1
|
Bergal HT, Kinoshita K, Wong WP. High-Throughput Centrifuge Force Microscopy Reveals Dynamic Immune-Cell Avidity at the Single-Cell Level. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640408. [PMID: 40060606 PMCID: PMC11888394 DOI: 10.1101/2025.02.27.640408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Cell-cell binding, mediated by the physical interactions of receptors and their ligands, plays a fundamental role in immune processes such as immune surveillance and T-cell activation. However, current approaches for measuring cell avidity often lack either throughput or quantitative precision. Here, we introduce a high-throughput approach for quantifying cell binding lifetimes and strength using a centrifuge force microscope (CFM)-a compact microscope operating within a standard benchtop centrifuge. The CFM enables live monitoring of single-cell interactions under force, conducting thousands of force experiments in parallel. To facilitate the real-time study of live cell interactions, we developed a next-generation CFM with multichannel fluorescence imaging capabilities. This system accommodates measurements in two modes: cell-protein binding and cell-cell avidity assays. Using this system, we investigated immune-cell binding mediated by Bispecific T-cell Engager (BiTE) molecules, a novel immunotherapy designed to enhance immune-cell targeting of cancer cells. In cell-protein assays, we quantified T- and B-cell unbinding from BiTE-functionalized surfaces, revealing receptor-specific relationships between ligand concentration and binding strength. In cell-cell assays, we examined BiTE-mediated binding of T-cells to Nalm6 B-cells, a precursor leukemia cell line, uncovering a strong, time-dependent increase in BiTE-mediated immune-cell avidity. By integrating high-throughput and quantitative single-cell force analysis, the CFM provides new insights into the dynamic nature of immunological interactions under force, with broad implications for immunotherapy and cellular mechanics.
Collapse
Affiliation(s)
- Hans T Bergal
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School
| | - Koji Kinoshita
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School
| | - Wesley P Wong
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School
- Department of Pediatrics, Harvard Medical School
- Wyss Institute for Biologically Inspired Engineering, Harvard University
| |
Collapse
|
2
|
Ho AD, Tanaka M. Novel techniques to quantitatively assess age-dependent alterations in biophysical properties of HSPCs and bone marrow niche. Exp Hematol 2025; 142:104686. [PMID: 39613289 DOI: 10.1016/j.exphem.2024.104686] [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: 09/09/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 12/01/2024]
Abstract
The present knowledge on hematopoietic stem and progenitor cell (HSPC) biology and aging is based largely on studies in mouse models. Although mouse models are invaluable, they are not without limitations for defining how physical properties of HSPCs and their niche change with age. The bone marrow (BM) niche is a complex, interactive environment with multiple cell types. The structure and organization of the BM niche, especially the extracellular matrix (ECM), change with age. Provided with recent advances in quantitative analytical techniques and in vitro niche models, we have developed novel tools to quantitatively assess the impact of specific biochemical and physical cues on homing, adhesion, and migration of HSPCs. Recent developments in in vitro niche models have also provided new insights into the interactions between HSPCs and their niche, particularly the role of matrix stiffness. Further research is needed to integrate physical biomarkers into comprehensive mathematical models of age-dependent HSPC-niche interactions. The key is to use mouse models in conjunction with direct analyses in in vitro niche models to achieve a more comprehensive understanding of age-dependent alterations in niche function and regulation.
Collapse
Affiliation(s)
- Anthony D Ho
- Department of Medicine V, Medical Center, Heidelberg University, Heidelberg, Germany; Center for Integrative Medicine and Physics, Institute for Advances Study, Kyoto University, Kyoto, Japan.
| | - Motomu Tanaka
- Center for Integrative Medicine and Physics, Institute for Advances Study, Kyoto University, Kyoto, Japan; Physical Chemistry of Biosystems, Institute of Physical Chemistry, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
3
|
Chen CW, Chavez JB, Kumar R, Go VA, Pant A, Jain A, Polusani SR, Hart MJ, Robinson RD, Gaczynska M, Osmulski P, Kirma NB, Nicholson BJ. Hypersensitive intercellular responses of endometrial stromal cells drive invasion in endometriosis. eLife 2024; 13:e94778. [PMID: 39660704 PMCID: PMC11729374 DOI: 10.7554/elife.94778] [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: 11/22/2023] [Accepted: 12/10/2024] [Indexed: 12/12/2024] Open
Abstract
Endometriosis is a debilitating disease affecting 190 million women worldwide and the greatest single contributor to infertility. The most broadly accepted etiology is that uterine endometrial cells retrogradely enter the peritoneum during menses, and implant and form invasive lesions in a process analogous to cancer metastasis. However, over 90% of women suffer retrograde menstruation, but only 10% develop endometriosis, and debate continues as to whether the underlying defect is endometrial or peritoneal. Processes implicated in invasion include: enhanced motility; adhesion to, and formation of gap junctions with, the target tissue. Endometrial stromal (ESCs) from 22 endometriosis patients at different disease stages show much greater invasiveness across mesothelial (or endothelial) monolayers than ESCs from 22 control subjects, which is further enhanced by the presence of EECs. This is due to the enhanced responsiveness of endometriosis ESCs to the mesothelium, which induces migration and gap junction coupling. ESC-PMC gap junction coupling is shown to be required for invasion, while coupling between PMCs enhances mesothelial barrier breakdown.
Collapse
Affiliation(s)
- Chun-Wei Chen
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| | - Jeffery B Chavez
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| | - Ritikaa Kumar
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| | - Virginia Arlene Go
- Department of Obstetrics and Gynecology, UT Health San AntonioSan AntonioUnited States
| | - Ahvani Pant
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| | - Anushka Jain
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| | - Srikanth R Polusani
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| | - Matthew J Hart
- Center for Innovative Drug Discovery, UT Health San AntonioSan AntonioUnited States
| | - Randal D Robinson
- Department of Obstetrics and Gynecology, UT Health San AntonioSan AntonioUnited States
| | - Maria Gaczynska
- Department of Molecular Medicine, UT Health San AntonioSan AntonioUnited States
| | - Pawel Osmulski
- Department of Molecular Medicine, UT Health San AntonioSan AntonioUnited States
| | - Nameer B Kirma
- Department of Molecular Medicine, UT Health San AntonioSan AntonioUnited States
| | - Bruce J Nicholson
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| |
Collapse
|
4
|
Teixeira Polez R, Huynh N, Pridgeon CS, Valle-Delgado JJ, Harjumäki R, Österberg M. Insights into spheroids formation in cellulose nanofibrils and Matrigel hydrogels using AFM-based techniques. Mater Today Bio 2024; 26:101065. [PMID: 38706731 PMCID: PMC11066555 DOI: 10.1016/j.mtbio.2024.101065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/30/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024] Open
Abstract
The recent FDA decision to eliminate animal testing requirements emphasises the role of cell models, such as spheroids, as regulatory test alternatives for investigations of cellular behaviour, drug responses, and disease modelling. The influence of environment on spheroid formation are incompletely understood, leading to uncertainty in matrix selection for scaffold-based 3D culture. This study uses atomic force microscopy-based techniques to quantify cell adhesion to Matrigel and cellulose nanofibrils (CNF), and cell-cell adhesion forces, and their role in spheroid formation of hepatocellular carcinoma (HepG2) and induced pluripotent stem cells (iPS(IMR90)-4). Results showed different cell behaviour in CNF and Matrigel cultures. Both cell lines formed compact spheroids in CNF but loose cell aggregates in Matrigel. Interestingly, the type of cell adhesion protein, and not the bond strength, appeared to be a key factor in the formation of compact spheroids. The gene expression of E- and N-cadherins, proteins on cell membrane responsible for cell-cell interactions, was increased in CNF culture, leading to formation of compact spheroids while Matrigel culture induced integrin-laminin binding and downregulated E-cadherin expression, resulting in looser cell aggregates. These findings enhance our understanding of cell-biomaterial interactions in 3D cultures and offer insights for improved 3D cell models, culture biomaterials, and applications in drug research.
Collapse
Affiliation(s)
- Roberta Teixeira Polez
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Ngoc Huynh
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Chris S. Pridgeon
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Juan José Valle-Delgado
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Riina Harjumäki
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Monika Österberg
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| |
Collapse
|
5
|
Zhang S, Felthaus O, Prantl L, Ma N, Machatschek R. Continuous protein-density gradients: A new approach to correlate physical cues with cell response. PNAS NEXUS 2024; 3:pgae202. [PMID: 38840799 PMCID: PMC11152205 DOI: 10.1093/pnasnexus/pgae202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/07/2024] [Indexed: 06/07/2024]
Abstract
To assess cellular behavior within heterogeneous tissues, such as bone, skin, and nerves, scaffolds with biophysical gradients are required to adequately replicate the in vivo interaction between cells and their native microenvironment. In this study, we introduce a strategy for depositing ultrathin films comprised of laminin-111 with precisely controlled biophysical gradients onto planar substrates using the Langmuir-Blodgett (LB) technique. The gradient is created by controlled desynchronization of the barrier compression and substrate withdrawal speed during the LB deposition process. Characterization of the films was performed using techniques such as atomic force microscopy and confocal fluorescence microscopy, enabling the comprehensive analysis of biophysical parameters along the gradient direction. Furthermore, human adipose-derived stem cells were seeded onto the gradient films to investigate the influence of protein density on cell attachment, showing that the distribution of the cells can be modulated by the arrangement of the laminin at the air-water interface. The presented approach not only allowed us to gain insights into the intricate interplay between biophysical cues and cell behavior within complex tissue environments, but it is also suited as a screening approach to determine optimal protein concentrations to achieve a target cellular output.
Collapse
Affiliation(s)
- Shanshan Zhang
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
- Helmholtz-Zentrum Hereon, Institute of Active Polymers, Kantstrasse 55, 14513 Teltow, Germany
| | - Oliver Felthaus
- Department of Plastic Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Nan Ma
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
- Helmholtz-Zentrum Hereon, Institute of Sustainable Materials, Kantstrasse 55, 14513 Teltow, Germany
| | - Rainhard Machatschek
- Helmholtz-Zentrum Hereon, Institute of Active Polymers, Kantstrasse 55, 14513 Teltow, Germany
| |
Collapse
|
6
|
Habli Z, Lahoud R, Zantout A, Abou-Kheir W, Khraiche ML. Single-cell fluid-based force spectroscopy reveals near lipid size nano-topography effects on neural cell adhesion. LAB ON A CHIP 2024; 24:707-718. [PMID: 38230917 DOI: 10.1039/d3lc00984j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Nano-roughness has shown great potential in enhancing high-fidelity electrogenic cell interfaces, owing to its characteristic topography comparable to proteins and lipids, which influences a wide range of cellular mechanical responses. Gaining a comprehensive understanding of how cells respond to nano-roughness at the single-cell level is not only imperative for implanted devices but also essential for tissue regeneration and interaction with complex biomaterial surfaces. In this study, we quantify cell adhesion and biomechanics of single cells to nano-roughened surfaces by measuring neural cell adhesion and biomechanics via fluidic-based single-cell force spectroscopy (SCFS). For this, we introduce nanoscale topographical features on polyimide (PI) surfaces achieving roughness up to 25 nm without chemical modifications. Initial adhesion experiments show cell-specific response to nano-roughness for neuroblastoma cells (SH-SY5Y) compared to human astrocytes (NHA) around 15 and 20 nm surface roughness. In addition, our SCFS measurements revealed a remarkable 2.5-fold increase in adhesion forces (150-164 nN) for SH-SY5Y cells cultured on roughened PI (rPI) surfaces compared to smooth surfaces (60-107 nN). Our data also shows that cells can distinguish changes in nano-roughness as small 2 nm (close to the diameter of a single lipid) and show roughness dependence adhesion while favoring 15 nm. Notably, this enhanced adhesion is accompanied by increased cell elongation upon cell detachment without any significant differences in cell area spreading. The study provides valuable insights into the interplay between nano-topography and cellular responses and offers practical implications for designing biomaterial surfaces with enhanced cellular interactions.
Collapse
Affiliation(s)
- Zeina Habli
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Rima Lahoud
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Ahmad Zantout
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Massoud L Khraiche
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon.
| |
Collapse
|
7
|
Huang LZY, Shaw ZL, Penman R, Cheeseman S, Truong VK, Higgins MJ, Caruso RA, Elbourne A. Cell Adhesion, Elasticity, and Rupture Forces Guide Microbial Cell Death on Nanostructured Antimicrobial Titanium Surfaces. ACS APPLIED BIO MATERIALS 2024; 7:344-361. [PMID: 38100088 DOI: 10.1021/acsabm.3c00943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Naturally occurring and synthetic nanostructured surfaces have been widely reported to resist microbial colonization. The majority of these studies have shown that both bacterial and fungal cells are killed upon contact and subsequent surface adhesion to such surfaces. This occurs because the presence of high-aspect-ratio structures can initiate a self-driven mechanical rupture of microbial cells during the surface adsorption process. While this technology has received a large amount of scientific and medical interest, one important question still remains: what factors drive microbial death on the surface? In this work, the interplay between microbial-surface adhesion, cell elasticity, cell membrane rupture forces, and cell lysis at the microbial-nanostructure biointerface during adsorptive processes was assessed using a combination of live confocal laser scanning microscopy, scanning electron microscopy, in situ amplitude atomic force microscopy, and single-cell force spectroscopy. Specifically, the adsorptive behavior and nanomechanical properties of live Gram-negative (Pseudomonas aeruginosa) and Gram-positive (methicillin-resistant Staphylococcus aureus) bacterial cells, as well as the fungal species Candida albicans and Cryptococcus neoformans, were assessed on unmodified and nanostructured titanium surfaces. Unmodified titanium and titanium surfaces with nanostructures were used as model substrates for investigation. For all microbial species, cell elasticity, rupture force, maximum cell-surface adhesion force, the work of adhesion, and the cell-surface tether behavior were compared to the relative cell death observed for each surface examined. For cells with a lower elastic modulus, lower force to rupture through the cell, and higher work of adhesion, the surfaces had a higher antimicrobial activity, supporting the proposed biocidal mode of action for nanostructured surfaces. This study provides direct quantification of the differences observed in the efficacy of nanostructured antimicrobial surface as a function of microbial species indicating that a universal, antimicrobial surface architecture may be hard to achieve.
Collapse
Affiliation(s)
- Louisa Z Y Huang
- Applied Chemistry and Environmental Science, School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia
| | - Z L Shaw
- School of Engineering, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia
| | - Rowan Penman
- Applied Chemistry and Environmental Science, School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia
| | - Samuel Cheeseman
- Applied Chemistry and Environmental Science, School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia
- Graeme Clark Institute, Faculty of Engineering and Information Technology & Faculty of Medicine, Dentistry and Health Services, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Vi Khanh Truong
- Applied Chemistry and Environmental Science, School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Michael J Higgins
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Rachel A Caruso
- Applied Chemistry and Environmental Science, School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia
| | - Aaron Elbourne
- Applied Chemistry and Environmental Science, School of Science, College of STEM, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
8
|
Wei J, Yang Y, Li M. Single-cell force spectroscopy of fluid flow-tuned cell adhesion for dissecting hemodynamics in tumor metastasis. NANOSCALE 2023; 16:360-372. [PMID: 38063483 DOI: 10.1039/d3nr04439d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Cell adhesion plays an important role in regulating the metastasis of cancer cells, and atomic force microscopy (AFM)-based single-cell force spectroscopy (SCFS) has become an important method to directly measure the adhesion forces of individual cells. Particularly, bodily fluid flow environments strongly affect the functions and behaviors of metastatic cells for successful dissemination. Nevertheless, the interactions between fluidic flow medium environment and cell adhesion remain poorly understood. In this work, AFM-based SCFS was exploited to examine the effects of fluidic flow environment on cellular adhesion. A fluidic cell culture medium device was used to simulate the fluidic flow environment experienced by cancer cells during metastasis, which was combined with AFM-based SCFS assay. A single living cancer cell was attached to the AFM tipless cantilever to prepare the single-cell probe for performing SCFS experiments on the mesothelial cells grown under the fluidic flow medium conditions, and the effects of experimental parameters (retraction speed, contact time, loading force) on the measured cellular adhesion forces were analyzed. Experimental results of SCFS assay show that cellular adhesion forces significantly decrease after growth in fluidic flow medium, whereas cellular adhesion forces increase after growth in static culture medium. Experiments performed with the use of spherical probes coated with cell adhesion-associated biomolecules also show the weakening of cell adhesion after growth in fluidic flow cell culture medium, which was subsequently confirmed by the confocal fluorescence microscopy experiments of cell adhesion molecules, vividly illustrating the remarkable effects of fluidic flow environment on cellular adhesion. The study provides a new approach to detect adhesion force dynamics involved in the interactions between cells and the fluidic flow environment at the single-cell level, which will facilitate dissecting the role of hemodynamics in tumor metastasis.
Collapse
Affiliation(s)
- Jiajia Wei
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanqi Yang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110169, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
Gulati K, Adachi T. Profiling to Probing: Atomic force microscopy to characterize nano-engineered implants. Acta Biomater 2023; 170:15-38. [PMID: 37562516 DOI: 10.1016/j.actbio.2023.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
Surface modification of implants in the nanoscale or implant nano-engineering has been recognized as a strategy for augmenting implant bioactivity and achieving long-term implant success. Characterizing and optimizing implant characteristics is crucial to achieving desirable effects post-implantation. Modified implant enables tailored, guided and accelerated tissue integration; however, our understanding is limited to multicellular (bulk) interactions. Finding the nanoscale forces experienced by a single cell on nano-engineered implants will aid in predicting implants' bioactivity and engineering the next generation of bioactive implants. Atomic force microscope (AFM) is a unique tool that enables surface characterization and understanding of the interactions between implant surface and biological tissues. The characterization of surface topography using AFM to gauge nano-engineered implants' characteristics (topographical, mechanical, chemical, electrical and magnetic) and bioactivity (adhesion of cells) is presented. A special focus of the review is to discuss the use of single-cell force spectroscopy (SCFS) employing AFM to investigate the minute forces involved with the adhesion of a single cell (resident tissue cell or bacterium) to the surface of nano-engineered implants. Finally, the research gaps and future perspectives relating to AFM-characterized current and emerging nano-engineered implants are discussed towards achieving desirable bioactivity performances. This review highlights the use of advanced AFM-based characterization of nano-engineered implant surfaces via profiling (investigating implant topography) or probing (using a single cell as a probe to study precise adhesive forces with the implant surface). STATEMENT OF SIGNIFICANCE: Nano-engineering is emerging as a surface modification platform for implants to augment their bioactivity and achieve favourable treatment outcomes. In this extensive review, we closely examine the use of Atomic Force Microscopy (AFM) to characterize the properties of nano-engineered implant surfaces (topography, mechanical, chemical, electrical and magnetic). Next, we discuss Single-Cell Force Spectroscopy (SCFS) via AFM towards precise force quantification encompassing a single cell's interaction with the implant surface. This interdisciplinary review will appeal to researchers from the broader scientific community interested in implants and cell adhesion to implants and provide an improved understanding of the surface characterization of nano-engineered implants.
Collapse
Affiliation(s)
- Karan Gulati
- Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto 606-8507, Japan; The University of Queensland, School of Dentistry, Herston QLD 4006, Australia.
| | - Taiji Adachi
- Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto 606-8507, Japan
| |
Collapse
|
10
|
Holuigue H, Nacci L, Di Chiaro P, Chighizola M, Locatelli I, Schulte C, Alfano M, Diaferia GR, Podestà A. Native extracellular matrix probes to target patient- and tissue-specific cell-microenvironment interactions by force spectroscopy. NANOSCALE 2023; 15:15382-15395. [PMID: 37700706 DOI: 10.1039/d3nr01568h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Atomic Force Microscopy (AFM) is successfully used for the quantitative investigation of the cellular mechanosensing of the microenvironment. To this purpose, several force spectroscopy approaches aim at measuring the adhesive forces between two living cells and also between a cell and an appropriate reproduction of the extracellular matrix (ECM), typically exploiting tips suitably functionalised with single components (e.g. collagen, fibronectin) of the ECM. However, these probes only poorly reproduce the complexity of the native cellular microenvironment and consequently of the biological interactions. We developed a novel approach to produce AFM probes that faithfully retain the structural and biochemical complexity of the ECM; this was achieved by attaching to an AFM cantilever a micrometric slice of native decellularised ECM, which was cut by laser microdissection. We demonstrate that these probes preserve the morphological, mechanical, and chemical heterogeneity of the ECM. Native ECM probes can be used in force spectroscopy experiments aimed at targeting cell-microenvironment interactions. Here, we demonstrate the feasibility of dissecting mechanotransductive cell-ECM interactions in the 10 pN range. As proof-of-principle, we tested a rat bladder ECM probe against the AY-27 rat bladder cancer cell line. On the one hand, we obtained reproducible results using different probes derived from the same ECM regions; on the other hand, we detected differences in the adhesion patterns of distinct bladder ECM regions (submucosa, detrusor, and adventitia), in line with the disparities in composition and biophysical properties of these ECM regions. Our results demonstrate that native ECM probes, produced from patient-specific regions of organs and tissues, can be used to investigate cell-microenvironment interactions and early mechanotransductive processes by force spectroscopy. This opens new possibilities in the field of personalised medicine.
Collapse
Affiliation(s)
- H Holuigue
- CIMAINA and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milano, Italy.
| | - L Nacci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy.
| | - P Di Chiaro
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy.
| | - M Chighizola
- CIMAINA and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milano, Italy.
| | - I Locatelli
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy.
| | - C Schulte
- CIMAINA and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milano, Italy.
- Department of Biomedical and Clinical Sciences "L. Sacco", Università degli Studi di Milano, Milano, Italy
| | - M Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy.
| | - G R Diaferia
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy.
| | - A Podestà
- CIMAINA and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
11
|
Zhou G, Wang C, Wu C, Zhang W. Active learning model for extracting elastic modulus of cell on substrate. Biophys J 2023; 122:2489-2499. [PMID: 37147802 PMCID: PMC10323012 DOI: 10.1016/j.bpj.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/02/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023] Open
Abstract
The cell elastic modulus (Ec) is widely used as the mechanics-based marker to analyze the biological effects of substrates on cells. However, the employment of the Hertz model to extract the apparent Ec can cause errors due to the disobedience of the small deformation assumption and the infinite half-space assumption, as well as an inability to deduct the deformation of the substrate. So far, no model can effectively solve the errors caused by the above-mentioned aspects simultaneously. In response to this, herein, we propose an active learning model to extract Ec. The numerical calculation with finite element suggests the good prediction accuracy of the model. The indentation experiments on both hydrogel and cell indicate that the established model can efficiently reduce the error caused by the method of extracting Ec. The application of this model may facilitate our understanding about the role of Ec in correlating the stiffness of substrate and the biological behavior of cell.
Collapse
Affiliation(s)
- Guanlin Zhou
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Chao Wang
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Chengwei Wu
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Wei Zhang
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China.
| |
Collapse
|
12
|
Kav B, Weikl TR, Schneck E. Measuring pico-Newton Forces with Lipid Anchors as Force Sensors in Molecular Dynamics Simulations. J Phys Chem B 2023; 127:4081-4089. [PMID: 37127845 PMCID: PMC10184124 DOI: 10.1021/acs.jpcb.3c00063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Binding forces between biomolecules are ubiquitous in nature but sometimes as weak as a few pico-Newtons (pN). In many cases, the binding partners are attached to biomembranes with the help of a lipid anchor. One important example are glycolipids that promote membrane adhesion through weak carbohydrate-carbohydrate binding between adjacent membranes. Here, we use molecular dynamics (MD) simulations to quantify the forces generated by bonds involving membrane-anchored molecules. We introduce a method in which the protrusion of the lipid anchors from the membrane acts as the force sensor. Our results with two different glycolipids reveal binding forces of up to 20 pN and corroborate the recent notion that carbohydrate-carbohydrate interactions are generic rather than specific.
Collapse
Affiliation(s)
- Batuhan Kav
- Max Planck Institute of Colloids and Interfaces, 14467, Potsdam, Germany
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Thomas R Weikl
- Max Planck Institute of Colloids and Interfaces, 14467, Potsdam, Germany
| | - Emanuel Schneck
- Max Planck Institute of Colloids and Interfaces, 14467, Potsdam, Germany
- Institute for Condensed Matter Physics, Technische Universität Darmstadt, 64289 Darmstadt, Germany
| |
Collapse
|
13
|
Tanaka M, Thoma J, Poisa-Beiro L, Wuchter P, Eckstein V, Dietrich S, Pabst C, Müller-Tidow C, Ohta T, Ho AD. Physical biomarkers for human hematopoietic stem and progenitor cells. Cells Dev 2023; 174:203845. [PMID: 37116713 DOI: 10.1016/j.cdev.2023.203845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023]
Abstract
Adhesion of hematopoietic stem and progenitor cells (HSPCs) to the bone marrow niche plays critical roles in the maintenance of the most primitive HSPCs. The interactions of HSPC-niche interactions are clinically relevant in acute myeloid leukemia (AML), because (i) leukemia-initiating cells adhered to the marrow niche are protected from the cytotoxic effect by chemotherapy and (ii) mobilization of HSPCs from healthy donors' bone marrow is crucial for the effective stem cell transplantation. However, although many clinical agents have been developed for the HSPC mobilization, the effects caused by the extrinsic molecular cues were traditionally evaluated based on phenomenological observations. This review highlights the recent interdisciplinary challenges of hematologists, biophysicists and cell biologists towards the design of defined in vitro niche models and the development of physical biomarkers for quantitative indexing of differential effects of clinical agents on human HSPCs.
Collapse
Affiliation(s)
- Motomu Tanaka
- Physical Chemistry of Biosystems, Institute of Physical Chemistry, INF253, Heidelberg University, 69120 Heidelberg, Germany; Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, 606-8501 Kyoto, Japan.
| | - Judith Thoma
- Physical Chemistry of Biosystems, Institute of Physical Chemistry, INF253, Heidelberg University, 69120 Heidelberg, Germany
| | - Laura Poisa-Beiro
- Department of Medicine V, Heidelberg University, INF410, 69120 Heidelberg, Germany
| | - Patrick Wuchter
- Department of Medicine V, Heidelberg University, INF410, 69120 Heidelberg, Germany
| | - Volker Eckstein
- Department of Medicine V, Heidelberg University, INF410, 69120 Heidelberg, Germany
| | - Sascha Dietrich
- Department of Medicine V, Heidelberg University, INF410, 69120 Heidelberg, Germany
| | - Caroline Pabst
- Department of Medicine V, Heidelberg University, INF410, 69120 Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Medicine V, Heidelberg University, INF410, 69120 Heidelberg, Germany
| | - Takao Ohta
- Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, 606-8501 Kyoto, Japan
| | - Anthony D Ho
- Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, 606-8501 Kyoto, Japan; Department of Medicine V, Heidelberg University, INF410, 69120 Heidelberg, Germany; Molecular Medicine Partnership Unit Heidelberg, European Molecular Biology Laboratory (EMBL), Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
14
|
He S, Zhang Y, Zhao C, Wang X, Baddi S, Wu B, Dou X, Feng C. Assembly of Helical Nanostructures: Solvent-Induced Morphology Transition and Its Effect on Cell Adhesion. Chemistry 2023; 29:e202202735. [PMID: 36404280 DOI: 10.1002/chem.202202735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022]
Abstract
Being able to precisely manipulate both the morphology and chiroptical signals of supramolecular assemblies will help to better understand the natural biological self-assembly mechanism. Two simple l/d-phenylalanine-based derivatives (L/DPFM) have been designed, and their solvent-dependent morphology evolutions are illustrated. It was found that, as the content of H2 O in aqueous ethanol solutions was increased, LPFM self-assembles first into right-handed nanofibers, then flat fibrous structures, and finally inversed left-handed nanofibers. Assemblies in ethanol and H2 O exhibit opposite conformations and circular dichroism (CD) signals even though they are constructed from the same molecules. Thus, the morphology-dependent cell adhesion and proliferation behaviors are further characterized. Left-handed nanofibers are found to be more favorable for cell adhesion than right-handed nanostructures. Quantitative AFM analysis showed that the L929 cell adhesion force on left-handed LPFM fibers is much higher than that on structures with inversed handedness. Moreover, the value of cell Young's modulus is lower for left-handed nanofibrous films, which indicates better flexibility. The difference in cell-substrate interactions might lead to different effects on cell behavior.
Collapse
Affiliation(s)
- Sijia He
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Yaqian Zhang
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Changli Zhao
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Xueqian Wang
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Sravan Baddi
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Beibei Wu
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Xiaoqiu Dou
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Chuanliang Feng
- State Key Lab of Metal Matrix Composites School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| |
Collapse
|
15
|
Angeloni L, Popa B, Nouri-Goushki M, Minneboo M, Zadpoor AA, Ghatkesar MK, Fratila-Apachitei LE. Fluidic Force Microscopy and Atomic Force Microscopy Unveil New Insights into the Interactions of Preosteoblasts with 3D-Printed Submicron Patterns. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204662. [PMID: 36373704 DOI: 10.1002/smll.202204662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Physical patterns represent potential surface cues for promoting osteogenic differentiation of stem cells and improving osseointegration of orthopedic implants. Understanding the early cell-surface interactions and their effects on late cellular functions is essential for a rational design of such topographies, yet still elusive. In this work, fluidic force microscopy (FluidFM) and atomic force microscopy (AFM) combined with optical and electron microscopy are used to quantitatively investigate the interaction of preosteoblasts with 3D-printed patterns after 4 and 24 h of culture. The patterns consist of pillars with the same diameter (200 nm) and interspace (700 nm) but distinct heights (500 and 1000 nm) and osteogenic properties. FluidFM reveals a higher cell adhesion strength after 24 h of culture on the taller pillars (32 ± 7 kPa versus 21.5 ± 12.5 kPa). This is associated with attachment of cells partly on the sidewalls of these pillars, thus requiring larger normal forces for detachment. Furthermore, the higher resistance to shear forces observed for these cells indicates an enhanced anchorage and can be related to the persistence and stability of lamellipodia. The study explains the differential cell adhesion behavior induced by different pillar heights, enabling advancements in the rational design of osteogenic patterns.
Collapse
Affiliation(s)
- Livia Angeloni
- Department of Precision and Microsystems Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Bogdan Popa
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Mahdiyeh Nouri-Goushki
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Michelle Minneboo
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Murali K Ghatkesar
- Department of Precision and Microsystems Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| | - Lidy E Fratila-Apachitei
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, Delft, 2628CD, The Netherlands
| |
Collapse
|
16
|
Fuchs M, Radeva MY, Spindler V, Vielmuth F, Kugelmann D, Waschke J. Cytoskeletal anchorage of different Dsg3 pools revealed by combination of hybrid STED/SMFS-AFM. Cell Mol Life Sci 2023; 80:25. [PMID: 36602635 PMCID: PMC9816259 DOI: 10.1007/s00018-022-04681-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023]
Abstract
Desmoglein 3 (Dsg3) is a desmosomal cadherin mediating cell adhesion within desmosomes and is the antigen of the autoimmune blistering skin disease pemphigus vulgaris. Therefore, understanding of the complex desmosome turnover process is of high biomedical relevance. Recently, super resolution microscopy was used to characterize desmosome composition and turnover. However, studies were limited because adhesion measurements on living cells were not possible in parallel. Before desmosomal cadherins are incorporated into nascent desmosomes, they are not bound to intermediate filaments but were suggested to be associated with the actin cytoskeleton. However, direct proof that adhesion of a pool of desmosomal cadherins is dependent on actin is missing. Here, we applied single-molecule force spectroscopy measurements with the novel single molecule hybrid-technique STED/SMFS-AFM to investigate the cytoskeletal anchorage of Dsg3 on living keratinocytes for the first time. By application of pharmacological agents we discriminated two different Dsg3 pools, only one of which is anchored to actin filaments. We applied the actin polymerization inhibitor Latrunculin B to modify the actin cytoskeleton and the PKCα activator PMA to modulate intermediate filament anchorage. On the cellular surface Dsg3 adhesion was actin-dependent. In contrast, at cell-cell contacts, Dsg3 adhesion was independent from actin but rather is regulated by PKC which is well established to control desmosome turn-over via intermediate filament anchorage. Taken together, using the novel STED/SMFS-AFM technique, we demonstrated the existence of two Dsg3 pools with different cytoskeletal anchorage mechanisms.
Collapse
Affiliation(s)
- Michael Fuchs
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Volker Spindler
- Department of Biomedicine and Institute of Anatomy, University of Basel, Basel, Switzerland
| | - Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Daniela Kugelmann
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| |
Collapse
|
17
|
Rosenbohm J, Minnick G, Safa BT, Esfahani AM, Jin X, Zhai H, Lavrik NV, Yang R. A multi-material platform for imaging of single cell-cell junctions under tensile load fabricated with two-photon polymerization. Biomed Microdevices 2022; 24:33. [PMID: 36207557 PMCID: PMC11104271 DOI: 10.1007/s10544-022-00633-z] [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] [Accepted: 09/04/2022] [Indexed: 11/29/2022]
Abstract
We previously reported a single-cell adhesion micro tensile tester (SCAμTT) fabricated from IP-S photoresin with two-photon polymerization (TPP) for investigating the mechanics of a single cell-cell junction under defined tensile loading. A major limitation of the platform is the autofluorescence of IP-S, the photoresin for TPP fabrication, which significantly increases background signal and makes fluorescent imaging of stretched cells difficult. In this study, we report the design and fabrication of a new SCAμTT platform that mitigates autofluorescence and demonstrate its capability in imaging a single cell pair as its mutual junction is stretched. By employing a two-material design using IP-S and IP-Visio, a photoresin with reduced autofluorescence, we show a significant reduction in autofluorescence of the platform. Further, by integrating apertures onto the substrate with a gold coating, the influence of autofluorescence on imaging is almost completely mitigated. With this new platform, we demonstrate the ability to image a pair of epithelial cells as they are stretched up to 250% strain, allowing us to observe junction rupture and F-actin retraction while simultaneously recording the accumulation of over 800 kPa of stress in the junction. The platform and methodology presented here can potentially enable detailed investigation of the mechanics of and mechanotransduction in cell-cell junctions and improve the design of other TPP platforms in mechanobiology applications.
Collapse
Affiliation(s)
- Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Grayson Minnick
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Bahareh Tajvidi Safa
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Haiwei Zhai
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Nickolay V Lavrik
- Center for Nanophase Materials Sciences, Oak Ridge National Laboratory, Oak Ridge, TN, 37831-6054, USA.
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
| |
Collapse
|
18
|
Nagy ÁG, Székács I, Bonyár A, Horvath R. Cell-substratum and cell-cell adhesion forces and single-cell mechanical properties in mono- and multilayer assemblies from robotic fluidic force microscopy. Eur J Cell Biol 2022; 101:151273. [PMID: 36088812 DOI: 10.1016/j.ejcb.2022.151273] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 12/14/2022] Open
Abstract
The epithelium covers, protects, and actively regulates various formations and cavities of the human body. During embryonic development the assembly of the epithelium is crucial to the organoid formation, and the invasion of the epithelium is an essential step in cancer metastasis. Live cell mechanical properties and associated forces presumably play an important role in these biological processes. However, the direct measurement of cellular forces in a precise and high-throughput manner is still challenging. We studied the cellular adhesion maturation of epithelial Vero monolayers by measuring single-cell force-spectra with high-throughput fluidic force microscopy (robotic FluidFM). Vero cells were grown on gelatin-covered plates in different seeding concentrations, and cell detachment forces were recorded from the single-cell state, through clustered island formation, to their complete assembly into a sparse and then into a tight monolayer. A methodology was proposed to separate cell-substratum and cell-cell adhesion force and energy (work of adhesion) contributions based on the recorded force-distance curves. For comparison, cancerous HeLa cells were also measured in the same settings. During Vero monolayer formation, a significantly strengthening adhesive tendency was found, showing the development of cell-cell contacts. Interestingly, this type of step-by-step maturation was absent in HeLa cells. The attachment of cancerous HeLa cells to the assembled epithelial monolayers was also measured, proposing a new high-throughput method to investigate the biomechanics of cancer cell invasion. We found that HeLa cells adhere significantly stronger to the tight Vero monolayer than cells of the same origin. Moreover, the mechanical characteristics of Vero monolayers upon cancerous HeLa cell influence were recorded and analyzed. All these results provide insight into the qualitative assessment of cell-substratum and cell-cell mechanical contacts in mono- and multilayered assemblies and demonstrate the robustness and speed of the robotic FluidFM technology to reveal biomechanical properties of live cell assemblies with statistical significances.
Collapse
Affiliation(s)
- Ágoston G Nagy
- Department of Electronics Technology, Faculty of Electrical Engineering and Informatics, Budapest University of Technology and Economics, Budapest, Hungary; Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Inna Székács
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Attila Bonyár
- Department of Electronics Technology, Faculty of Electrical Engineering and Informatics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Robert Horvath
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary.
| |
Collapse
|
19
|
Cell Adhesion Strength Indicates the Antithrombogenicity of Poly(2-Methoxyethyl Acrylate) (PMEA): Potential Candidate for Artificial Small-Diameter Blood Vessel. SURFACES 2022. [DOI: 10.3390/surfaces5030027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Poly (2-methoxyethyl acrylate) (PMEA) is a US FDA-approved biocompatible polymer, although there is insufficient work on human umbilical vein endothelial cells (HUVECs) and platelet interaction analysis on PMEA-analogous polymers. In this study, we extensively investigated HUVEC–polymer and platelet–polymer interaction behavior by measuring the adhesion strength using single-cell force spectroscopy. Furthermore, the hydration layer of the polymer interface was observed using frequency-modulation atomic force microscopy. We found that endothelial cells can attach and spread on the PMEA surface with strong adhesion strength compared to other analogous polymers. We found that the hydration layers on the PMEA-analogous polymers were closely related to their weak platelet adhesion behavior. Based on our results, it can be concluded that PMEA is a promising candidate for the construction of artificial small-diameter blood vessels owing to the presence of IW and a hydration layer on the interface.
Collapse
|
20
|
Nishida K, Anada T, Tanaka M. Roles of interfacial water states on advanced biomedical material design. Adv Drug Deliv Rev 2022; 186:114310. [PMID: 35487283 DOI: 10.1016/j.addr.2022.114310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/12/2022] [Accepted: 04/21/2022] [Indexed: 12/15/2022]
Abstract
When biomedical materials come into contact with body fluids, the first reaction that occurs on the material surface is hydration; proteins are then adsorbed and denatured on the hydrated material surface. The amount and degree of denaturation of adsorbed proteins affect subsequent cell behavior, including cell adhesion, migration, proliferation, and differentiation. Biomolecules are important for understanding the interactions and biological reactions of biomedical materials to elucidate the role of hydration in biomedical materials and their interaction partners. Analysis of the water states of hydrated materials is complicated and remains controversial; however, knowledge about interfacial water is useful for the design and development of advanced biomaterials. Herein, we summarize recent findings on the hydration of synthetic polymers, supramolecular materials, inorganic materials, proteins, and lipid membranes. Furthermore, we present recent advances in our understanding of the classification of interfacial water and advanced polymer biomaterials, based on the intermediate water concept.
Collapse
Affiliation(s)
- Kei Nishida
- Institute for Materials Chemistry and Engineering Kyushu university, 744 Motooka, Nishi-ku Fukuoka 819-0395, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Japan(1)
| | - Takahisa Anada
- Institute for Materials Chemistry and Engineering Kyushu university, 744 Motooka, Nishi-ku Fukuoka 819-0395, Japan
| | - Masaru Tanaka
- Institute for Materials Chemistry and Engineering Kyushu university, 744 Motooka, Nishi-ku Fukuoka 819-0395, Japan.
| |
Collapse
|
21
|
Al-madani H, Du H, Yao J, Peng H, Yao C, Jiang B, Wu A, Yang F. Living Sample Viability Measurement Methods from Traditional Assays to Nanomotion. BIOSENSORS 2022; 12:453. [PMID: 35884256 PMCID: PMC9313330 DOI: 10.3390/bios12070453] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/18/2022]
Abstract
Living sample viability measurement is an extremely common process in medical, pharmaceutical, and biological fields, especially drug pharmacology and toxicology detection. Nowadays, there are a number of chemical, optical, and mechanical methods that have been developed in response to the growing demand for simple, rapid, accurate, and reliable real-time living sample viability assessment. In parallel, the development trend of viability measurement methods (VMMs) has increasingly shifted from traditional assays towards the innovative atomic force microscope (AFM) oscillating sensor method (referred to as nanomotion), which takes advantage of the adhesion of living samples to an oscillating surface. Herein, we provide a comprehensive review of the common VMMs, laying emphasis on their benefits and drawbacks, as well as evaluating the potential utility of VMMs. In addition, we discuss the nanomotion technique, focusing on its applications, sample attachment protocols, and result display methods. Furthermore, the challenges and future perspectives on nanomotion are commented on, mainly emphasizing scientific restrictions and development orientations.
Collapse
Affiliation(s)
- Hamzah Al-madani
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Du
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junlie Yao
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hao Peng
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenyang Yao
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Jiang
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| | - Fang Yang
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS), Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China; (H.A.-m.); (H.D.); (J.Y.); (H.P.); (C.Y.); (B.J.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, China
| |
Collapse
|
22
|
Poly(2-Methoxyethyl Acrylate) (PMEA)-Coated Anti-Platelet Adhesive Surfaces to Mimic Native Blood Vessels through HUVECs Attachment, Migration, and Monolayer Formation. COATINGS 2022. [DOI: 10.3390/coatings12060869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Confluent monolayers of human umbilical vein endothelial cells (HUVECs) on a poly(2-methoxyethyl acrylate) (PMEA) antithrombogenic surface play a major role in mimicking the inner surface of native blood vessels. In this study, we extensively investigated the behavior of cell–polymer and cell–cell interactions by measuring adhesion strength using single-cell force spectroscopy. In addition, the attachment and migration of HUVECs on PMEA-analogous substrates were detected, and the migration rate was estimated. Moreover, the bilateral migration of HUVECs between two adjacent surfaces was observed. Furthermore, the outer surface of HUVEC was examined using frequency-modulation atomic force microscopy (FM-AFM). Hydration was found to be an indication of a healthy glycocalyx layer. The results were compared with the hydration states of individual PMEA-analogous polymers to understand the adhesion mechanism between the cells and substrates in the interface region. HUVECs could attach and spread on the PMEA surface with stronger adhesion strength than self-adhesion strength, and migration occurred over the surface of analogue polymers. We confirmed that platelets could not adhere to HUVEC monolayers cultured on the PMEA surface. FM-AFM images revealed a hydration layer on the HUVEC surfaces, indicating the presence of components of the glycocalyx layer in the presence of intermediate water. Our findings show that PMEA can mimic original blood vessels through an antithrombogenic HUVEC monolayer and is thus suitable for the construction of artificial small-diameter blood vessels.
Collapse
|
23
|
Qiu Y, Chien CC, Maroulis B, Bei J, Gaitas A, Gong B. Extending applications of AFM to fluidic AFM in single living cell studies. J Cell Physiol 2022; 237:3222-3238. [PMID: 35696489 PMCID: PMC9378449 DOI: 10.1002/jcp.30809] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/25/2022] [Indexed: 12/30/2022]
Abstract
In this article, a review of a series of applications of atomic force microscopy (AFM) and fluidic Atomic Force Microscopy (fluidic AFM, hereafter fluidFM) in single-cell studies is presented. AFM applications involving single-cell and extracellular vesicle (EV) studies, colloidal force spectroscopy, and single-cell adhesion measurements are discussed. FluidFM is an offshoot of AFM that combines a microfluidic cantilever with AFM and has enabled the research community to conduct biological, pathological, and pharmacological studies on cells at the single-cell level in a liquid environment. In this review, capacities of fluidFM are discussed to illustrate (1) the speed with which sequential measurements of adhesion using coated colloid beads can be done, (2) the ability to assess lateral binding forces of endothelial or epithelial cells in a confluent cell monolayer in an appropriate physiological environment, and (3) the ease of measurement of vertical binding forces of intercellular adhesion between heterogeneous cells. Furthermore, key applications of fluidFM are reviewed regarding to EV absorption, manipulation of a single living cell by intracellular injection, sampling of cellular fluid from a single living cell, patch clamping, and mass measurements of a single living cell.
Collapse
Affiliation(s)
- Yuan Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Chen-Chi Chien
- The Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Basile Maroulis
- The Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Jiani Bei
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Angelo Gaitas
- The Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,BioMedical Engineering & Imaging Institute, Leon and Norma Hess Center for Science and Medicine, New York City, New York, USA
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA.,Sealy Center for Vector Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, Texas, USA.,Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA.,Institute for Human Infectious and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
24
|
Xie L, Sun Z, Brown NJ, Glinskii OV, Meininger GA, Glinsky VV. Changes in dynamics of tumor/endothelial cell adhesive interactions depending on endothelial cell growth state and elastic properties. PLoS One 2022; 17:e0269552. [PMID: 35666755 PMCID: PMC9170101 DOI: 10.1371/journal.pone.0269552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer cell adhesion to the endothelium is a crucial process in hematogenous metastasis, but how the integrity of the endothelial barrier and endothelial cell (EC) mechanical properties influence the adhesion between metastatic cancer cells and the endothelium remain unclear. In the present study, we have measured the adhesion between single cancer cells and two types of ECs at various growth states and their mechanical properties (elasticity) using atomic force microscopy single cell force spectroscopy. We demonstrated that the EC stiffness increased and adhesion with cancer cells decreased, as ECs grew from a single cell to a confluent state and developed cell-cell contacts, but this was reversed when confluent cells returned to a single state in a scratch assay. Our results suggest that the integrity of the endothelial barrier is an important factor in reducing the ability of the metastatic tumor cells to adhere to the vascular endothelium, extravasate and lodge in the vasculature of a distant organ where secondary metastatic tumors would develop.
Collapse
Affiliation(s)
- Leike Xie
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Zhe Sun
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
| | - Nicola J. Brown
- Microcirculation Research Group, Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Olga V. Glinskii
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States of America
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States of America
| | - Gerald A. Meininger
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Vladislav V. Glinsky
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, United States of America
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States of America
| |
Collapse
|
25
|
Nagy ÁG, Kanyó N, Vörös A, Székács I, Bonyár A, Horvath R. Population distributions of single-cell adhesion parameters during the cell cycle from high-throughput robotic fluidic force microscopy. Sci Rep 2022; 12:7747. [PMID: 35546603 PMCID: PMC9095720 DOI: 10.1038/s41598-022-11770-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022] Open
Abstract
Single-cell adhesion plays an essential role in biological and biomedical sciences, but its precise measurement for a large number of cells is still a challenging task. At present, typical force measuring techniques usually offer low throughput, a few cells per day, and therefore are unable to uncover phenomena emerging at the population level. In this work, robotic fluidic force microscopy (FluidFM) was utilized to measure the adhesion parameters of cells in a high-throughput manner to study their population distributions in-depth. The investigated cell type was the genetically engineered HeLa Fucci construct with cell cycle-dependent expression of fluorescent proteins. This feature, combined with the high-throughput measurement made it possible for the first time to characterize the single-cell adhesion distributions at various stages of the cell cycle. It was found that parameters such as single-cell adhesion force and energy follow a lognormal population distribution. Therefore, conclusions based on adhesion data of a low number of cells or treating the population as normally distributed can be misleading. Moreover, we found that the cell area was significantly the smallest, and the area normalized maximal adhesion force was significantly the largest for the colorless cells (the mitotic (M) and early G1 phases). Notably, the parameter characterizing the elongation of the cells until the maximum level of force between the cell and its substratum was also dependent on the cell cycle, which quantity was the smallest for the colorless cells. A novel parameter, named the spring coefficient of the cell, was introduced as the fraction of maximal adhesion force and maximal cell elongation during the mechanical detachment, which was found to be significantly the largest for the colorless cells. Cells in the M phase adhere in atypical way, with so-called reticular adhesions, which are different from canonical focal adhesions. We first revealed that reticular adhesion can exert a higher force per unit area than canonical focal adhesions, and cells in this phase are significantly stiffer. The possible biological consequences of these findings were also discussed, together with the practical relevance of the observed population-level adhesion phenomena.
Collapse
Affiliation(s)
- Ágoston G Nagy
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary.,Department of Electronics Technology, Faculty of Electrical Engineering and Informatics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Nicolett Kanyó
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Alexandra Vörös
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Inna Székács
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Attila Bonyár
- Department of Electronics Technology, Faculty of Electrical Engineering and Informatics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Robert Horvath
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary.
| |
Collapse
|
26
|
Atomic Force Microscopy (AFM) Applications in Arrhythmogenic Cardiomyopathy. Int J Mol Sci 2022; 23:ijms23073700. [PMID: 35409059 PMCID: PMC8998711 DOI: 10.3390/ijms23073700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/06/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart muscle disorder characterized by progressive replacement of cardiomyocytes by fibrofatty tissue, ventricular dilatation, cardiac dysfunction, arrhythmias, and sudden cardiac death. Interest in molecular biomechanics for these disorders is constantly growing. Atomic force microscopy (AFM) is a well-established technic to study the mechanobiology of biological samples under physiological and pathological conditions at the cellular scale. However, a review which described all the different data that can be obtained using the AFM (cell elasticity, adhesion behavior, viscoelasticity, beating force, and frequency) is still missing. In this review, we will discuss several techniques that highlight the potential of AFM to be used as a tool for assessing the biomechanics involved in ACM. Indeed, analysis of genetically mutated cells with AFM reveal abnormalities of the cytoskeleton, cell membrane structures, and defects of contractility. The higher the Young’s modulus, the stiffer the cell, and it is well known that abnormal tissue stiffness is symptomatic of a range of diseases. The cell beating force and frequency provide information during the depolarization and repolarization phases, complementary to cell electrophysiology (calcium imaging, MEA, patch clamp). In addition, original data is also presented to emphasize the unique potential of AFM as a tool to assess fibrosis in cardiac tissue.
Collapse
|
27
|
Qin Y, Yang W, Chu H, Li Y, Cai S, Yu H, Liu L. Atomic Force Microscopy for Tumor Research at Cell and Molecule Levels. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2022; 28:1-18. [PMID: 35257653 DOI: 10.1017/s1431927622000290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumors have posed a serious threat to human life and health. Researchers can determine whether or not cells are cancerous, whether the cancer cells are invasive or metastatic, and what the effects of drugs are on cancer cells by the physical properties such as hardness, adhesion, and Young's modulus. The atomic force microscope (AFM) has emerged as a key important tool for biomechanics research on tumor cells due to its ability to image and collect force spectroscopy information of biological samples with nano-level spatial resolution and under near-physiological conditions. This article reviews the existing results of the study of cancer cells with AFM. The main foci are the operating principle of AFM and research advances in mechanical property measurement, ultra-microtopography, and molecular recognition of tumor cells, which allows us to outline what we do know it in a systematic way and to summarize and to discuss future directions.
Collapse
Affiliation(s)
- Yitong Qin
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai264005, China
| | - Wenguang Yang
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai264005, China
| | - Honghui Chu
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai264005, China
| | - Yan Li
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai264005, China
| | - Shuxiang Cai
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai264005, China
| | - Haibo Yu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang110016, China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang110016, China
| |
Collapse
|
28
|
Beeghly GF, Amofa KY, Fischbach C, Kumar S. Regulation of Tumor Invasion by the Physical Microenvironment: Lessons from Breast and Brain Cancer. Annu Rev Biomed Eng 2022; 24:29-59. [PMID: 35119915 DOI: 10.1146/annurev-bioeng-110220-115419] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The success of anticancer therapies is often limited by heterogeneity within and between tumors. While much attention has been devoted to understanding the intrinsic molecular diversity of tumor cells, the surrounding tissue microenvironment is also highly complex and coevolves with tumor cells to drive clinical outcomes. Here, we propose that diverse types of solid tumors share common physical motifs that change in time and space, serving as universal regulators of malignancy. We use breast cancer and glioblastoma as instructive examples and highlight how invasion in both diseases is driven by the appropriation of structural guidance cues, contact-dependent heterotypic interactions with stromal cells, and elevated interstitial fluid pressure and flow. We discuss how engineering strategies show increasing value for measuring and modeling these physical properties for mechanistic studies. Moreover, engineered systems offer great promise for developing and testing novel therapies that improve patient prognosis by normalizing the physical tumor microenvironment. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Garrett F Beeghly
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA;
| | - Kwasi Y Amofa
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA; .,Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, USA
| | - Sanjay Kumar
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, USA.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
29
|
Sun W, Gao X, Lei H, Wang W, Cao Y. Biophysical Approaches for Applying and Measuring Biological Forces. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105254. [PMID: 34923777 PMCID: PMC8844594 DOI: 10.1002/advs.202105254] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Indexed: 05/13/2023]
Abstract
Over the past decades, increasing evidence has indicated that mechanical loads can regulate the morphogenesis, proliferation, migration, and apoptosis of living cells. Investigations of how cells sense mechanical stimuli or the mechanotransduction mechanism is an active field of biomaterials and biophysics. Gaining a further understanding of mechanical regulation and depicting the mechanotransduction network inside cells require advanced experimental techniques and new theories. In this review, the fundamental principles of various experimental approaches that have been developed to characterize various types and magnitudes of forces experienced at the cellular and subcellular levels are summarized. The broad applications of these techniques are introduced with an emphasis on the difficulties in implementing these techniques in special biological systems. The advantages and disadvantages of each technique are discussed, which can guide readers to choose the most suitable technique for their questions. A perspective on future directions in this field is also provided. It is anticipated that technical advancement can be a driving force for the development of mechanobiology.
Collapse
Affiliation(s)
- Wenxu Sun
- School of SciencesNantong UniversityNantong226019P. R. China
| | - Xiang Gao
- Key Laboratory of Intelligent Optical Sensing and IntegrationNational Laboratory of Solid State Microstructureand Department of PhysicsCollaborative Innovation Center of Advanced MicrostructuresNanjing UniversityNanjing210023P. R. China
- Institute of Brain ScienceNanjing UniversityNanjing210023P. R. China
| | - Hai Lei
- Key Laboratory of Intelligent Optical Sensing and IntegrationNational Laboratory of Solid State Microstructureand Department of PhysicsCollaborative Innovation Center of Advanced MicrostructuresNanjing UniversityNanjing210023P. R. China
- Institute of Brain ScienceNanjing UniversityNanjing210023P. R. China
- Chemistry and Biomedicine Innovation CenterNanjing UniversityNanjing210023P. R. China
| | - Wei Wang
- Key Laboratory of Intelligent Optical Sensing and IntegrationNational Laboratory of Solid State Microstructureand Department of PhysicsCollaborative Innovation Center of Advanced MicrostructuresNanjing UniversityNanjing210023P. R. China
- Institute of Brain ScienceNanjing UniversityNanjing210023P. R. China
| | - Yi Cao
- Key Laboratory of Intelligent Optical Sensing and IntegrationNational Laboratory of Solid State Microstructureand Department of PhysicsCollaborative Innovation Center of Advanced MicrostructuresNanjing UniversityNanjing210023P. R. China
- Institute of Brain ScienceNanjing UniversityNanjing210023P. R. China
- MOE Key Laboratory of High Performance Polymer Materials and TechnologyDepartment of Polymer Science & EngineeringCollege of Chemistry & Chemical EngineeringNanjing UniversityNanjing210023P. R. China
- Chemistry and Biomedicine Innovation CenterNanjing UniversityNanjing210023P. R. China
| |
Collapse
|
30
|
Luo M, Yang W, Cartwright TN, Higgins JMG, Chen J. Simultaneous Measurement of Single-Cell Mechanics and Cell-to-Materials Adhesion Using Fluidic Force Microscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:620-628. [PMID: 34981921 DOI: 10.1021/acs.langmuir.1c01973] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The connection between cells and their substrate is essential for biological processes such as cell migration. Atomic force microscopy nanoindentation has often been adopted to measure single-cell mechanics. Very recently, fluidic force microscopy has been developed to enable rapid measurements of cell adhesion. However, simultaneous characterization of the cell-to-material adhesion and viscoelastic properties of the same cell is challenging. In this study, we present a new approach to simultaneously determine these properties for single cells, using fluidic force microscopy. For MCF-7 cells grown on tissue-culture-treated polystyrene surfaces, we found that the adhesive force and adhesion energy were correlated for each cell. Well-spread cells tended to have stronger adhesion, which may be due to the greater area of the contact between cellular adhesion receptors and the surface. By contrast, the viscoelastic properties of MCF-7 cells cultured on the same surface appeared to have little dependence on cell shape. This methodology provides an integrated approach to better understand the biophysics of multiple cell types.
Collapse
Affiliation(s)
- Ma Luo
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K
| | - Wenjian Yang
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K
- Research Center for Intelligent Sensing Systems, Zhijiang Laboratory, Hangzhou 311100, China
| | - Tyrell N Cartwright
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
| | - Jonathan M G Higgins
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, U.K
| | - Jinju Chen
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K
| |
Collapse
|
31
|
Schoenit A, Lo Giudice C, Hahnen N, Ollech D, Jahnke K, Göpfrich K, Cavalcanti-Adam EA. Tuning Epithelial Cell-Cell Adhesion and Collective Dynamics with Functional DNA-E-Cadherin Hybrid Linkers. NANO LETTERS 2022; 22:302-310. [PMID: 34939414 PMCID: PMC8759084 DOI: 10.1021/acs.nanolett.1c03780] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/12/2021] [Indexed: 06/14/2023]
Abstract
The binding strength between epithelial cells is crucial for tissue integrity, signal transduction and collective cell dynamics. However, there is no experimental approach to precisely modulate cell-cell adhesion strength at the cellular and molecular level. Here, we establish DNA nanotechnology as a tool to control cell-cell adhesion of epithelial cells. We designed a DNA-E-cadherin hybrid system consisting of complementary DNA strands covalently bound to a truncated E-cadherin with a modified extracellular domain. DNA sequence design allows to tune the DNA-E-cadherin hybrid molecular binding strength, while retaining its cytosolic interactions and downstream signaling capabilities. The DNA-E-cadherin hybrid facilitates strong and reversible cell-cell adhesion in E-cadherin deficient cells by forming mechanotransducive adherens junctions. We assess the direct influence of cell-cell adhesion strength on intracellular signaling and collective cell dynamics. This highlights the scope of DNA nanotechnology as a precision technology to study and engineer cell collectives.
Collapse
Affiliation(s)
- Andreas Schoenit
- Biophysical
Engineering Group, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Department
of Cellular Biophysics, Growth Factor Mechanobiology Group, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
| | - Cristina Lo Giudice
- Department
of Cellular Biophysics, Growth Factor Mechanobiology Group, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
| | - Nina Hahnen
- Biophysical
Engineering Group, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Department
of Cellular Biophysics, Growth Factor Mechanobiology Group, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
| | - Dirk Ollech
- Department
of Cellular Biophysics, Growth Factor Mechanobiology Group, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
| | - Kevin Jahnke
- Biophysical
Engineering Group, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Department
of Physics and Astronomy, Heidelberg University, D-69120 Heidelberg, Germany
| | - Kerstin Göpfrich
- Biophysical
Engineering Group, Max Planck Institute
for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
- Department
of Physics and Astronomy, Heidelberg University, D-69120 Heidelberg, Germany
| | - Elisabetta Ada Cavalcanti-Adam
- Department
of Cellular Biophysics, Growth Factor Mechanobiology Group, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120 Heidelberg, Germany
| |
Collapse
|
32
|
Parreira P, Martins MCL. The biophysics of bacterial infections: Adhesion events in the light of force spectroscopy. Cell Surf 2021; 7:100048. [PMID: 33665520 PMCID: PMC7898176 DOI: 10.1016/j.tcsw.2021.100048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 08/10/2020] [Accepted: 12/03/2020] [Indexed: 02/08/2023] Open
Abstract
Bacterial infections are the most eminent public health challenge of the 21st century. The primary step leading to infection is bacterial adhesion to the surface of host cells or medical devices, which is mediated by a multitude of molecular interactions. At the interface of life sciences and physics, last years advances in atomic force microscopy (AFM)-based force spectroscopy techniques have made possible to measure the forces driving bacteria-cell and bacteria-materials interactions on a single molecule/cell basis (single molecule/cell force spectroscopy). Among the bacteria-(bio)materials surface interactions, the life-threatening infections associated to medical devices involving Staphylococcus aureus and Escherichia coli are the most eminent. On the other hand, Pseudomonas aeruginosa binding to the pulmonary and urinary tract or the Helicobacter pylori binding to the gastric mucosa, are classical examples of bacteria-host cell interactions that end in serious infections. As we approach the end of the antibiotic era, acquisition of a deeper knowledge of the fundamental forces involved in bacteria - host cells/(bio)materials surface adhesion is crucial for the identification of new ligand-binding events and its assessment as novel targets for alternative anti-infective therapies. This article aims to highlight the potential of AFM-based force spectroscopy for new targeted therapies development against bacterial infections in which adhesion plays a pivotal role and does not aim to be an extensive overview on the AFM technical capabilities and theory of single molecule force spectroscopy.
Collapse
Affiliation(s)
- Paula Parreira
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Portugal
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - M. Cristina L. Martins
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Portugal
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| |
Collapse
|
33
|
Nishida K, Anada T, Kobayashi S, Ueda T, Tanaka M. Effect of bound water content on cell adhesion strength to water-insoluble polymers. Acta Biomater 2021; 134:313-324. [PMID: 34332104 DOI: 10.1016/j.actbio.2021.07.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
Adhesion of cells on biomaterials plays an essential role in modulating cellular functions. Although hydration of biomaterials occurs under biological conditions, it is challenging to systematically evaluate the correlation of hydrated water content in biomaterials with the cell adhesion strength. In this report, we investigated the effect of bound water content on the adhesion strength of cells on poly(2-methoxyethyl acrylate) (PMEA) analogue substrates. Water-insoluble PMEA analogues were synthesized to fabricate substrates with a systemically controlled bound water content. To assess the surface properties of their substrates, contact angle measurement, atomic force microscopy (AFM), and fluorescence measurement were conducted. To reflect the effect of bound water of PMEA analogues, the relationship between the bound water content and cell adhesion behavior was evaluated under serum-free condition. From the single cell force spectrometry (SCFS) and microscopic analysis, it revealed that the increment of bound water content on the substrates decreased cell adhesion strength and cell spreading on the substrates. The bound water content exhibited a good correlation with adhesion strength, spreading area, circularity, and aspect ratio of cells. Our findings indicate that the bound water content could contribute to the development of a novel biomaterial and evaluation of cell behaviors on biomaterials. STATEMENT OF SIGNIFICANCE: For coordinating cell functions, such as growth, mobility, and differentiation, modulating the adhesion strength between cells and their environments is important. Although the hydration to biomaterials has been reported to be closely related to a antifouling property, the effect of hydration water on the cell adhesion behavior is not well understood. We present the first demonstration of essential relationship between cell adhesion strength and hydrated water on a biomaterials surface using the water-insoluble polymers with different hydrated water content. The results reveal that the hydrated water content of polymer substrates strong correlation with adhesion strength of cells. Collectively, the hydrated water content of the biomaterials will be a predominant factor affecting the cell adhesion strength and behavior.
Collapse
|
34
|
Fourier Transform Infrared Microspectroscopy Combined with Principal Component Analysis and Artificial Neural Networks for the Study of the Effect of β-Hydroxy-β-Methylbutyrate (HMB) Supplementation on Articular Cartilage. Int J Mol Sci 2021; 22:ijms22179189. [PMID: 34502096 PMCID: PMC8430473 DOI: 10.3390/ijms22179189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022] Open
Abstract
The potential of Fourier Transform infrared microspectroscopy (FTIR microspectroscopy) and multivariate analyses were applied for the classification of the frequency ranges responsible for the distribution changes of the main components of articular cartilage (AC) that occur during dietary β-hydroxy-β-methyl butyrate (HMB) supplementation. The FTIR imaging analysis of histological AC sections originating from 35-day old male piglets showed the change in the collagen and proteoglycan contents of the HMB-supplemented group compared to the control. The relative amount of collagen content in the superficial zone increased by more than 23% and in the middle zone by about 17%, while no changes in the deep zone were observed compared to the control group. Considering proteoglycans content, a significant increase was registered in the middle and deep zones, respectively; 62% and 52% compared to the control. AFM nanoindentation measurements collected from animals administered with HMB displayed an increase in AC tissue stiffness by detecting a higher value of Young’s modulus in all investigated AC zones. We demonstrated that principal component analysis and artificial neural networks could be trained with spectral information to distinguish AC histological sections and the group under study accurately. This work may support the use and effectiveness of FTIR imaging combined with multivariate analyses as a quantitative alternative to traditional collagenous tissue-related histology.
Collapse
|
35
|
Nouri-Goushki M, Angeloni L, Modaresifar K, Minneboo M, Boukany PE, Mirzaali MJ, Ghatkesar MK, Fratila-Apachitei LE, Zadpoor AA. 3D-Printed Submicron Patterns Reveal the Interrelation between Cell Adhesion, Cell Mechanics, and Osteogenesis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:33767-33781. [PMID: 34250808 PMCID: PMC8323101 DOI: 10.1021/acsami.1c03687] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The surface topography of implantable devices is of crucial importance for guiding the cascade of events that starts from the initial contact of the cells with the surface and continues until the complete integration of the device in its immediate environment. There is, however, limited quantitative information available regarding the relationships between the different stages of such cascade(s) and how the design of surface topography influences them. We, therefore, used direct laser writing to 3D-print submicron pillars with precisely controlled dimensions and spatial arrangements to perform a systematic study of such relationships. Using single-cell force spectroscopy, we measured the adhesion force and the work of adhesion of the preosteoblast cells residing on the different types of surfaces. Not only the adhesion parameters (after 2-60 s) but also the formation of focal adhesions was strongly dependent on the geometry and arrangement of the pillars: sufficiently tall and dense pillars enhanced both adhesion parameters and the formation of focal adhesions. Our morphological study of the cells (after 24 h) showed that those enhancements were associated with a specific way of cell settlement onto the surface (i.e., "top state"). The cells interacting with tall and dense pillars were also characterized by numerous thick actin stress fibers in the perinuclear region and possibly high internal stresses. Furthermore, living cells with highly organized cytoskeletal networks exhibited greater values of the elastic modulus. The early responses of the cells predicted their late response including matrix mineralization: tall and dense submicron pillars significantly upregulated the expression of osteopontin after 21 days of culture under both osteogenic and nonosteogenic conditions. Our findings paint a detailed picture of at least one possible cascade of events that starts from initial cell adhesion and continues to subsequent cellular functions and eventual matrix mineralization. These observations could inform the future developments of instructive surfaces for medical devices based on physical surface cues and early markers.
Collapse
Affiliation(s)
- Mahdiyeh Nouri-Goushki
- Department
of Biomechanical Engineering, Faculty of Mechanical, Maritime, and
Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
- ,
| | - Livia Angeloni
- Department
of Biomechanical Engineering, Faculty of Mechanical, Maritime, and
Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
- Department
of Precision and Microsystems Engineering, Faculty of Mechanical,
Maritime, and Materials Engineering, Delft
University of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
- . Phone: +31-152786980
| | - Khashayar Modaresifar
- Department
of Biomechanical Engineering, Faculty of Mechanical, Maritime, and
Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Michelle Minneboo
- Department
of Biomechanical Engineering, Faculty of Mechanical, Maritime, and
Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Pouyan E. Boukany
- Department
of Chemical Engineering, Delft University
of Technology (TU Delft), van der Maasweg 9, 2629 HZ Delft, The Netherlands
| | - Mohammad J. Mirzaali
- Department
of Biomechanical Engineering, Faculty of Mechanical, Maritime, and
Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Murali K. Ghatkesar
- Department
of Precision and Microsystems Engineering, Faculty of Mechanical,
Maritime, and Materials Engineering, Delft
University of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Lidy E. Fratila-Apachitei
- Department
of Biomechanical Engineering, Faculty of Mechanical, Maritime, and
Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Amir A. Zadpoor
- Department
of Biomechanical Engineering, Faculty of Mechanical, Maritime, and
Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| |
Collapse
|
36
|
Characterization of the strain-rate-dependent mechanical response of single cell-cell junctions. Proc Natl Acad Sci U S A 2021; 118:2019347118. [PMID: 33531347 DOI: 10.1073/pnas.2019347118] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cell-cell adhesions are often subjected to mechanical strains of different rates and magnitudes in normal tissue function. However, the rate-dependent mechanical behavior of individual cell-cell adhesions has not been fully characterized due to the lack of proper experimental techniques and therefore remains elusive. This is particularly true under large strain conditions, which may potentially lead to cell-cell adhesion dissociation and ultimately tissue fracture. In this study, we designed and fabricated a single-cell adhesion micro tensile tester (SCAµTT) using two-photon polymerization and performed displacement-controlled tensile tests of individual pairs of adherent epithelial cells with a mature cell-cell adhesion. Straining the cytoskeleton-cell adhesion complex system reveals a passive shear-thinning viscoelastic behavior and a rate-dependent active stress-relaxation mechanism mediated by cytoskeleton growth. Under low strain rates, stress relaxation mediated by the cytoskeleton can effectively relax junctional stress buildup and prevent adhesion bond rupture. Cadherin bond dissociation also exhibits rate-dependent strengthening, in which increased strain rate results in elevated stress levels at which cadherin bonds fail. This bond dissociation becomes a synchronized catastrophic event that leads to junction fracture at high strain rates. Even at high strain rates, a single cell-cell junction displays a remarkable tensile strength to sustain a strain as much as 200% before complete junction rupture. Collectively, the platform and the biophysical understandings in this study are expected to build a foundation for the mechanistic investigation of the adaptive viscoelasticity of the cell-cell junction.
Collapse
|
37
|
Wanuske M, Brantschen D, Schinner C, Stüdle C, Walter E, Hiermaier M, Vielmuth F, Waschke J, Spindler V. Clustering of desmosomal cadherins by desmoplakin is essential for cell-cell adhesion. Acta Physiol (Oxf) 2021; 231:e13609. [PMID: 33354837 DOI: 10.1111/apha.13609] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/14/2022]
Abstract
AIM Desmoplakin (Dp) is a crucial component of the desmosome, a supramolecular cell junction complex anchoring intermediate filaments. The mechanisms how Dp modulates cell-cell adhesion are only partially understood. Here, we studied the impact of Dp on the function of desmosomal adhesion molecules, desmosome turnover and intercellular adhesion. METHODS CRISPR/Cas9 was used for gene editing of human keratinocytes which were characterized by Western blot and immunostaining. Desmosomal ultrastructure and function were assessed by electron microscopy and cell adhesion assays. Single molecule binding properties and localization of desmosomal cadherins were studied by atomic force microscopy and super-resolution imaging. RESULTS Knockout (ko) of Dp impaired cell cohesion to drastically higher extents as ko of another desmosomal protein, plakoglobin (Pg). In contrast to Pg ko, desmosomes were completely absent in Dp ko. Binding properties of the desmosomal adhesion molecules desmocollin (Dsc) 3 and desmoglein (Dsg) 3 remained unaltered under loss of Dp. Dp was required for assembling desmosomal cadherins into large clusters, as Dsg2 and Dsc3, adhesion molecules primarily localized within desmosomes, were redistributed into small puncta in the cell membrane of Dp ko cells. Additional silencing of desmosomal cadherins in Dp ko did not further increase loss of intercellular adhesion. CONCLUSION Our data demonstrate that Dp is essential for desmosome formation but does not influence intercellular adhesion on the level of individual cadherin binding properties. Rather, macro-clustering of desmosomal adhesion molecules through Dp is crucial. These results may help to better understand severe diseases which are caused by Dp dysfunction.
Collapse
Affiliation(s)
- Marie‐Therès Wanuske
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | | | - Camilla Schinner
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Chiara Stüdle
- Department of Biomedicine University of Basel Basel Switzerland
| | - Elias Walter
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Matthias Hiermaier
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Franziska Vielmuth
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Jens Waschke
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Volker Spindler
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| |
Collapse
|
38
|
Li M, Xi N, Wang YC, Liu LQ. Atomic force microscopy for revealing micro/nanoscale mechanics in tumor metastasis: from single cells to microenvironmental cues. Acta Pharmacol Sin 2021; 42:323-339. [PMID: 32807839 PMCID: PMC8027022 DOI: 10.1038/s41401-020-0494-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023]
Abstract
Mechanics are intrinsic properties which appears throughout the formation, development, and aging processes of biological systems. Mechanics have been shown to play important roles in regulating the development and metastasis of tumors, and understanding tumor mechanics has emerged as a promising way to reveal the underlying mechanisms guiding tumor behaviors. In particular, tumors are highly complex diseases associated with multifaceted factors, including alterations in cancerous cells, tissues, and organs as well as microenvironmental cues, indicating that investigating tumor mechanics on multiple levels is significantly helpful for comprehensively understanding the effects of mechanics on tumor progression. Recently, diverse techniques have been developed for probing the mechanics of tumors, among which atomic force microscopy (AFM) has appeared as an excellent platform enabling simultaneously characterizing the structures and mechanical properties of living biological systems ranging from individual molecules and cells to tissue samples with unprecedented spatiotemporal resolution, offering novel possibilities for understanding tumor physics and contributing much to the studies of cancer. In this review, we survey the recent progress that has been achieved with the use of AFM for revealing micro/nanoscale mechanics in tumor development and metastasis. Challenges and future progress are also discussed.
Collapse
Affiliation(s)
- Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Ning Xi
- Department of Industrial and Manufacturing Systems Engineering, The University of Hong Kong, Hong Kong, China
| | - Yue-Chao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lian-Qing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
39
|
Omidvar R, Ayala YA, Brandel A, Hasenclever L, Helmstädter M, Rohrbach A, Römer W, Madl J. Quantification of nanoscale forces in lectin-mediated bacterial attachment and uptake into giant liposomes. NANOSCALE 2021; 13:4016-4028. [PMID: 33503085 DOI: 10.1039/d0nr07726g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Interactions of the bacterial lectin LecA with the host cells glycosphingolipid Gb3 have been shown to be crucial for the cellular uptake of the bacterium Pseudomonas aeruginosa. LecA-induced Gb3 clustering, referred to as lipid zipper mechanism, leads to full membrane engulfment of the bacterium. Here, we aim for a nanoscale force characterization of this mechanism using two complementary force probing techniques, atomic force microscopy (AFM) and optical tweezers (OT). The LecA-Gb3 interactions are reconstituted using giant unilamellar vesicles (GUVs), a well-controlled minimal system mimicking the plasma membrane and nanoscale forces between either bacteria (PAO1 wild-type and LecA-deletion mutant strains) or LecA-coated probes (as minimal, synthetic bacterial model) and vesicles are measured. LecA-Gb3 interactions strengthen the bacterial attachment to the membrane (1.5-8-fold) depending on the membrane tension and the applied technique. Moreover, significantly less energy (reduction up to 80%) is required for the full uptake of LecA-coated beads into Gb3-functionalized vesicles. This quantitative approach highlights that lectin-glycolipid interactions provide adequate forces and energies to drive bacterial attachment and uptake.
Collapse
Affiliation(s)
- Ramin Omidvar
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany. and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany and Freiburg Center for Interactive Materials and Bioinspired Technologies (FIT), University of Freiburg, Georges-Köhler-Allee 105, 79110 Freiburg, Germany
| | - Yareni A Ayala
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Georges-Köhler-Allee 105, 79110 Freiburg, Germany.
| | - Annette Brandel
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany. and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Lukas Hasenclever
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany. and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Martin Helmstädter
- Renal Division, Department of Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Freiburg, Germany
| | - Alexander Rohrbach
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Georges-Köhler-Allee 105, 79110 Freiburg, Germany.
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany. and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany and Freiburg Center for Interactive Materials and Bioinspired Technologies (FIT), University of Freiburg, Georges-Köhler-Allee 105, 79110 Freiburg, Germany
| | - Josef Madl
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany. and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany and Freiburg Center for Interactive Materials and Bioinspired Technologies (FIT), University of Freiburg, Georges-Köhler-Allee 105, 79110 Freiburg, Germany
| |
Collapse
|
40
|
Abu Quba AA, Schaumann GE, Karagulyan M, Diehl D. Quality control of direct cell–mineral adhesion measurements in air and liquid using inverse AFM imaging. RSC Adv 2021; 11:5384-5392. [PMID: 35423094 PMCID: PMC8694684 DOI: 10.1039/d1ra00110h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/22/2021] [Indexed: 12/03/2022] Open
Abstract
The study of interaction forces between biological and non-living systems requires in-house production of probes modified with, e.g., bacterial cells or with minerals, in order to map irregularly shaped natural surfaces. In order to avoid artifacts, it is essential to control the functionality of the modified probes. Current methods for this purpose require removing the modified probe from the liquid-cell, inserting it into another device and/or have a too low resolution to detect local changes within the interacting areas. Therefore, we present a fast and cost-effective method that overcomes the above mentioned problems by the inverse AFM imaging principle. First, the 3-D shape of a fresh sharp AFM tip is modeled by measuring the shape of a standard rough pattern and post blind tip reconstruction analysis. The so calibrated characterizer tip was extracted and upside-down fixed rigidly on a disc together with the sample. Before and after the cell–mineral interaction, the modified probe is then inversely imaged by the fixed characterizer controlling changes in finest 3-D details of the modified probe. The characterization of probes modified with kaolinite and P. fluorescens cells and their interactions with R. erythropolis and montmorillonite samples show that the method allows a fast precise investigation of tip modifications before and after cell–mineral interactions in air and liquid such that artifacts in adhesion between cell and mineral at the single-cell level can be excluded. Setup for a reliable cell-mineral interaction at the single-cell level, (a) study of the mineral by a sharp tip, (b) study of the bacterial modified probe by a characterizer, (c) cell-mineral interaction, (d) subsequent check of the modified probe.![]()
Collapse
Affiliation(s)
- Abd Alaziz Abu Quba
- Environmental and Soil Chemistry Group
- iES Institute for Environmental Sciences
- University of Koblenz-Landau
- 76829 Landau
- Germany
| | - Gabriele E. Schaumann
- Environmental and Soil Chemistry Group
- iES Institute for Environmental Sciences
- University of Koblenz-Landau
- 76829 Landau
- Germany
| | - Mariam Karagulyan
- Department of Environmental Biotechnology
- Helmholtz Centre for Environmental Research – UFZ
- Leipzig
- Germany
| | - Doerte Diehl
- Environmental and Soil Chemistry Group
- iES Institute for Environmental Sciences
- University of Koblenz-Landau
- 76829 Landau
- Germany
| |
Collapse
|
41
|
Cheirdaris DG. Force Spectroscopy in Mechanical Protein Domains Unfolding. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1339:187-193. [DOI: 10.1007/978-3-030-78787-5_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
42
|
Maynard S, Gelmi A, Skaalure SC, Pence IJ, Lee-Reeves C, Sero JE, Whittaker TE, Stevens MM. Nanoscale Molecular Quantification of Stem Cell-Hydrogel Interactions. ACS NANO 2020; 14:17321-17332. [PMID: 33215498 PMCID: PMC7760213 DOI: 10.1021/acsnano.0c07428] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/17/2020] [Indexed: 05/07/2023]
Abstract
A common approach to tailoring synthetic hydrogels for regenerative medicine applications involves incorporating RGD cell adhesion peptides, yet assessing the cellular response to engineered microenvironments at the nanoscale remains challenging. To date, no study has demonstrated how RGD concentration in hydrogels affects the presentation of individual cell surface receptors. Here we studied the interaction between human mesenchymal stem cells (hMSCs) and RGD-functionalized poly(ethylene glycol) hydrogels, by correlating macro- and nanoscale single-cell interfacial quantification techniques. We quantified RGD unbinding forces on a synthetic hydrogel using single cell atomic force spectroscopy, revealing that short-term binding of hMSCs was sensitive to RGD concentration. We also performed direct stochastic optical reconstruction microscopy (dSTORM) to quantify the molecular interactions between integrin α5β1 and a biomaterial, unexpectedly revealing that increased integrin clustering at the hydrogel-cell interface correlated with fewer available RGD binding sites. Our complementary, quantitative approach uncovered mechanistic insights into specific stem cell-hydrogel interactions, where dSTORM provides nanoscale sensitivity to RGD-dependent differences in cell surface localization of integrin α5β1. Our findings reveal that it is possible to precisely determine how peptide-functionalized hydrogels interact with cells at the molecular scale, thus providing a basis to fine-tune the spatial presentation of bioactive ligands.
Collapse
Affiliation(s)
| | | | - Stacey C. Skaalure
- Department of Materials,
Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Isaac J. Pence
- Department of Materials,
Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Charlotte Lee-Reeves
- Department of Materials,
Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | | | - Thomas E. Whittaker
- Department of Materials,
Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Molly M. Stevens
- Department of Materials,
Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, United Kingdom
| |
Collapse
|
43
|
Benito-Jardón M, Strohmeyer N, Ortega-Sanchís S, Bharadwaj M, Moser M, Müller DJ, Fässler R, Costell M. αv-Class integrin binding to fibronectin is solely mediated by RGD and unaffected by an RGE mutation. J Cell Biol 2020; 219:e202004198. [PMID: 33141174 PMCID: PMC7644020 DOI: 10.1083/jcb.202004198] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/20/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Fibronectin (FN) is an essential glycoprotein of the extracellular matrix; binds integrins, syndecans, collagens, and growth factors; and is assembled by cells into complex fibrillar networks. The RGD motif in FN facilitates cell binding- and fibrillogenesis through binding to α5β1 and αv-class integrins. However, whether RGD is the sole binding site for αv-class integrins is unclear. Most notably, substituting aspartate with glutamate (RGE) was shown to eliminate integrin binding in vitro, while mouse genetics revealed that FNRGE preserves αv-class integrin binding and fibrillogenesis. To address this conflict, we employed single-cell force spectroscopy, engineered cells, and RGD motif-deficient mice (Fn1ΔRGD/ΔRGD) to search for additional αv-class integrin-binding sites. Our results demonstrate that α5β1 and αv-class integrins solely recognize the FN-RGD motif and that αv-class, but not α5β1, integrins retain FN-RGE binding. Furthermore, Fn1ΔRGD/ΔRGD tissues and cells assemble abnormal and dysfunctional FNΔRGD fibrils in a syndecan-dependent manner. Our data highlight the central role of FN-RGD and the functionality of FN-RGE for αv-class integrins.
Collapse
Affiliation(s)
- María Benito-Jardón
- Department of Biochemistry and Molecular Biology, Universitat de València, Burjassot, Spain
- Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| | - Nico Strohmeyer
- Eidgenössische Technische Hochschule Zürich, Basel, Switzerland
| | - Sheila Ortega-Sanchís
- Department of Biochemistry and Molecular Biology, Universitat de València, Burjassot, Spain
- Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| | | | - Markus Moser
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | | | - Mercedes Costell
- Department of Biochemistry and Molecular Biology, Universitat de València, Burjassot, Spain
- Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| |
Collapse
|
44
|
Nie Y, Xu X, Wang W, Ma N, Lendlein A. Spheroid formation of human keratinocyte: Balancing between cell-substrate and cell-cell interaction. Clin Hemorheol Microcirc 2020; 76:329-340. [PMID: 32925021 DOI: 10.3233/ch-209217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The formation of spheroids is tightly regulated by intrinsic cell-cell and cell-substrate interactions. OBJECTIVE The chitosan (CS)-coating was applied to investigate the driven force directed the spheroid formation. METHODS The effects of CS on cell functions were studied. Atomic force microscopy was employed to measure the cell- biomaterial interplay at single cell level. RESULTS HaCaT cells shifted from their flattened sheet to a compact 3D spheroidal morphology when increasing CS-coating concentration. The proliferative capacity of HaCaT was preserved in the spheroid. The expression and activation of integrin β1 (ITGB1) were enhanced on CS modified surfaces, while the active to total ratio of ITGB1 was decreased. The adhesive force of a single HaCaT cell to the tissue culture plate (TCP) was 4.84±0.72 nN. It decreased on CS-coated surfaces as CS concentration increased, from 2.16±0.26 nN to 0.96±0.17 nN. The adhesive force between the single HaCaT cell to its neighbor cell increased as CS concentration increased, from 1.15±0.09 nN to 2.60±0.51 nN. CONCLUSIONS Conclusively, the decreased cell- substrate adhesion was the main driven force in the spheroid formation. This finding might serve as a design criterion for biomaterials facilitating the formation of epithelial spheroids.
Collapse
Affiliation(s)
- Yan Nie
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
45
|
Hiermaier M, Kliewe F, Schinner C, Stüdle C, Maly IP, Wanuske MT, Rötzer V, Endlich N, Vielmuth F, Waschke J, Spindler V. The Actin-Binding Protein α-Adducin Modulates Desmosomal Turnover and Plasticity. J Invest Dermatol 2020; 141:1219-1229.e11. [PMID: 33098828 DOI: 10.1016/j.jid.2020.09.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 01/01/2023]
Abstract
Intercellular adhesion is essential for tissue integrity and homeostasis. Desmosomes are abundant in the epidermis and the myocardium-tissues, which are under constantly changing mechanical stresses. Yet, it is largely unclear whether desmosomal adhesion can be rapidly adapted to changing demands, and the mechanisms underlying desmosome turnover are only partially understood. In this study we show that the loss of the actin-binding protein α-adducin resulted in reduced desmosome numbers and prevented the ability of cultured keratinocytes or murine epidermis to withstand mechanical stress. This effect was not primarily caused by decreased levels or impaired adhesive properties of desmosomal molecules but rather by altered desmosome turnover. Mechanistically, reduced cortical actin density in α-adducin knockout keratinocytes resulted in increased mobility of the desmosomal adhesion molecule desmoglein 3 and impaired interactions with E-cadherin, a crucial step in desmosome formation. Accordingly, the loss of α-adducin prevented increased membrane localization of desmoglein 3 in response to cyclic stretch or shear stress. Our data demonstrate the plasticity of desmosomal molecules in response to mechanical stimuli and unravel a mechanism of how the actin cytoskeleton indirectly shapes intercellular adhesion by restricting the membrane mobility of desmosomal molecules.
Collapse
Affiliation(s)
- Matthias Hiermaier
- Department of Biomedicine, University of Basel, Basel, Switzerland; Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Camilla Schinner
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Chiara Stüdle
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - I Piotr Maly
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marie-Therès Wanuske
- Department of Biomedicine, University of Basel, Basel, Switzerland; Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Vera Rötzer
- Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Franziska Vielmuth
- Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Jens Waschke
- Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland; Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| |
Collapse
|
46
|
Huang H, Dai C, Shen H, Gu M, Wang Y, Liu J, Chen L, Sun L. Recent Advances on the Model, Measurement Technique, and Application of Single Cell Mechanics. Int J Mol Sci 2020; 21:E6248. [PMID: 32872378 PMCID: PMC7504142 DOI: 10.3390/ijms21176248] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since the cell was discovered by humans, it has been an important research subject for researchers. The mechanical response of cells to external stimuli and the biomechanical response inside cells are of great significance for maintaining the life activities of cells. These biomechanical behaviors have wide applications in the fields of disease research and micromanipulation. In order to study the mechanical behavior of single cells, various cell mechanics models have been proposed. In addition, the measurement technologies of single cells have been greatly developed. These models, combined with experimental techniques, can effectively explain the biomechanical behavior and reaction mechanism of cells. In this review, we first introduce the basic concept and biomechanical background of cells, then summarize the research progress of internal force models and experimental techniques in the field of cell mechanics and discuss the latest mechanical models and experimental methods. We summarize the application directions of cell mechanics and put forward the future perspectives of a cell mechanics model.
Collapse
Affiliation(s)
| | | | | | | | | | - Jizhu Liu
- School of Mechanical and Electric Engineering, Jiangsu Provincial Key Laboratory of Advanced Robotics, Soochow University, Suzhou 215123, China; (H.H.); (C.D.); (H.S.); (M.G.); (Y.W.); (L.S.)
| | - Liguo Chen
- School of Mechanical and Electric Engineering, Jiangsu Provincial Key Laboratory of Advanced Robotics, Soochow University, Suzhou 215123, China; (H.H.); (C.D.); (H.S.); (M.G.); (Y.W.); (L.S.)
| | | |
Collapse
|
47
|
Alunda BO, Lee YJ. Review: Cantilever-Based Sensors for High Speed Atomic Force Microscopy. SENSORS (BASEL, SWITZERLAND) 2020; 20:E4784. [PMID: 32854193 PMCID: PMC7506678 DOI: 10.3390/s20174784] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022]
Abstract
This review critically summarizes the recent advances of the microcantilever-based force sensors for atomic force microscope (AFM) applications. They are one the most common mechanical spring-mass systems and are extremely sensitive to changes in the resonant frequency, thus finding numerous applications especially for molecular sensing. Specifically, we comment on the latest progress in research on the deflection detection systems, fabrication, coating and functionalization of the microcantilevers and their application as bio- and chemical sensors. A trend on the recent breakthroughs on the study of biological samples using high-speed atomic force microscope is also reported in this review.
Collapse
Affiliation(s)
- Bernard Ouma Alunda
- School of Mines and Engineering, Taita Taveta University, P.O. Box 635-80300 Voi, Kenya;
| | - Yong Joong Lee
- School of Mechanical Engineering, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
48
|
Obeid S, Guyomarc'h F. Atomic force microscopy of food assembly: Structural and mechanical insights at the nanoscale and potential opportunities from other fields. FOOD BIOSCI 2020. [DOI: 10.1016/j.fbio.2020.100654] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
49
|
Kim H, Witt H, Oswald TA, Tarantola M. Adhesion of Epithelial Cells to PNIPAm Treated Surfaces for Temperature-Controlled Cell-Sheet Harvesting. ACS APPLIED MATERIALS & INTERFACES 2020; 12:33516-33529. [PMID: 32631046 PMCID: PMC7467562 DOI: 10.1021/acsami.0c09166] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Stimuli responsive polymer coatings are a common motive for designing surfaces for cell biological applications. In the present study, we have characterized temperature dependent adhesive properties of poly(N-isopropylacrylamide) (PNIPAm) microgel coated surfaces (PMS) using various atomic force microscopy based approaches. We imaged and quantified the material properties of PMS upon a temperature switch using quantitative AFM imaging but also employed single-cell force spectroscopy (SCFS) before and after decreasing the temperature to assess the forces and work of initial adhesion between cells and PMS. We performed a detailed analysis of steps in the force-distance curves. Finally, we applied colloid probe atomic force microscopy (CP-AFM) to analyze the adhesive properties of two major components of the extracellular matrix to PMS under temperature control, namely collagen I and fibronectin. In combination with confocal imaging, we could show that these two ECM components differ in their detachment properties from PNIPAm microgel films upon cell harvesting, and thus gained a deeper understanding of cell-sheet maturation and harvesting process and the involved partial ECM dissolution.
Collapse
Affiliation(s)
- Hyejeong Kim
- Max Planck Institute
for Dynamics and Self Organization (MPIDS), Am Fassberg 17, 37077 Göttingen, Germany
| | - Hannes Witt
- Max Planck Institute
for Dynamics and Self Organization (MPIDS), Am Fassberg 17, 37077 Göttingen, Germany
| | - Tabea A. Oswald
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Tammannstrasse 2, 37077 Göttingen, Germany
| | - Marco Tarantola
- Max Planck Institute
for Dynamics and Self Organization (MPIDS), Am Fassberg 17, 37077 Göttingen, Germany
- Institute for Dynamics of Complex Systems, University of Göttingen, Friedrich-Hund Platz 1, 37073 Göttingen, Germany
- E-mail: . Phone: +49-551-5176-316
| |
Collapse
|
50
|
Chighizola M, Previdi A, Dini T, Piazzoni C, Lenardi C, Milani P, Schulte C, Podestà A. Adhesion force spectroscopy with nanostructured colloidal probes reveals nanotopography-dependent early mechanotransductive interactions at the cell membrane level. NANOSCALE 2020; 12:14708-14723. [PMID: 32618323 DOI: 10.1039/d0nr01991g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Mechanosensing, the ability of cells to perceive and interpret the microenvironmental biophysical cues (such as the nanotopography), impacts strongly cellular behaviour through mechanotransductive processes and signalling. These events are predominantly mediated by integrins, the principal cellular adhesion receptors located at the cell/extracellular matrix (ECM) interface. Because of the typical piconewton force range and nanometre length scale of mechanotransductive interactions, achieving a detailed understanding of the spatiotemporal dynamics occurring at the cell/microenvironment interface is challenging; sophisticated interdisciplinary methodologies are required. Moreover, an accurate control over the nanotopographical features of the microenvironment is essential, in order to systematically investigate and precisely assess the influence of the different nanotopographical motifs on the mechanotransductive process. In this framework, we were able to study and quantify the impact of microenvironmental nanotopography on early cellular adhesion events by means of adhesion force spectroscopy based on innovative colloidal probes mimicking the nanotopography of natural ECMs. These probes provided the opportunity to detect nanotopography-specific modulations of the molecular clutch force loading dynamics and integrin clustering at the level of single binding events, in the critical time window of nascent adhesion formation. Following this approach, we found that the nanotopographical features are responsible for an excessive force loading in single adhesion sites after 20-60 s of interaction, causing a drop in the number of adhesion sites. However, by manganese treatment we demonstrated that the availability of activated integrins is a critical regulatory factor for these nanotopography-dependent dynamics.
Collapse
Affiliation(s)
- M Chighizola
- C.I.Ma.I.Na. and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, via Celoria 16, 20133 Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|