBN1) expression. Taken together, our results demonstrated that CDH11 improves the mechanical strength of tissue-engineered elastic cartilage by promoting ECM synthesis and elastic fiber assembly.
Collapse
Affiliation(s)
- Jia Li
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Rui Cao
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qian Wang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hang Shi
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yi Wu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kexin Sun
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xia Liu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Haiyue Jiang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
19
|
Sanchez JG, Espinosa FM, Miguez R, Garcia R. The viscoelasticity of adherent cells follows a single power-law with distinct local variations within a single cell and across cell lines. NANOSCALE 2021; 13:16339-16348. [PMID: 34581722 DOI: 10.1039/d1nr03894j] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
AFM-based force-distance curves are commonly used to characterize the nanomechanical properties of live cells. The transformation of these curves into nanomechanical properties requires the development of contact mechanics models. Spatially-resolved force-distance curves involving 1 to 2 μm deformations were obtained on HeLa and NIH 3T3 (fibroblast) cells. An elastic and two viscoelastic models were used to describe the experimental force-distance curves. The best agreement was obtained by applying a contact mechanics model that accounts for the geometry of the contact and the finite-thickness of the cell and assumes a single power-law dependence with time. Our findings show the shortcomings of elastic and semi-infinite viscoelastic models to characterize the mechanical response of a mammalian cell under micrometer-scale deformations. The parameters of the 3D power-law viscoelastic model, compressive modulus and fluidity exponent showed local variations within a single cell and across the two cell lines. The corresponding nanomechanical maps revealed structures that were not visible in the AFM topographic maps.
Collapse
Affiliation(s)
- Juan G Sanchez
- Instituto de Ciencia de Materiales de Madrid, CSIC, c/Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain.
| | - Francisco M Espinosa
- Instituto de Ciencia de Materiales de Madrid, CSIC, c/Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain.
| | - Ruben Miguez
- Instituto de Ciencia de Materiales de Madrid, CSIC, c/Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain.
| | - Ricardo Garcia
- Instituto de Ciencia de Materiales de Madrid, CSIC, c/Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain.
| |
Collapse
|
20
|
Lekka M, Gnanachandran K, Kubiak A, Zieliński T, Zemła J. Traction force microscopy - Measuring the forces exerted by cells. Micron 2021; 150:103138. [PMID: 34416532 DOI: 10.1016/j.micron.2021.103138] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/15/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
Cells generate mechanical forces (traction forces, TFs) while interacting with the extracellular matrix or neighbouring cells. Forces are generated by both cells and extracellular matrix (ECM) and transmitted within the cell-ECM or cell-cell contacts involving focal adhesions or adherens junctions. Within more than two decades, substantial progress has been achieved in techniques that measure TFs. One of the techniques is traction force microscopy (TFM). This review discusses the TFM and its advances in measuring TFs exerted by cells (single cells and multicellular systems) at cell-ECM and cell-cell junctional intracellular interfaces. The answers to how cells sense, adapt and respond to mechanical forces unravel their role in controlling and regulating cell behaviour in normal and pathological conditions.
Collapse
Affiliation(s)
- Małgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland.
| | | | - Andrzej Kubiak
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland
| | - Tomasz Zieliński
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland
| | - Joanna Zemła
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland
| |
Collapse
|
21
|
Rheinlaender J, Wirbel H, Schäffer TE. Spatial correlation of cell stiffness and traction forces in cancer cells measured with combined SICM and TFM. RSC Adv 2021; 11:13951-13956. [PMID: 35423943 PMCID: PMC8697701 DOI: 10.1039/d1ra01277k] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/06/2021] [Indexed: 12/31/2022] Open
Abstract
The mechanical properties of cancer cells at the single-cell and the subcellular level might be the key for answering long-standing questions in the diagnosis and treatment of cancer. However, the subcellular distribution of two main mechanical properties, cell stiffness and traction forces, has been investigated only rarely and qualitatively yet. Here, we present the first direct combination of scanning ion conductance microscopy (SICM) and traction force microscopy (TFM), which we used to identify a correlation between the local stiffness and the local traction force density in living cells. We found a correlation in normal breast epithelial cells, but no correlation in cancerous breast epithelial cells. This indicates that the interplay between cell stiffness and traction forces is altered in cancer cells as compared to healthy cells, which might give new insight in the research field of cancer cell mechanobiology.
Collapse
Affiliation(s)
- Johannes Rheinlaender
- Institute of Applied Physics, University of Tübingen Auf der Morgenstelle 10 72076 Tübingen Germany +49 7071 29 5093 +49 7071 29 76030
| | - Hannes Wirbel
- Institute of Applied Physics, University of Tübingen Auf der Morgenstelle 10 72076 Tübingen Germany +49 7071 29 5093 +49 7071 29 76030
| | - Tilman E Schäffer
- Institute of Applied Physics, University of Tübingen Auf der Morgenstelle 10 72076 Tübingen Germany +49 7071 29 5093 +49 7071 29 76030
| |
Collapse
|
22
|
Pfannenstill V, Barbotin A, Colin-York H, Fritzsche M. Quantitative Methodologies to Dissect Immune Cell Mechanobiology. Cells 2021; 10:851. [PMID: 33918573 PMCID: PMC8069647 DOI: 10.3390/cells10040851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 12/25/2022] Open
Abstract
Mechanobiology seeks to understand how cells integrate their biomechanics into their function and behavior. Unravelling the mechanisms underlying these mechanobiological processes is particularly important for immune cells in the context of the dynamic and complex tissue microenvironment. However, it remains largely unknown how cellular mechanical force generation and mechanical properties are regulated and integrated by immune cells, primarily due to a profound lack of technologies with sufficient sensitivity to quantify immune cell mechanics. In this review, we discuss the biological significance of mechanics for immune cells across length and time scales, and highlight several experimental methodologies for quantifying the mechanics of immune cells. Finally, we discuss the importance of quantifying the appropriate mechanical readout to accelerate insights into the mechanobiology of the immune response.
Collapse
Affiliation(s)
- Veronika Pfannenstill
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK; (V.P.); (A.B.)
| | - Aurélien Barbotin
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK; (V.P.); (A.B.)
| | - Huw Colin-York
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK; (V.P.); (A.B.)
| | - Marco Fritzsche
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK; (V.P.); (A.B.)
- Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0FA, UK
| |
Collapse
|
23
|
Fischer LS, Rangarajan S, Sadhanasatish T, Grashoff C. Molecular Force Measurement with Tension Sensors. Annu Rev Biophys 2021; 50:595-616. [PMID: 33710908 DOI: 10.1146/annurev-biophys-101920-064756] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ability of cells to generate mechanical forces, but also to sense, adapt to, and respond to mechanical signals, is crucial for many developmental, postnatal homeostatic, and pathophysiological processes. However, the molecular mechanisms underlying cellular mechanotransduction have remained elusive for many decades, as techniques to visualize and quantify molecular forces across individual proteins in cells were missing. The development of genetically encoded molecular tension sensors now allows the quantification of piconewton-scale forces that act upon distinct molecules in living cells and even whole organisms. In this review, we discuss the physical principles, advantages, and limitations of this increasingly popular method. By highlighting current examples from the literature, we demonstrate how molecular tension sensors can be utilized to obtain access to previously unappreciated biophysical parameters that define the propagation of mechanical forces on molecular scales. We discuss how the methodology can be further developed and provide a perspective on how the technique could be applied to uncover entirely novel aspects of mechanobiology in the future.
Collapse
Affiliation(s)
- Lisa S Fischer
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, Münster D-48149, Germany;
| | - Srishti Rangarajan
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, Münster D-48149, Germany;
| | - Tanmay Sadhanasatish
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, Münster D-48149, Germany;
| | - Carsten Grashoff
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, Münster D-48149, Germany;
| |
Collapse
|
24
|
Kar S, Katti DR, Katti KS. Evaluation of quasi-static and dynamic nanomechanical properties of bone-metastatic breast cancer cells using a nanoclay cancer testbed. Sci Rep 2021; 11:3096. [PMID: 33542384 PMCID: PMC7862348 DOI: 10.1038/s41598-021-82664-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/20/2021] [Indexed: 01/07/2023] Open
Abstract
In recent years, there has been increasing interest in investigating the mechanical properties of individual cells to delineate disease mechanisms. Reorganization of cytoskeleton facilitates the colonization of metastatic breast cancer at bone marrow space, leading to bone metastasis. Here, we report evaluation of mechanical properties of two breast cancer cells with different metastatic ability at the site of bone metastases, using quasi-static and dynamic nanoindentation methods. Our results showed that the significant reduction in elastic modulus along with increased liquid-like behavior of bone metastasized MCF-7 cells was induced by depolymerization and reorganization of F-actin to the adherens junctions, whereas bone metastasized MDA-MB-231 cells showed insignificant changes in elastic modulus and F-actin reorganization over time, compared to their respective as-received counterparts. Taken together, our data demonstrate evolution of breast cancer cell mechanics at bone metastases.
Collapse
Affiliation(s)
- Sumanta Kar
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering, North Dakota State University, Fargo, ND, 58108, USA
| | - Dinesh R Katti
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering, North Dakota State University, Fargo, ND, 58108, USA
| | - Kalpana S Katti
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering, North Dakota State University, Fargo, ND, 58108, USA.
| |
Collapse
|
25
|
Manipulation of Axonal Outgrowth via Exogenous Low Forces. Int J Mol Sci 2020; 21:ijms21218009. [PMID: 33126477 PMCID: PMC7663625 DOI: 10.3390/ijms21218009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022] Open
Abstract
Neurons are mechanosensitive cells. The role of mechanical force in the process of neurite initiation, elongation and sprouting; nerve fasciculation; and neuron maturation continues to attract considerable interest among scientists. Force is an endogenous signal that stimulates all these processes in vivo. The axon is able to sense force, generate force and, ultimately, transduce the force in a signal for growth. This opens up fascinating scenarios. How are forces generated and sensed in vivo? Which molecular mechanisms are responsible for this mechanotransduction signal? Can we exploit exogenously applied forces to mimic and control this process? How can these extremely low forces be generated in vivo in a non-invasive manner? Can these methodologies for force generation be used in regenerative therapies? This review addresses these questions, providing a general overview of current knowledge on the applications of exogenous forces to manipulate axonal outgrowth, with a special focus on forces whose magnitude is similar to those generated in vivo. We also review the principal methodologies for applying these forces, providing new inspiration and insights into the potential of this approach for future regenerative therapies.
Collapse
|
26
|
The mechanobiology of kidney podocytes in health and disease. Clin Sci (Lond) 2020; 134:1245-1253. [PMID: 32501496 DOI: 10.1042/cs20190764] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/15/2020] [Accepted: 05/22/2020] [Indexed: 01/01/2023]
Abstract
Chronic kidney disease (CKD) substantially reduces quality of life and leads to premature death for thousands of people each year. Dialysis and kidney organ transplants remain prevalent therapeutic avenues but carry significant medical, economic and social burden. Podocytes are responsible for blood filtration selectivity in the kidney, where they extend a network of foot processes (FPs) from their cell bodies which surround endothelial cells and interdigitate with those on neighbouring podocytes to form narrow slit diaphragms (SDs). During aging, some podocytes are lost naturally but accelerated podocyte loss is a hallmark of CKD. Insights into the origin of degenerative podocyte loss will help answer important questions about kidney function and lead to substantial health benefits. Here, approaches that uncover insights into podocyte mechanobiology are reviewed, both those that interrogate the biophysical properties of podocytes and how the external physical environment affects podocyte behaviour, and also those that interrogate the biophysical effects that podocytes exert on their surroundings.
Collapse
|
27
|
Efremov YM, Kotova SL, Timashev PS. Viscoelasticity in simple indentation-cycle experiments: a computational study. Sci Rep 2020; 10:13302. [PMID: 32764637 PMCID: PMC7413555 DOI: 10.1038/s41598-020-70361-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/23/2020] [Indexed: 11/17/2022] Open
Abstract
Instrumented indentation has become an indispensable tool for quantitative analysis of the mechanical properties of soft polymers and biological samples at different length scales. These types of samples are known for their prominent viscoelastic behavior, and attempts to calculate such properties from the indentation data are constantly made. The simplest indentation experiment presents a cycle of approach (deepening into the sample) and retraction of the indenter, with the output of the force and indentation depth as functions of time and a force versus indentation dependency (force curve). The linear viscoelastic theory based on the elastic–viscoelastic correspondence principle might predict the shape of force curves based on the experimental conditions and underlying relaxation function of the sample. Here, we conducted a computational analysis based on this theory and studied how the force curves were affected by the indenter geometry, type of indentation (triangular or sinusoidal ramp), and the relaxation functions. The relaxation functions of both traditional and fractional viscoelastic models were considered. The curves obtained from the analytical solutions, numerical algorithm and finite element simulations matched each other well. Common trends for the curve-related parameters (apparent Young’s modulus, normalized hysteresis area, and curve exponent) were revealed. Importantly, the apparent Young’s modulus, obtained by fitting the approach curve to the elastic model, demonstrated a direct relation to the relaxation function for all the tested cases. The study will help researchers to verify which model is more appropriate for the sample description without extensive calculations from the basic curve parameters and their dependency on the indentation rate.
Collapse
Affiliation(s)
- Yu M Efremov
- Institute for Regenerative Medicine, Sechenov University, 8 Trubetskaya St., Moscow, 119991, Russia.
| | - S L Kotova
- Institute for Regenerative Medicine, Sechenov University, 8 Trubetskaya St., Moscow, 119991, Russia.,N.N. Semenov Institute of Chemical Physics, 4 Kosygin St., Moscow, 119991, Russia
| | - P S Timashev
- Institute for Regenerative Medicine, Sechenov University, 8 Trubetskaya St., Moscow, 119991, Russia.,N.N. Semenov Institute of Chemical Physics, 4 Kosygin St., Moscow, 119991, Russia.,Institute of Photon Technologies of Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences, Pionerskaya 2, Troitsk, Moscow, 108840, Russia.,Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, Moscow, 119991, Russia
| |
Collapse
|
28
|
Cell response to substrate rigidity is regulated by active and passive cytoskeletal stress. Proc Natl Acad Sci U S A 2020; 117:12817-12825. [PMID: 32444491 DOI: 10.1073/pnas.1917555117] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Morphogenesis, tumor formation, and wound healing are regulated by tissue rigidity. Focal adhesion behavior is locally regulated by stiffness; however, how cells globally adapt, detect, and respond to rigidity remains unknown. Here, we studied the interplay between the rheological properties of the cytoskeleton and matrix rigidity. We seeded fibroblasts onto flexible microfabricated pillar arrays with varying stiffness and simultaneously measured the cytoskeleton organization, traction forces, and cell-rigidity responses at both the adhesion and cell scale. Cells adopted a rigidity-dependent phenotype whereby the actin cytoskeleton polarized on stiff substrates but not on soft. We further showed a crucial role of active and passive cross-linkers in rigidity-sensing responses. By reducing myosin II activity or knocking down α-actinin, we found that both promoted cell polarization on soft substrates, whereas α-actinin overexpression prevented polarization on stiff substrates. Atomic force microscopy indentation experiments showed that this polarization response correlated with cell stiffness, whereby cell stiffness decreased when active or passive cross-linking was reduced and softer cells polarized on softer matrices. Theoretical modeling of the actin network as an active gel suggests that adaptation to matrix rigidity is controlled by internal mechanical properties of the cytoskeleton and puts forward a universal scaling between nematic order of the actin cytoskeleton and the substrate-to-cell elastic modulus ratio. Altogether, our study demonstrates the implication of cell-scale mechanosensing through the internal stress within the actomyosin cytoskeleton and its coupling with local rigidity sensing at focal adhesions in the regulation of cell shape changes and polarity.
Collapse
|
29
|
Blair CA, Pruitt BL. Mechanobiology Assays with Applications in Cardiomyocyte Biology and Cardiotoxicity. Adv Healthc Mater 2020; 9:e1901656. [PMID: 32270928 PMCID: PMC7480481 DOI: 10.1002/adhm.201901656] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/19/2022]
Abstract
Cardiomyocytes are the motor units that drive the contraction and relaxation of the heart. Traditionally, testing of drugs for cardiotoxic effects has relied on primary cardiomyocytes from animal models and focused on short-term, electrophysiological, and arrhythmogenic effects. However, primary cardiomyocytes present challenges arising from their limited viability in culture, and tissue from animal models suffers from a mismatch in their physiology to that of human heart muscle. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can address these challenges. They also offer the potential to study not only electrophysiological effects but also changes in cardiomyocyte contractile and mechanical function in response to cardiotoxic drugs. With growing recognition of the long-term cardiotoxic effects of some drugs on subcellular structure and function, there is increasing interest in using hiPSC-CMs for in vitro cardiotoxicity studies. This review provides a brief overview of techniques that can be used to quantify changes in the active force that cardiomyocytes generate and variations in their inherent stiffness in response to cardiotoxic drugs. It concludes by discussing the application of these tools in understanding how cardiotoxic drugs directly impact the mechanobiology of cardiomyocytes and how cardiomyocytes sense and respond to mechanical load at the cellular level.
Collapse
Affiliation(s)
- Cheavar A. Blair
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Beth L. Pruitt
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
30
|
Rianna C, Radmacher M, Kumar S. Direct evidence that tumor cells soften when navigating confined spaces. Mol Biol Cell 2020; 31:1726-1734. [PMID: 31995446 PMCID: PMC7521845 DOI: 10.1091/mbc.e19-10-0588] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The mechanical properties of cells strongly regulate many physiological and pathological processes. For example, in cancer, invasive and metastatic tumor cells have often been reported to be softer than nontumor cells, raising speculation that cancer cells might adaptively soften to facilitate migration through narrow tissue spaces. Despite growing interest in targeting cell softening to impede invasion and metastasis, it remains to be directly demonstrated that tumor cells soften as they migrate through confined spaces. Here, we address this open question by combining topographically patterned substrates with atomic force microscopy (AFM). Using a polydimethylsiloxane open-roof microdevice featuring tapered, fibronectin-coated channels, we followed the migration of U2OS cells through various stages of confinement while simultaneously performing AFM indentation. As cells progress from unconfined migration to fully confined migration, cells soften and exclude Yes-associated protein from the nucleus. Superresolution imaging reveals that confinement induces remodeling of actomyosin stress fiber architecture. Companion studies with flat one-dimensional microlines indicate that the changes in cytoarchitecture and mechanics are intrinsically driven by topographical confinement rather than changes in cellular aspect ratio. Our studies represent among the most direct evidence to date that tumor cells soften during confined migration and support cell softening as a mechanoadaptive mechanism during invasion.
Collapse
Affiliation(s)
- Carmela Rianna
- Institute of Biophysics, University of Bremen, 28359 Bremen, Germany.,Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720
| | - Manfred Radmacher
- Institute of Biophysics, University of Bremen, 28359 Bremen, Germany
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720
| |
Collapse
|
31
|
Efremov YM, Okajima T, Raman A. Measuring viscoelasticity of soft biological samples using atomic force microscopy. SOFT MATTER 2020; 16:64-81. [PMID: 31720656 DOI: 10.1039/c9sm01020c] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mechanical properties play important roles at different scales in biology. At the level of a single cell, the mechanical properties mediate mechanosensing and mechanotransduction, while at the tissue and organ levels, changes in mechanical properties are closely connected to disease and physiological processes. Over the past three decades, atomic force microscopy (AFM) has become one of the most widely used tools in the mechanical characterization of soft samples, ranging from molecules, cell organoids and cells to whole tissue. AFM methods can be used to quantify both elastic and viscoelastic properties, and significant recent developments in the latter have been enabled by the introduction of new techniques and models for data analysis. Here, we review AFM techniques developed in recent years for examining the viscoelastic properties of cells and soft gels, describe the main steps in typical data acquisition and analysis protocols, and discuss relevant viscoelastic models and how these have been used to characterize the specific features of cellular and other biological samples. We also discuss recent trends and potential directions for this field.
Collapse
Affiliation(s)
- Yuri M Efremov
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, USA. and Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana, USA and Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Takaharu Okajima
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Arvind Raman
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, USA. and Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
32
|
Halonen HT, Hyttinen JA, Ihalainen TO. Mechanical impact stimulation platform tailored for high-resolution light microscopy. HEALTH AND TECHNOLOGY 2020. [DOI: 10.1007/s12553-019-00382-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
AbstractHigh frequency (HF) mechanical vibration has been used in vitro to study the cellular response to mechanical stimulation and induce stem cell differentiation. However, detailed understanding of the effect of the mechanical cues on cellular physiology is lacking. To meet this limitation, we have designed a system, which enables monitoring of living cells by high-resolution light microscopy during mechanical stimulation by HF vibration or mechanical impacts. The system consists of a commercial speaker, and a 3D printed sample vehicle and frame. The speaker moves the sample in the horizontal plane, allowing simultaneous microscopy. The HF vibration (30–200 Hz) performances of two vehicles made of polymer and aluminum were characterized with accelerometer. The mechanical impacts were characterized by measuring the acceleration of the aluminum vehicle and by time lapse imaging. The lighter polymer vehicle produced higher HF vibration magnitudes at 30–50 Hz frequencies than the aluminum vehicle. However, the aluminum vehicle performed better at higher frequencies (60–70 Hz, 90–100 Hz, 150 Hz). Compatibility of the system in live cell experiments was investigated with epithelial cells (MDCKII, expressing Emerald-Occludin) and HF (0.56Gpeak,30 Hz and 60 Hz) vibration. Our findings indicated that our system is compatible with high-resolution live cell microscopy. Furthermore, the epithelial cells were remarkable stable under mechanical vibration stimulation. To conclude, we have designed an inexpensive tool for the studies of cellular biophysics, which combines versatile in vivo like mechanical stimuli with live cell imaging, showing a great potential for several cellular applications.
Collapse
|
33
|
Neurite regrowth stimulation by a red-light spot focused on the neuronal cell soma following blue light-induced retraction. Sci Rep 2019; 9:18210. [PMID: 31796850 PMCID: PMC6890775 DOI: 10.1038/s41598-019-54687-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 11/18/2019] [Indexed: 11/13/2022] Open
Abstract
The interaction of light with biological tissues has been considered for various therapeutic applications. Light-induced neurite growth has the potential to be a clinically useful technique for neuron repair. However, most previous studies used either a large illumination area to accelerate overall neurite growth or employed a light spot to guide a growing neurite. It is not clear if optical stimulation can induce the regrowth of a retracted neurite. In the present work, we used blue light (wavelength: 473 nm) to cause neurite retraction, and we proved that using a red-light (wavelength: 650 nm) spot to illuminate the soma near the junction of the retracted neurite could induce neurite regrowth. As a comparison, we found that green light (wavelength 550 nm) had a 62% probability of inducing neurite regrowth, while red light had a 75% probability of inducing neurite regrowth at the same power level. Furthermore, the neurite regrowth length induced by red light was increased by the pre-treatment with inhibitors of myosin functions. We also observed actin propagation from the soma to the tip of the re-growing neurite following red-light stimulation of the soma. The red light-induced extension and regrowth were abrogated in the calcium-free medium. These results suggest that illumination with a red-light spot on the soma may trigger the regrowth of a neurite after the retraction caused by blue-light illumination.
Collapse
|
34
|
Chighizola M, Dini T, Lenardi C, Milani P, Podestà A, Schulte C. Mechanotransduction in neuronal cell development and functioning. Biophys Rev 2019; 11:701-720. [PMID: 31617079 PMCID: PMC6815321 DOI: 10.1007/s12551-019-00587-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/29/2019] [Indexed: 12/21/2022] Open
Abstract
Although many details remain still elusive, it became increasingly evident in recent years that mechanosensing of microenvironmental biophysical cues and subsequent mechanotransduction are strongly involved in the regulation of neuronal cell development and functioning. This review gives an overview about the current understanding of brain and neuronal cell mechanobiology and how it impacts on neurogenesis, neuronal migration, differentiation, and maturation. We will focus particularly on the events in the cell/microenvironment interface and the decisive extracellular matrix (ECM) parameters (i.e. rigidity and nanometric spatial organisation of adhesion sites) that modulate integrin adhesion complex-based mechanosensing and mechanotransductive signalling. It will also be outlined how biomaterial approaches mimicking essential ECM features help to understand these processes and how they can be used to control and guide neuronal cell behaviour by providing appropriate biophysical cues. In addition, principal biophysical methods will be highlighted that have been crucial for the study of neuronal mechanobiology.
Collapse
Affiliation(s)
- Matteo Chighizola
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Tania Dini
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Cristina Lenardi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Paolo Milani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Alessandro Podestà
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy
| | - Carsten Schulte
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics ``Aldo Pontremoli'', Università degli Studi di Milano, via Celoria 16, 20133, Milan, Italy.
| |
Collapse
|
35
|
Liu L, Wei Y, Liu J, Wang K, Zhang J, Zhang P, Zhou Y, Li B. Spatial high resolution of actin filament organization by PeakForce atomic force microscopy. Cell Prolif 2019; 53:e12670. [PMID: 31568631 PMCID: PMC6985672 DOI: 10.1111/cpr.12670] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/08/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives To investigate the heterogeneous feature of actin filaments (ACFs) associated with the cellular membrane in HeLa and HCT‐116 cells at the nanoscale level. Materials and Methods Fluorescence microscopy coupled with atomic force microscopy (AFM) was used to identify and characterize ACFs of cells. The distribution of ACFs was detected by Fluor‐488‐phalloidin–labelled actin. The morphology of the ACFs was probed by AFM images. The spatial correlation of the microvilli and ACFs was explored with different forces of AFM loading on cells. Results Intricate but ordered structures of the actin cytoskeletons associated with cellular membrane were characterized and revealed. Two different layers of ACFs with distinct structural organizations were directly observed in HCT‐116 and HeLa cells. Bundle‐shaped ACFs protruding the cellular membrane forming the microvilli, and the network ACFs underneath the cellular membrane were resolved with high resolution under near‐physiological conditions. Approximately 14 nm lateral resolution was achieved when imaging single ACF beneath the cellular membrane. On the basis of the observed spatial distribution of the ultrastructure of the ACF organization, a model for this organization of ACFs was proposed. Conclusions We revealed the two layers of the ACF organization in Hela and HCT‐116 cells. The resolved heterogeneous structures at the nanoscale level provide a spatial view of the ACFs, which would contribute to the understanding of the essential biological functions of the actin cytoskeleton.
Collapse
Affiliation(s)
- Lin Liu
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuhui Wei
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| | - Jingyuan Liu
- Fourth Military Medical University, Xi'an, China
| | - Kaizhe Wang
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jinjin Zhang
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| | - Ping Zhang
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yi Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Li
- Division of Physical Biology & Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
36
|
Zhang T, Day JH, Su X, Guadarrama AG, Sandbo NK, Esnault S, Denlinger LC, Berthier E, Theberge AB. Investigating Fibroblast-Induced Collagen Gel Contraction Using a Dynamic Microscale Platform. Front Bioeng Biotechnol 2019; 7:196. [PMID: 31475142 PMCID: PMC6702460 DOI: 10.3389/fbioe.2019.00196] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/30/2019] [Indexed: 11/14/2022] Open
Abstract
Mechanical forces have long been recognized as fundamental drivers in biological processes, such as embryogenesis, tissue formation and disease regulation. The collagen gel contraction (CGC) assay has served as a classic tool in the field of mechanobiology to study cell-induced contraction of extracellular matrix (ECM), which plays an important role in inflammation and wound healing. In a conventional CGC assay, cell-laden collagen is loaded into a cell culture vessel (typically a well plate) and forms a disk-shaped gel adhering to the bottom of the vessel. The decrement in diameter or surface area of the gel is used as a parameter to quantify the degree of cell contractility. In this study, we developed a microscale CGC assay with an engineered well plate insert that uses surface tension forces to load and manipulate small volumes (14 μL) of cell-laden collagen. The system is easily operated with two pipetting steps and the microscale device moves dynamically as a result of cellular forces. We used a straightforward one-dimensional measurement as the gel contraction readout. We adapted a conventional lung fibroblast CGC assay to demonstrate the functionality of the device, observing significantly more gel contraction when human lung fibroblasts were cultured in serum-containing media vs. serum-free media (p ≤ 0.05). We further cocultured eosinophils and fibroblasts in the system, two important cellular components that lead to fibrosis in asthma, and observed that soluble factors from eosinophils significantly increase fibroblast-mediated gel contraction (p ≤ 0.01). Our microscale CGC device provides a new method for studying downstream ECM effects of intercellular cross talk using 7- to 35-fold less cell-laden gel than traditional CGC assays.
Collapse
Affiliation(s)
- Tianzi Zhang
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - John H Day
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Xiaojing Su
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Arthur G Guadarrama
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Nathan K Sandbo
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Stephane Esnault
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Loren C Denlinger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Ashleigh B Theberge
- Department of Chemistry, University of Washington, Seattle, WA, United States.,Department of Urology, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
37
|
Huang J, Lin F, Xiong C. Mechanical characterization of single cells based on microfluidic techniques. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.07.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
38
|
Efremov YM, Shpichka AI, Kotova SL, Timashev PS. Viscoelastic mapping of cells based on fast force volume and PeakForce Tapping. SOFT MATTER 2019; 15:5455-5463. [PMID: 31231747 DOI: 10.1039/c9sm00711c] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Development of fast force volume (FFV), PeakForce Tapping (PFT), and related AFM techniques allow fast acquisition and mapping of a sample's mechanical properties. The methods are well-suited for studying soft biological samples like living cells in a liquid environment. However, the question remains how the measured mechanical properties are related to those acquired with the classical force volume (FV) technique conducted at low indentation rates. The difference is coming mostly from the pronounced viscoelastic behavior of cells, making apparent elastic parameters depending on the probing rate. Here, the viscoelastic analysis was applied directly to the force curves acquired with force volume or PeakForce Tapping by their post-processing based on the Ting's model. Maps from classical force volume, FFV and PFT obtained using special PFT cantilevers and cantilevers modified with microspheres were compared here. With the correct viscoelastic model, which was found to be the power-law rheology model, all the techniques have provided self-consistent results. The techniques were further modified for the mapping of the viscoelastic model-independent complex Young's modulus.
Collapse
Affiliation(s)
- Yu M Efremov
- Institute for Regenerative Medicine, Sechenov University, 8 Trubetskaya St., Moscow, 119991, Russia.
| | - A I Shpichka
- Institute for Regenerative Medicine, Sechenov University, 8 Trubetskaya St., Moscow, 119991, Russia.
| | - S L Kotova
- Institute for Regenerative Medicine, Sechenov University, 8 Trubetskaya St., Moscow, 119991, Russia. and N.N. Semenov Institute of Chemical Physics, 4 Kosygin St., Moscow, 119991, Russia
| | - P S Timashev
- Institute for Regenerative Medicine, Sechenov University, 8 Trubetskaya St., Moscow, 119991, Russia. and N.N. Semenov Institute of Chemical Physics, 4 Kosygin St., Moscow, 119991, Russia and Institute of Photonic Technologies, Research center "Crystallography and Photonics", 2 Pionerskaya St., Troitsk, Moscow 108840, Russia
| |
Collapse
|