1
|
Zhou W, Li Y, Liu Y, Quan H, Li P, Li F, Niu L, Zheng H, Meng L. An acoustic squeezer for assessment of multiparameter cell mechanical properties. ULTRASONICS 2025; 151:107622. [PMID: 40068409 DOI: 10.1016/j.ultras.2025.107622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Cells' ability to sense and respond to mechanical stimuli is fundamental to various biological processes and serves as a crucial biomarker of their physiological and pathological states. Traditional methods for assessing cell mechanical properties, such as atomic force microscopy and micropipette aspiration, are hindered by complex procedures and the risk of cellular damage due to direct contact. Here we introduce a novel non-contact acoustic squeezer that leverages focused interdigital transducers to induce cell deformation through a robust standing surface acoustic wave (SSAW) field. This approach enables the multiparametric quantification of multiple mechanical properties, including elasticity (Young's modulus, stiffness) and viscosity, without requiring labeling or physical contact, providing a comprehensive understanding of the cell mechanical properties. Our acoustic squeezer is capable of generating a maximum squeezing force of 25.70 pN, inducing a deformability of 1.27 ± 0.017. Combined with thin-shell deformation model, the quantized Young's modulus of normal red blood cells (RBCs) is approximately 919.04 ± 55.64 Pa. Furthermore, our method demonstrates that cells treated with the anti-cancer drug (doxorubicin) exhibited reduced deformability, increased Young's modulus and viscosity. Our acoustic squeezer offers a standardized, non-invasive, and highly sensitive approach for characterizing cell mechanical properties, with significant promise for clinical applications in disease diagnosis and drug development.
Collapse
Affiliation(s)
- Wei Zhou
- Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, PR China
| | - Yingyin Li
- Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, PR China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110016, PR China
| | - Yifan Liu
- Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, PR China
| | - Hao Quan
- Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, PR China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110016, PR China
| | - Pengqi Li
- Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, PR China
| | - Fei Li
- Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, PR China
| | - Lili Niu
- Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, PR China
| | - Hairong Zheng
- Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, PR China
| | - Long Meng
- Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, PR China.
| |
Collapse
|
2
|
Huse M. Mechanoregulation of lymphocyte cytotoxicity. Nat Rev Immunol 2025:10.1038/s41577-025-01173-2. [PMID: 40312550 DOI: 10.1038/s41577-025-01173-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 05/03/2025]
Abstract
Cytotoxic lymphocytes counter intracellular pathogens and cancer by recognizing and destroying infected or transformed target cells. The basis for their function is the cytolytic immune synapse, a structurally stereotyped cell-cell interface through which lymphocytes deliver toxic proteins to target cells. The immune synapse is a highly dynamic contact capable of exerting nanonewton-scale forces against the target cell. In recent years, it has become clear that the interplay between these forces and the biophysical properties of the target influences the entirety of the cytotoxic response, from the initial activation of cytotoxic lymphocytes to the release of dying target cells. As a result, cellular cytotoxicity has become an exemplar of the ways in which biomechanics can regulate immune cell activation and effector function. This Review covers recent progress in this area, which has prompted a reconsideration of target cell killing from a more mechanobiological perspective.
Collapse
Affiliation(s)
- Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
3
|
Pustovalova M, Mohammad R, Wang Y, Xue W, Malakhov P, Nekrasov V, Kontareva E, Nofal Z, Saburov V, Kolesov D, Osipov A, Leonov S. High-LET Proton Irradiation Significantly Alters the Clonogenic and Tumorigenic Potential of Human Breast Cancer Cell Lines In Vitro and In Vivo. FRONT BIOSCI-LANDMRK 2025; 30:36415. [PMID: 40302350 DOI: 10.31083/fbl36415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/19/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND The implementation of proton beam irradiation (PBI) for breast cancer (BC) treatment is rapidly advancing due to its enhanced target coverage and reduced toxicities to organs at risk. However, the effects of PBI can vary depending on the cell type. This study aimed to explore the effects of PBI on two BC cell lines, MCF7 and MDA-MB-231. METHODS The relative biological effectiveness (RBE) of PBI was assessed using a clonogenic assay. DNA double-strand break (DSB) repair, epithelial-mesenchymal transition (EMT), and filamentous actin (F-actin) were evaluated using immunofluorescence analysis. The extent of entosis and the senescence-associated β-galactosidase (SA-β-gal) activity were estimated by cytochemistry analysis. The influence of the extracellular matrix was evaluated by cultivating cells in both adherent two-dimensional (2D) environments and within 3D fibrin gels of varying stiffness. The metastatic propensity of cells was investigated using migration tests and the cell encapsulation of carboxylate-modified fluorescent nanoparticles. The comparative tumorigenic potential of cells was investigated using an in vivo model of the chick embryo chorioallantoic membrane (CAM). RESULTS PBI demonstrated superior efficacy in eliminating MCF7 and MDA-MB-231 cells with RBE 1.7 and 1.75, respectively. Following PBI, MDA-MB-231 cells exhibited significantly lower clonogenic survival compared to MCF7, which was accompanied by the accumulation of phosphorylated histone H2AX (γH2AX), p53-binding protein 1 (53BP1) and Rad51 foci of DNA DSBs repair proteins. After surviving 7 days post-PBI, MCF7 cells exhibited 2.5-fold higher levels of the senescence phenotype and entosis compared to the MDA-MB-231 offspring. Both PBI-survived cell lines had greater capability for 2D collective migration, but their metastatic potential was significantly reduced. A significant influence of extracellular matrix stiffness on the correlation between F-actin expression in PBI-survived cells-an indicator of cell stiffness-and their ability to uptake nanoparticles, a trait associated with metastatic potential, was observed. PBI-survived MDA-MB-231RP subline exhibited a hybrid EMT phenotype and a 70% reduction in tumor growth in the in vivo model of the chick embryo CAM. In contrast, PBI-survived MCF7RP cells exhibit mesenchymal-to-epithelial transition (MET)-like features, and their in vivo tumor growth increased by 66% compared to parental cells. CONCLUSIONS PBI triggers various cellular responses in different BC cell lines, influencing tumor growth through mechanisms like DNA damage repair, stress-induced premature senescence (SIPS), and alterations in the stiffness of tumor cell membranes. Our insights into entosis and the effect of extracellular matrix stiffness on metastatic propensity (nanoparticle uptake) enhance the understanding of the role of PBI in BC cells, emphasizing the need for more research to optimize its therapeutic application.
Collapse
Affiliation(s)
- Margarita Pustovalova
- Institute of Future Biophysics, 141701 Dolgoprudny, Russia
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123098 Moscow, Russia
| | - Rita Mohammad
- Institute of Future Biophysics, 141701 Dolgoprudny, Russia
| | - Yuzhe Wang
- Institute of Future Biophysics, 141701 Dolgoprudny, Russia
| | - Wenyu Xue
- Institute of Future Biophysics, 141701 Dolgoprudny, Russia
| | | | | | | | - Zain Nofal
- Institute of Future Biophysics, 141701 Dolgoprudny, Russia
| | - Vyacheslav Saburov
- A. Tsyb Medical Radiological Research Center-Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 249031 Obninsk, Russia
| | - Dmitry Kolesov
- Laboratory of Scanning Probe Microscopy, Moscow Polytechnic University, 107023 Moscow, Russia
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| | - Andreyan Osipov
- Institute of Future Biophysics, 141701 Dolgoprudny, Russia
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, 123098 Moscow, Russia
| | - Sergey Leonov
- Institute of Future Biophysics, 141701 Dolgoprudny, Russia
- Institute of Cell Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia
| |
Collapse
|
4
|
Mierke CT. Softness or Stiffness What Contributes to Cancer and Cancer Metastasis? Cells 2025; 14:584. [PMID: 40277910 PMCID: PMC12026216 DOI: 10.3390/cells14080584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Beyond the genomic and proteomic analysis of bulk and single cancer cells, a new focus of cancer research is emerging that is based on the mechanical analysis of cancer cells. Therefore, several biophysical techniques have been developed and adapted. The characterization of cancer cells, like human cancer cell lines, started with their mechanical characterization at mostly a single timepoint. A universal hypothesis has been proposed that cancer cells need to be softer to migrate and invade tissues and subsequently metastasize in targeted organs. Thus, the softness of cancer cells has been suggested to serve as a universal physical marker for the malignancy of cancer types. However, it has turned out that there exists the opposite phenomenon, namely that stiffer cancer cells are more migratory and invasive and therefore lead to more metastases. These contradictory results question the universality of the role of softness of cancer cells in the malignant progression of cancers. Another problem is that the various biophysical techniques used can affect the mechanical properties of cancer cells, making it even more difficult to compare the results of different studies. Apart from the instrumentation, the culture and measurement conditions of the cancer cells can influence the mechanical measurements. The review highlights the main advances of the mechanical characterization of cancer cells, discusses the strength and weaknesses of the approaches, and questions whether the passive mechanical characterization of cancer cells is still state-of-the art. Besides the cell models, conditions and biophysical setups, the role of the microenvironment on the mechanical characteristics of cancer cells is presented and debated. Finally, combinatorial approaches to determine the malignant potential of tumors, such as the involvement of the ECM, the cells in a homogeneous or heterogeneous association, or biological multi-omics analyses, together with the dynamic-mechanical analysis of cancer cells, are highlighted as new frontiers of research.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Sciences, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
5
|
Libring S, Reinhart-King CA. Premetastatic niche mechanics and organotropism in breast cancer. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:11. [PMID: 40191104 PMCID: PMC11968405 DOI: 10.1038/s44341-025-00015-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025]
Abstract
Numerous physical and mechanical changes occur in the premetastatic niche. Here, we review the mechanics of the premetastatic niche and how the altered extracellular matrix and cancer cell mechanics may play a role in organotropism in breast cancer. Future research into premetastatic niche development and organotropic cell behavior should address physical alterations and biomechanical effects to the same rigor that biochemical alterations are studied.
Collapse
Affiliation(s)
- Sarah Libring
- Department of Bioengineering, Rice University, Houston, TX USA
| | | |
Collapse
|
6
|
Hallfors N, Lamprou C, Luo S, Alkhatib SA, Sapudom J, Aubry C, Alhammadi J, Chan V, Stefanini C, Teo J, Hadjileontiadis L, Pappa AM. Data-driven analysis for the evaluation of cortical mechanics of non-adherent cells. Sci Rep 2025; 15:9700. [PMID: 40113954 PMCID: PMC11926262 DOI: 10.1038/s41598-025-94315-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
Atomic Force Microscopy (AFM) analysis of single cells, especially nonadherent, is inherently slow and analysis-heavy. To address the inherent difficulty of measuring individual cells, and to scale up toward a large number of cells, we take a two-fold approach; first, we introduce an easy-to-fabricate reusable poly(dimethylsiloxane)-based array that consists of micron-sized traps for single-cell trapping, second, we apply a deep-learning method directly on the extracted curves to facilitate and automate the analysis. Our approach is validated using suspended cells, and by applying a small compression with a tipless cantilever AFM probe, we investigate the effect of various cytoskeletal drugs on their deformability. We then apply deep learning models to extract the elasticity of the cell directly from the raw data (with a Coefficient of Determination of 0.47) as well as for binary (with an Area Under the Curve score of 0.91) and multi-class classification (with accuracy scores exceeding 0.9 for each drug). Overall, the versatility to fabricate the microwells in conjunction with the automated analysis and classification streamline the analysis process and demonstrate their ability to generalize to other tasks, such as drug detection.
Collapse
Affiliation(s)
- Nicholas Hallfors
- Department of Biomedical Engineering and Biotechnology, Khalifa University, 127788, Abu Dhabi, UAE
- Healthcare Engineering Innovation Group, Khalifa University, 127788, Abu Dhabi, UAE
- Biotechnology Research Center, Technology Innovation Institute, 9639, Abu Dhabi, UAE
| | - Charalampos Lamprou
- Department of Biomedical Engineering and Biotechnology, Khalifa University, 127788, Abu Dhabi, UAE
- Healthcare Engineering Innovation Group, Khalifa University, 127788, Abu Dhabi, UAE
| | - Shaohong Luo
- Department of Biomedical Engineering and Biotechnology, Khalifa University, 127788, Abu Dhabi, UAE
| | - Sara Awni Alkhatib
- Department of Biomedical Engineering and Biotechnology, Khalifa University, 127788, Abu Dhabi, UAE
- Center for Catalysis and Separations, Khalifa University, 127788, Abu Dhabi, UAE
| | - Jiranuwat Sapudom
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Cyril Aubry
- Research Laboratories, Khalifa University, 127788, Abu Dhabi, UAE
| | - Jawaher Alhammadi
- Department of Biomedical Engineering and Biotechnology, Khalifa University, 127788, Abu Dhabi, UAE
| | - Vincent Chan
- Department of Biomedical Engineering and Biotechnology, Khalifa University, 127788, Abu Dhabi, UAE
- Healthcare Engineering Innovation Group, Khalifa University, 127788, Abu Dhabi, UAE
| | | | - Jeremy Teo
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Leontios Hadjileontiadis
- Department of Biomedical Engineering and Biotechnology, Khalifa University, 127788, Abu Dhabi, UAE.
- Healthcare Engineering Innovation Group, Khalifa University, 127788, Abu Dhabi, UAE.
- Department of Department of Electrical and Computer Engineering, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Anna-Maria Pappa
- Department of Biomedical Engineering and Biotechnology, Khalifa University, 127788, Abu Dhabi, UAE.
- Healthcare Engineering Innovation Group, Khalifa University, 127788, Abu Dhabi, UAE.
- Center for Catalysis and Separations, Khalifa University, 127788, Abu Dhabi, UAE.
| |
Collapse
|
7
|
Abdioğlu HB, Işık Y, Sevgi M, Demircali AA, Gorkem Kirabali U, Esmer GB, Uvet H. Noninvasive holographic sensor system for measuring stiffness of soft micro samples. JOURNAL OF BIOMEDICAL OPTICS 2025; 30:036501. [PMID: 40093760 PMCID: PMC11907929 DOI: 10.1117/1.jbo.30.3.036501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/02/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
Significance: Measuring cell stiffness is essential in cellular biomechanics, particularly in understanding disease progression, including cancer metastasis and tissue mechanics. However, conventional techniques such as atomic force microscopy and optical stretching present limitations, including invasiveness, low throughput, and complex sample preparation. These factors restrict their applicability in dynamic and sensitive biological environments. Aim: This study introduces a noninvasive holographic sensor system for evaluating the stiffness of soft microscale samples. Approach: The proposed system integrates holographic imaging with acoustic stimulation using an off-axis Mach-Zehnder interferometer combined with bulk acoustic waves. This setup allows for label-free, high-throughput measurements while preserving sample integrity. The system was validated with polyacrylamide beads engineered to mimic cellular stiffness, ensuring precise and repeatable stiffness assessments. Results: Measurement errors caused by spatial variations were minimized through a structured imaging approach and a calibration strategy, improving uniformity across different regions. These corrections enhanced the consistency and reliability of stiffness assessments. Experimental validation demonstrated stable stiffness measurements regardless of sample size variations. Repeatability tests further confirmed the system's robustness, producing consistent results across multiple trials. Conclusion: The findings highlight the potential of this holographic sensor system in advancing cell biomechanics research, cancer diagnostics, and mechanobiology. By offering a noninvasive, high-throughput alternative for mechanical property assessments in biological samples, this method contributes to improved characterization of cellular stiffness in biomedical applications.
Collapse
Affiliation(s)
- Hasan Berkay Abdioğlu
- Yıldız Technical University, Department of Mechatronics Engineering, Istanbul, Turkey
| | - Yağmur Işık
- Yıldız Technical University, Department of Mechatronics Engineering, Istanbul, Turkey
| | - Merve Sevgi
- Yıldız Technical University, Department of Bioengineering, Istanbul, Turkey
| | - Ali Anil Demircali
- Imperial College London, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, London, United Kingdom
| | | | - Gokhan Bora Esmer
- Marmara University, Department of Electrical and Electronics Engineering, Faculty of Engineering, Istanbul, Turkey
| | - Huseyin Uvet
- Yıldız Technical University, Department of Mechatronics Engineering, Istanbul, Turkey
| |
Collapse
|
8
|
Paumier A, Verre J, Runel G, Chlasta J, Tribolo S, Chanut S. Anas barbariae 200K Modulates Cell Stiffness and Oxidative Stress in Microglial Cells In Vitro. Int J Mol Sci 2025; 26:1451. [PMID: 40003917 PMCID: PMC11855513 DOI: 10.3390/ijms26041451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Anas barbariae 200K, a homeopathic medicine, is traditionally used for influenza-like illnesses. We investigated the effects of Anas barbariae 200K on microglial cells, a subpopulation of macrophages specific to the central nervous system often used to study the inflammatory processes and oxidative stress generated during influenza-like episodes. The study demonstrates the effect of Anas barbariae 200K on cell stiffness and the reactive oxygen species production using atomic force microscopy and fluorescence microscopy techniques, respectively. Our results showed that Anas barbariae 200K rapidly increased cell stiffness in resting cells by 41% compared with the vehicle. In inflamed cells, cell stiffness was decreased by 21% when treated with Anas barbariae 200K compared with the vehicle. Finally, Anas barbariae 200K caused a reorganisation of filamentous actin, with marked relocation of actin at the cell extremities. Moreover, Anas barbariae 200K significantly decreased the reactive oxygen species (ROS) production in inflamed microglial cells by 40% (total intracellular ROS) and by 67% (mitochondrial ROS) compared with the vehicle. These results strongly suggest an effect of Anas barbariae 200K at a cellular level on cell stiffness and actin cytoskeleton. This sheds light on the biological mechanism of action of this homeopathic preparation.
Collapse
Affiliation(s)
- Anne Paumier
- Laboratoires BOIRON, Research Department, 2 Avenue de l’Ouest Lyonnais, 69510 Messimy, France; (A.P.); (J.V.); (S.C.)
| | - Justine Verre
- Laboratoires BOIRON, Research Department, 2 Avenue de l’Ouest Lyonnais, 69510 Messimy, France; (A.P.); (J.V.); (S.C.)
| | - Gaël Runel
- BioMeca, 60C Avenue Rockfeller, 69008 Lyon, France;
| | | | - Sandra Tribolo
- Laboratoires BOIRON, Research Department, 2 Avenue de l’Ouest Lyonnais, 69510 Messimy, France; (A.P.); (J.V.); (S.C.)
| | - Stéphanie Chanut
- Laboratoires BOIRON, Research Department, 2 Avenue de l’Ouest Lyonnais, 69510 Messimy, France; (A.P.); (J.V.); (S.C.)
| |
Collapse
|
9
|
Cornell CE, Chorlay A, Krishnamurthy D, Martin NR, Baldauf L, Fletcher DA. Target cell tension regulates macrophage trogocytosis. RESEARCH SQUARE 2025:rs.3.rs-5806746. [PMID: 39975908 PMCID: PMC11838726 DOI: 10.21203/rs.3.rs-5806746/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Macrophages are known to engulf small membrane fragments, or trogocytose, target cells and pathogens, rather than fully phagocytose them. However, little is known about what causes macrophages to choose trogocytosis versus phagocytosis. Here, we report that cortical tension of target cells is a key regulator of macrophage trogocytosis. At low tension, macrophages will preferentially trogocytose antibody-opsonized cells, while at high tension they tend towards phagocytosis. Using model vesicles, we demonstrate that macrophages will rapidly switch from trogocytosis to phagocytosis when membrane tension is increased. Stiffening the cortex of target cells also biases macrophages to phagocytose them, a trend that can be countered by increasing antibody surface density and is captured in a mechanical model of trogocytosis. This work suggests that the target cell, rather than the macrophage, determines phagocytosis versus trogocytosis, and that macrophages do not require a distinct molecular pathway for trogocytosis.
Collapse
Affiliation(s)
- Caitlin E. Cornell
- Department of Bioengineering, University of California Berkeley; Berkeley, CA USA
| | - Aymeric Chorlay
- Department of Bioengineering, University of California Berkeley; Berkeley, CA USA
| | - Deepak Krishnamurthy
- Department of Bioengineering, University of California Berkeley; Berkeley, CA USA
| | - Nicholas R. Martin
- Cardiovascular Research Institute, University of California San Francisco; San Francisco, CA USA
| | - Lucia Baldauf
- London Centre for Nanotechnology, University College London; London, UK
| | - Daniel A. Fletcher
- Department of Bioengineering, University of California Berkeley; Berkeley, CA USA
- University of California Berkeley/University of California San Francisco Graduate Group in Bioengineering, CA USA
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory; Berkeley CA USA
- Chan Zuckerberg Biohub; San Francisco CA USA
| |
Collapse
|
10
|
Isert L, Passi M, Freystetter B, Grab M, Roidl A, Müller C, Mehta A, Sundararaghavan HG, Zahler S, Merkel OM. Cellular EMT-status governs contact guidance in an electrospun TACS-mimicking in vitro model. Mater Today Bio 2025; 30:101401. [PMID: 39759848 PMCID: PMC11699613 DOI: 10.1016/j.mtbio.2024.101401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
In this study, an advanced nanofiber breast cancer in vitro model was developed and systematically characterized including physico-chemical, cell-biological and biophysical parameters. Using electrospinning, the architecture of tumor-associated collagen signatures (TACS5 and TACS6) was mimicked. By employing a rotating cylinder or static plate collector set-up, aligned fibers (TACS5-like structures) and randomly orientated fibers (TACS6-like structures) fibers were produced, respectively. The biocompatibility of these fibers was enhanced by collagen coating, ensuring minimal toxicity and improved cell attachment. Various breast cancer cell lines (MCF7, HCC1954, MDA-MB-468, and MDA-MB-231) were cultured on these fibers to assess epithelial-to-mesenchymal transition (EMT) markers, cellular morphology, and migration. Aligned fibers (TACS5) significantly influenced EMT-related changes, promoting cellular alignment, spindle-shaped morphology and a highly migratory phenotype in mesenchymal and hybrid EMT cells (MDA-MB-468, MDA-MB-231). Conversely, epithelial cells (MCF7, HCC1954) showed limited response, but - under growth factor treatment - started to infiltrate the fibrous scaffold and underwent EMT-like changes, particularly on TACS5-mimicks, emphasizing the interplay of topographical cues and EMT induction. The biophysical analysis revealed a clear correlation between cellular EMT status and cell mechanics, with increased EMT correlating to decreased total cellular stiffness. Cancer cell mechanics, however, were found to be dynamic during biochemical and topographical EMT-induction, exceeding initial stiffness by up to 2-fold. These findings highlight the potential of TACS5-like nanofiber scaffolds in modeling the tumor microenvironment and studying cancer cell behavior and mechanics.
Collapse
Affiliation(s)
- Lorenz Isert
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich, Germany
| | - Mehak Passi
- Pharmaceutical Biology, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Benedikt Freystetter
- Department of Cardiac Surgery, Ludwig Maximilians University München, Munich, Germany
| | - Maximilian Grab
- Department of Cardiac Surgery, Ludwig Maximilians University München, Munich, Germany
| | - Andreas Roidl
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christoph Müller
- Center of Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Aditi Mehta
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich, Germany
| | | | - Stefan Zahler
- Pharmaceutical Biology, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Olivia M. Merkel
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich, Germany
| |
Collapse
|
11
|
Wan W, Liu H, Zou J, Xie T, Zhang G, Ying W, Zou X. The optimization and application of photodynamic diagnosis and autofluorescence imaging in tumor diagnosis and guided surgery: current status and future prospects. Front Oncol 2025; 14:1503404. [PMID: 39845324 PMCID: PMC11750647 DOI: 10.3389/fonc.2024.1503404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Photodynamic diagnosis (PDD) and autofluorescence imaging (AFI) are emerging cancer diagnostic technologies that offer significant advantages over traditional white-light endoscopy in detecting precancerous lesions and early-stage cancers; moreover, they hold promising potential in fluorescence-guided surgery (FGS) for tumors. However, their shortcomings have somewhat hindered the clinical application of PDD and AFI. Therefore, it is imperative to enhance the efficacy of PDD and AFI, thereby maximizing their potential for practical clinical use. This article reviews the principles, characteristics, current research status, and advancements of PDD and AFI, focusing on analyzing and discussing the optimization strategies of PDD and AFI in tumor diagnosis and FGS scenarios. Considering the practical and technical feasibility, optimizing PDD and AFI may result in an effective real-time diagnostic tool to guide clinicians in tumor diagnosis and surgical guidance to achieve the best results.
Collapse
Affiliation(s)
- Wei Wan
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Huiquan Liu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junrong Zou
- Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tianpeng Xie
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Guoxi Zhang
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Weihai Ying
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaofeng Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
12
|
Lan H, Tan XHM, Le MTT, Chien HY, Zheng R, Rowat AC, Teitell MA, Chiou PY. Optomagnetic Micromirror Arrays for Mapping Large Area Stiffness Distributions of Biomimetic Materials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406389. [PMID: 39614709 PMCID: PMC11710979 DOI: 10.1002/smll.202406389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/17/2024] [Indexed: 12/01/2024]
Abstract
A new device termed "Optomagnetic Micromirror Arrays" (OMA) is demonstrated capable of mapping the stiffness distribution of biomimetic materials across a 5.1 mm × 7.2 mm field of view with cellular resolution. The OMA device comprises an array of 50 000 magnetic micromirrors with optical grating structures embedded beneath an elastic PDMS film, with biomimetic materials affixed on top. Illumination of a broadband white light beam onto these micromirrors results in the reflection of microscale rainbow light rays on each micromirror. When a magnetic field is applied, it causes each micromirror to tilt differently depending on the local stiffness of the biomimetic materials. Through imaging these micromirrors with low N.A. optics, a specific narrow band of reflection light rays from each micromirror is captured. Changing a micromirror's tilt angle also alters the color spectrum it reflects back to the imaging system and the color of the micromirror image it represents. As a result, OMA can infer the local stiffness of the biomimetic materials through the color change detected on each micromirror. OMA offers the potential for high-throughput stiffness mapping at the tissue-level while maintaining spatial resolution at the cellular level.
Collapse
Affiliation(s)
- Hsin Lan
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Xing Haw Marvin Tan
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), Singapore, 138632, Republic of Singapore
| | - Minh-Tam Tran Le
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Hao-Yu Chien
- Department of Electrical and Computer Enigeering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ruoda Zheng
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Amy C Rowat
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Integrative Biology & Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Michael A Teitell
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Pei-Yu Chiou
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
13
|
Owusu SB, Russell E, Ekanayake AB, Tivanski AV, Petronek MS. Iron promotes isocitrate dehydrogenase mutant glioma cell motility. Free Radic Biol Med 2025; 226:109-116. [PMID: 39551451 DOI: 10.1016/j.freeradbiomed.2024.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/19/2024]
Abstract
Enriched iron metabolic features such as high transferrin receptor (TfR) expression and high iron content are commonly observed in aggressive gliomas and can be associated with poor clinical responses. However, the underlying question of how iron contributes to tumor aggression remains elusive. Gliomas harboring isocitrate dehydrogenase (IDH) mutations account for a high percentage (>70 %) of recurrent tumors and cells with an acquired IDH mutation have been reported to have increased motility and invasion. This study aims to investigate how an acquired IDH mutation modulates iron metabolism and the implication(s) of iron on tumor cell growth. IDH mutant cells (U87R132H) grow significantly faster which is accompanied with increased TfR expression and iron uptake in vitro compared to wild-type U87 cells. This phenotype is retained in vivo. Biomechanically, U87R132H cells are significantly less stiff and supplementation with ferrous ammonium sulfate (Fe2+) augments membrane fluidity to drive U87R132H cells into a super motile state. These findings provide insight into how an acquired IDH mutation may be able to modulate iron metabolism, allowing iron to serve as a biomechanical driver of tumor progression.
Collapse
Affiliation(s)
- Stephenson Boakye Owusu
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, University of Iowa, Iowa City, IA, USA
| | - Emily Russell
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, University of Iowa, Iowa City, IA, USA
| | | | | | - Michael S Petronek
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
14
|
Yin Y, Liu Z. Microfluidic Technology for Measuring Mechanical Properties of Single Cells and Its Application. Bioengineering (Basel) 2024; 11:1266. [PMID: 39768083 PMCID: PMC11673902 DOI: 10.3390/bioengineering11121266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Cellular mechanical properties are critical for tissue and organ homeostasis, which are associated with many diseases and are very promising non-labeled biomarkers. Over the past two decades, many research tools based on microfluidic methods have been developed to measure the biophysical properties of single cells; however, it has still not been possible to develop a technique that allows for high-throughput, easy-to-operate and precise measurements of single-cell biophysical properties. In this paper, we review the emerging technologies implemented based on microfluidic approaches for characterizing the mechanical properties of single cells and discuss the methodological principles, advantages, limitations, and applications of various technologies.
Collapse
Affiliation(s)
| | - Ziyuan Liu
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Department of Ophthalmology, Peking University Third Hospital, Peking University Institute of Laser Medicine, No. 49 North Garden Road, Haidian District, Beijing 100191, China;
| |
Collapse
|
15
|
Asghari M, Ivetich SD, Aslan MK, Aramesh M, Melkonyan O, Meng Y, Xu R, Colombo M, Weiss T, Balabanov S, Stavrakis S, deMello AJ. Real-time viscoelastic deformability cytometry: High-throughput mechanical phenotyping of liquid and solid biopsies. SCIENCE ADVANCES 2024; 10:eabj1133. [PMID: 39630916 PMCID: PMC11616701 DOI: 10.1126/sciadv.abj1133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024]
Abstract
In principle, the measurement of mechanical property differences between cancer cells and their benign counterparts enables the detection, diagnosis, and classification of diseases. Despite the existence of various mechanophenotyping methods, the ability to perform high-throughput single-cell deformability measurements on liquid and/or solid tissue biopsies remains an unmet challenge within clinical settings. To address this issue, we present an ultrahigh-throughput viscoelastic microfluidic platform able to measure the mechanical properties of single cells at rates of up to 100,000 cells per second (and up to 10,000 cells per second in real time). To showcase the utility of the presented platform in clinical scenarios, we perform single-cell phenotyping of both liquid and solid tumor biopsies, cytoskeletal drug analysis, and identification of malignant lymphocytes in peripheral blood samples. Our viscoelastic microfluidic methodology offers opportunities for high-throughput, label-free single-cell analysis, with diverse applications in clinical diagnostics and personalized medicine.
Collapse
Affiliation(s)
- Mohammad Asghari
- Institute for Chemical and Bioengineering, ETH Zürich, 8093 Zürich, Switzerland
| | | | - Mahmut Kamil Aslan
- Institute for Chemical and Bioengineering, ETH Zürich, 8093 Zürich, Switzerland
| | - Morteza Aramesh
- Department of Materials Science and Engineering, Uppsala University, Uppsala, Sweden
- Department of Information Technology and Electrical Engineering, ETH Zürich, 8092 Zürich, Switzerland
| | - Oleksandr Melkonyan
- Institute for Chemical and Bioengineering, ETH Zürich, 8093 Zürich, Switzerland
| | - Yingchao Meng
- Institute for Chemical and Bioengineering, ETH Zürich, 8093 Zürich, Switzerland
| | - Rong Xu
- Department of Neurology, University Hospital Zürich, 8091 Zürich, Switzerland
- Clinical Neuroscience Center, University of Zürich, 8091 Zürich, Switzerland
| | - Monika Colombo
- Institute for Chemical and Bioengineering, ETH Zürich, 8093 Zürich, Switzerland
- Department of Mechanical and Production Engineering, Aarhus University, Aarhus, Denmark
| | - Tobias Weiss
- Department of Neurology, University Hospital Zürich, 8091 Zürich, Switzerland
- Clinical Neuroscience Center, University of Zürich, 8091 Zürich, Switzerland
| | - Stefan Balabanov
- Clinic for Medical Oncology and Hematology, University Hospital Zürich, 8091 Zürich, Switzerland
- University Center for Laboratory Medicine and Pathology, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Stavros Stavrakis
- Institute for Chemical and Bioengineering, ETH Zürich, 8093 Zürich, Switzerland
| | - Andew J. deMello
- Institute for Chemical and Bioengineering, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
16
|
Cornell CE, Chorlay A, Krishnamurthy D, Martin NR, Baldauf L, Fletcher DA. Target cell tension regulates macrophage trogocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626490. [PMID: 39677802 PMCID: PMC11642796 DOI: 10.1101/2024.12.02.626490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Macrophages are known to engulf small membrane fragments, or trogocytose, target cells and pathogens, rather than fully phagocytose them. However, little is known about what causes macrophages to choose trogocytosis versus phagocytosis. Here, we report that cortical tension of target cells is a key regulator of macrophage trogocytosis. At low tension, macrophages will preferentially trogocytose antibody-opsonized cells, while at high tension they tend towards phagocytosis. Using model vesicles, we demonstrate that macrophages will rapidly switch from trogocytosis to phagocytosis when membrane tension is increased. Stiffening the cortex of target cells also biases macrophages to phagocytose them, a trend that can be countered by increasing antibody surface density and is captured in a mechanical model of trogocytosis. This work suggests that a distinct molecular pathway for trogocytosis is not required to explain differences in trogocytosis among target cell types and points to a mechanism for target cells to modulate trogocytosis.
Collapse
|
17
|
Qiu Y, Gao T, Smith BR. Mechanical deformation and death of circulating tumor cells in the bloodstream. Cancer Metastasis Rev 2024; 43:1489-1510. [PMID: 38980581 PMCID: PMC11900898 DOI: 10.1007/s10555-024-10198-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024]
Abstract
The circulation of tumor cells through the bloodstream is a significant step in tumor metastasis. To better understand the metastatic process, circulating tumor cell (CTC) survival in the circulation must be explored. While immune interactions with CTCs in recent decades have been examined, research has yet to sufficiently explain some CTC behaviors in blood flow. Studies related to CTC mechanical responses in the bloodstream have recently been conducted to further study conditions under which CTCs might die. While experimental methods can assess the mechanical properties and death of CTCs, increasingly sophisticated computational models are being built to simulate the blood flow and CTC mechanical deformation under fluid shear stresses (FSS) in the bloodstream.Several factors contribute to the mechanical deformation and death of CTCs as they circulate. While FSS can damage CTC structure, diverse interactions between CTCs and blood components may either promote or hinder the next metastatic step-extravasation at a remote site. Overall understanding of how these factors influence the deformation and death of CTCs could serve as a basis for future experiments and simulations, enabling researchers to predict CTC death more accurately. Ultimately, these efforts can lead to improved metastasis-specific therapeutics and diagnostics specific in the future.
Collapse
Affiliation(s)
- Yunxiu Qiu
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, 48824, USA
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Tong Gao
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Department of Computational Mathematics, Science, and Engineering, East Lansing, MI, 48824, USA
| | - Bryan Ronain Smith
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, 48824, USA.
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
18
|
O’Dowling AT, Rodriguez BJ, Gallagher TK, Thorpe SD. Machine learning and artificial intelligence: Enabling the clinical translation of atomic force microscopy-based biomarkers for cancer diagnosis. Comput Struct Biotechnol J 2024; 24:661-671. [PMID: 39525667 PMCID: PMC11543504 DOI: 10.1016/j.csbj.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
The influence of biomechanics on cell function has become increasingly defined over recent years. Biomechanical changes are known to affect oncogenesis; however, these effects are not yet fully understood. Atomic force microscopy (AFM) is the gold standard method for measuring tissue mechanics on the micro- or nano-scale. Due to its complexity, however, AFM has yet to become integrated in routine clinical diagnosis. Artificial intelligence (AI) and machine learning (ML) have the potential to make AFM more accessible, principally through automation of analysis. In this review, AFM and its use for the assessment of cell and tissue mechanics in cancer is described. Research relating to the application of artificial intelligence and machine learning in the analysis of AFM topography and force spectroscopy of cancer tissue and cells are reviewed. The application of machine learning and artificial intelligence to AFM has the potential to enable the widespread use of nanoscale morphologic and biomechanical features as diagnostic and prognostic biomarkers in cancer treatment.
Collapse
Affiliation(s)
- Aidan T. O’Dowling
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- Department of Hepatobiliary and Transplant Surgery, St Vincent’s University Hospital, Dublin, Ireland
| | - Brian J. Rodriguez
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- UCD School of Physics, University College Dublin, Dublin, Ireland
| | - Tom K. Gallagher
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- Department of Hepatobiliary and Transplant Surgery, St Vincent’s University Hospital, Dublin, Ireland
| | - Stephen D. Thorpe
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
19
|
Sanoria M, Chelakkot R, Nandi A. Percolation transitions in a binary mixture of active Brownian particles with different softness. SOFT MATTER 2024; 20:9184-9192. [PMID: 39530663 DOI: 10.1039/d4sm00981a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Homogeneous active Brownian particle (ABP) systems with purely repulsive interactions are considered to exhibit a simple phase behavior, but various physical attributes of active entities can lead to variation in the collective dynamics. Recent studies have shown that even homogeneous ABPs exhibit complex behavior due to an interplay between particle softness and motility. However, the heterogeneity in the composition of ABPs has not been explored yet. In this paper, we study the structural properties of a binary mixture of ABPs with different particle softness by varying the relative softness and composition. We found that upon varying the motility parameter, the system underwent a motility-induced phase separation (MIPS) followed by a percolation transition similar to the homogeneous systems. However, we observed a novel feature: the formation of a space-filling structure made of particles with higher stiffness, within the dense cluster of MIPS containing both types of particles. Our systematic analysis shows that this structure formation occurs only if the difference in softness of both types of particles is sufficiently large. Furthermore, the presence of a non-linear scaling for different compositions of binary ABPs suggests that there is a complex relationship between the composition and the structural properties. Our study demonstrates that the composition heterogeneity of ABPs can lead to complex phase behavior.
Collapse
Affiliation(s)
- Monika Sanoria
- Department of Physics, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
- Center for Cellular and Biomolecular Machines, University of California Merced, CA, 95343, USA.
| | - Raghunath Chelakkot
- Department of Physics, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - Amitabha Nandi
- Department of Physics, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| |
Collapse
|
20
|
Lai A, Hinz S, Dong A, Lustig M, LaBarge MA, Sohn LL. Multi-Zone Visco-Node-Pore Sensing: A Microfluidic Platform for Multi-Frequency Viscoelastic Phenotyping of Single Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406013. [PMID: 39308179 DOI: 10.1002/advs.202406013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/19/2024] [Indexed: 11/22/2024]
Abstract
This study introduces multi-zone visco-Node-Pore Sensing (mz-visco-NPS), an electronic-based microfluidic platform for single-cell viscoelastic phenotyping. mz-visco-NPS implements a series of sinusoidal-shaped contraction zones that periodically deform a cell at specific strain frequencies, leading to changes in resistance across the zones that correspond to the cell's frequency-dependent elastic G' and viscous G″ moduli. mz-visco-NPS is validated by measuring the viscoelastic changes of MCF-7 cells when their cytoskeleton is disrupted. mz-visco-NPS is also employed to measure the viscoelastic properties of human mammary epithelial cells across the entire continuum of epithelial transformation states, from average- and high-risk primary epithelial cells, to immortal non-malignant (MCF-10A), malignant (MCF-7), and metastatic (MDA-MB-231) cell lines. With a throughput of 600 cells per hour and demonstrated ease-of-use, mz-visco-NPS reveals a remarkable level of single-cell heterogeneity that would otherwise be masked by ensemble averaging.
Collapse
Affiliation(s)
- Andre Lai
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Stefan Hinz
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, 91010, USA
| | - Alan Dong
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, 94720, USA
| | - Michael Lustig
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, 94720, USA
| | - Mark A LaBarge
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, 91010, USA
| | - Lydia L Sohn
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, University of California, Berkeley, CA, 94720, USA
- Department of Mechanical Engineering, University of California, Berkeley, 94720, USA
| |
Collapse
|
21
|
Sneh T, Corsetti S, Notaros M, Kikkeri K, Voldman J, Notaros J. Optical tweezing of microparticles and cells using silicon-photonics-based optical phased arrays. Nat Commun 2024; 15:8493. [PMID: 39362852 PMCID: PMC11450221 DOI: 10.1038/s41467-024-52273-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/29/2024] [Indexed: 10/05/2024] Open
Abstract
Integrated optical tweezers have the potential to enable highly-compact, low-cost, mass-manufactured, and broadly-accessible optical manipulation when compared to standard bulk-optical tweezers. However, integrated demonstrations to date have been fundamentally limited to micron-scale standoff distances and, often, passive trapping functionality, making them incompatible with many existing applications and significantly limiting their utility, especially for biological studies. In this work, we demonstrate optical trapping and tweezing using an integrated OPA for the first time, increasing the standoff distance of integrated optical tweezers by over two orders of magnitude compared to prior demonstrations. First, we demonstrate trapping of polystyrene microspheres 5 mm above the surface of the chip and calibrate the trap force. Next, we show tweezing of polystyrene microspheres in one dimension by non-mechanically steering the trap by varying the input laser wavelength. Finally, we use the OPA tweezers to demonstrate, to the best of our knowledge, the first cell experiments using single-beam integrated optical tweezers, showing controlled deformation of mouse lymphoblast cells. This work introduces a new modality for integrated optical tweezers, significantly expanding their utility and compatibility with existing applications, especially for biological experiments.
Collapse
Affiliation(s)
- Tal Sneh
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sabrina Corsetti
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Milica Notaros
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kruthika Kikkeri
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Joel Voldman
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jelena Notaros
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
22
|
Vessella T, Rozen EJ, Shohet J, Wen Q, Zhou HS. Investigation of Cell Mechanics and Migration on DDR2-Expressing Neuroblastoma Cell Line. Life (Basel) 2024; 14:1260. [PMID: 39459560 PMCID: PMC11509142 DOI: 10.3390/life14101260] [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: 09/13/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Neuroblastoma is a devastating disease accounting for ~15% of all childhood cancer deaths. Collagen content and fiber association within the tumor stroma influence tumor progression and metastasis. High expression levels of collagen receptor kinase, Discoidin domain receptor II (DDR2), are associated with the poor survival of neuroblastoma patients. Additionally, cancer cells generate and sustain mechanical forces within their environment as a part of their normal physiology. Despite this, evidence regarding whether collagen-activated DDR2 signaling dysregulates these migration forces is still elusive. To address these questions, a novel shRNA DDR2 knockdown neuroblastoma cell line (SH-SY5Y) was engineered to evaluate the consequence of DDR2 on cellular mechanics. Atomic force microscopy (AFM) and traction force microscopy (TFM) were utilized to unveil the biophysical altercations. DDR2 downregulation was found to significantly reduce proliferation, cell stiffness, and cellular elongation. Additionally, DDR2-downregulated cells had decreased traction forces when plated on collagen-coated elastic substrates. Together, these results highlight the important role that DDR2 has in reducing migration mechanics in neuroblastoma and suggest DDR2 may be a promising novel target for future therapies.
Collapse
Affiliation(s)
- Theadora Vessella
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Rd., Worcester, MA 01609, USA;
| | - Esteban J. Rozen
- Crnic Institute Bolder Branch, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Avenue, Boulder, CO 80303, USA
- Department of Pediatrics, University of Massachusetts Medical School, 55 Lake Ave North, Worcester, MA 01655, USA
| | - Jason Shohet
- Department of Pediatrics, University of Massachusetts Medical School, 55 Lake Ave North, Worcester, MA 01655, USA
| | - Qi Wen
- Department of Physics, Worcester Polytechnic Institute, 100 Institute Rd., Worcester, MA 01609, USA
| | - Hong Susan Zhou
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Rd., Worcester, MA 01609, USA;
| |
Collapse
|
23
|
Rosvold JR, Zanini G, Handler C, Frank E, Li J, Vitolo MI, Martin SS, Scarcelli G. Stimulated Brillouin scattering flow cytometry. BIOMEDICAL OPTICS EXPRESS 2024; 15:6024-6035. [PMID: 39421786 PMCID: PMC11482170 DOI: 10.1364/boe.537602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 10/19/2024]
Abstract
We present the use of stimulated Brillouin scattering spectroscopy to achieve rapid measurements of cell biomechanics in a flow cytometer setup. Specifically, our stimulated Brillouin scattering flow cytometry can acquire at a rate of 200 Hz, with a spectral acquisition time of 5 ms, which marks a 10x improvement compared to previous demonstrations of spontaneous Brillouin scattering flow cytometry. We experimentally validate our stimulated Brillouin scattering flow cytometer by measuring cell populations of normal breast epithelial cells and metastatic breast epithelial cancer cells.
Collapse
Affiliation(s)
- Jake R. Rosvold
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Giulia Zanini
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Chenchen Handler
- Department of Mechanical Engineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, USA
| | - Eric Frank
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Jiarui Li
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Michele I. Vitolo
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Stuart S. Martin
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Giuliano Scarcelli
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, College Park, MD 20742, USA
| |
Collapse
|
24
|
Ghézali G, Ribot J, Curry N, Pillet LE, Boutet-Porretta F, Mozheiko D, Calvo CF, Ezan P, Perfettini I, Lecoin L, Janel S, Zapata J, Escartin C, Etienne-Manneville S, Kaminski CF, Rouach N. Connexin 30 locally controls actin cytoskeleton and mechanical remodeling in motile astrocytes. Glia 2024; 72:1915-1929. [PMID: 38982826 DOI: 10.1002/glia.24590] [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: 06/07/2022] [Revised: 06/04/2024] [Accepted: 06/25/2024] [Indexed: 07/11/2024]
Abstract
During brain maturation, astrocytes establish complex morphologies unveiling intense structural plasticity. Connexin 30 (Cx30), a gap-junction channel-forming protein expressed postnatally, dynamically regulates during development astrocyte morphological properties by controlling ramification and extension of fine processes. However, the underlying mechanisms remain unexplored. Here, we found in vitro that Cx30 interacts with the actin cytoskeleton in astrocytes and inhibits its structural reorganization and dynamics during cell migration. This translates into an alteration of local physical surface properties, as assessed by correlative imaging using stimulated emission depletion (STED) super resolution imaging and atomic force microscopy (AFM). Specifically, Cx30 impaired astrocyte cell surface topology and cortical stiffness in motile astrocytes. As Cx30 alters actin organization, dynamics, and membrane physical properties, we assessed whether it controls astrocyte migration. We found that Cx30 reduced persistence and directionality of migrating astrocytes. Altogether, these data reveal Cx30 as a brake for astrocyte structural and mechanical plasticity.
Collapse
Affiliation(s)
- Grégory Ghézali
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Doctoral School N° 158, Sorbonne Université, Paris, France
| | - Jérôme Ribot
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Nathan Curry
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Laure-Elise Pillet
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Doctoral School N°562, Université Paris Cité, Paris, France
| | - Flora Boutet-Porretta
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Doctoral School N° 158, Sorbonne Université, Paris, France
| | - Daria Mozheiko
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Doctoral School N° 158, Sorbonne Université, Paris, France
| | - Charles-Félix Calvo
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Pascal Ezan
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Isabelle Perfettini
- Institut Pasteur, Université de Paris, CNRS, Cell Polarity, Migration and Cancer Unit, Paris, France
| | - Laure Lecoin
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Sébastien Janel
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Jonathan Zapata
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | | | - Clemens F Kaminski
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| |
Collapse
|
25
|
Dutta D, Ziemke M, Sindelar P, Vargas H, Lim JY, Chandra S. Cytoarchitecture of Breast Cancer Cells under Diabetic Conditions: Role of Regulatory Kinases-Rho Kinase and Focal Adhesion Kinase. Cancers (Basel) 2024; 16:3166. [PMID: 39335137 PMCID: PMC11430325 DOI: 10.3390/cancers16183166] [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: 08/08/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Diabetes greatly reduces the survival rates in breast cancer patients due to chemoresistance and metastasis. Reorganization of the cytoskeleton is crucial to cell migration and metastasis. Regulatory cytoskeletal protein kinases such as the Rho kinase (ROCK) and focal adhesion kinase (FAK) play a key role in cell mobility and have been shown to be affected in cancer. It is hypothesized that diabetes/high-glucose conditions alter the cytoskeletal structure and, thus, the elasticity of breast cancer cells through the ROCK and FAK pathway, which can cause rapid metastasis of cancer. The aim of the study was to investigate the role of potential mediators that affect the morphology of cancer cells in diabetes, thus leading to aggressive cancer. Breast cancer cells (MDA-MB-231 and MCF-7) were treated with 5 mM glucose (low glucose) or 25 mM glucose (high glucose) in the presence of Rho kinase inhibitor (Y-27632, 10 mM) or FAK inhibitor (10 mM). Cell morphology and elasticity were monitored using atomic force microscopy (AFM), and actin staining was performed by fluorescence microscopy. For comparative study, normal mammary breast epithelial cells (MCF-10A) were used. It was observed that high-glucose treatments modified the cytoskeleton of the cells, as observed through AFM and fluorescence microscopy, and significantly reduced the elasticity of the cells. Blocking the ROCK or FAK pathway diminished the high-glucose effects. These changes were more evident in the breast cancer cells as compared to the normal cells. This study improves the knowledge on the cytoarchitecture properties of diabetic breast cancer cells and provides potential pathways that can be targeted to prevent such effects.
Collapse
Affiliation(s)
- Diganta Dutta
- Department of Physics, Astronomy and Engineering, University of Nebraska at Kearney, Kearney, NE 68849, USA
| | - Matthew Ziemke
- Department of Physics, Astronomy and Engineering, University of Nebraska at Kearney, Kearney, NE 68849, USA
| | - Payton Sindelar
- Department of Biology, University of Nebraska at Kearney, Kearney, NE 68849, USA (H.V.)
| | - Hernan Vargas
- Department of Biology, University of Nebraska at Kearney, Kearney, NE 68849, USA (H.V.)
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Surabhi Chandra
- Department of Biology, University of Nebraska at Kearney, Kearney, NE 68849, USA (H.V.)
| |
Collapse
|
26
|
Zheng Y, Li J, Xu D, Liu L, Li Y, Yi J, Dong J, Pang D, Tang H. Tunneling nanotubes mediate KRas transport: Inducing tumor heterogeneity and altering cellular membrane mechanical properties. Acta Biomater 2024; 185:312-322. [PMID: 38969079 DOI: 10.1016/j.actbio.2024.06.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024]
Abstract
Mutation in oncogene KRas plays a crucial role in the occurrence and progression of numerous malignant tumors. Malignancy involves changes in cell mechanics for extensive cellular deformation during metastatic dissemination. We hypothesize that oncogene KRas mutations are intrinsic to alterations in cellular mechanics that promote malignant tumor generation and progression. Here, we demonstrate the use of optical tweezers coupled with a confocal fluorescence imaging system and gene interference technique to reveal that the mutant KRas protein can be transported between homogeneous and heterogeneous tumor cells by tunneling nanotubes (TNTs), resulting in a significant reduction of membrane tension and acceleration of membrane phospholipid flow in the recipient cells. Simultaneously, the changes in membrane mechanical properties of the tumor cells also enhance the metastatic and invasive ability of the tumors, which further contribute to the deterioration of the tumors. This finding helps to clarify the association between oncogene mutations and changes in the mechanical properties of tumor cells, which provides a theoretical basis for the development of cancer treatment strategies. STATEMENT OF SIGNIFICANCE: Here, we present a laser confocal fluorescence system integrated with optical tweezers to observe the transfer of mutant KRasG12D protein from mutant cells to wild-type cells through TNTs. Malignancy involves changes in cell mechanics for extensive cellular deformation during metastatic dissemination. Our results demonstrate a significant decrease in membrane tension and an increase in membrane phospholipid flow in recipient cells. These alterations in mechanical properties augment the migration and invasive capabilities of tumor cells, contributing to tumor malignancy. Our findings propose that cellular mechanical properties could serve as new markers for tumor development, and targeting membrane tension may hold potential as a therapeutic strategy.
Collapse
Affiliation(s)
- Yawen Zheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Jiangtao Li
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Dadi Xu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Liu Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Yuyao Li
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Jing Yi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Jiayao Dong
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Daiwen Pang
- College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Hongwu Tang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China.
| |
Collapse
|
27
|
Islam MM, Gaska I, Oshinowo O, Otumala A, Shekhar S, Au Yong N, Myers DR. Single-pericyte nanomechanics measured by contraction cytometry. APL Bioeng 2024; 8:036109. [PMID: 39131206 PMCID: PMC11316606 DOI: 10.1063/5.0213761] [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: 04/12/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
Pericytes line the microvasculature throughout the body and play a key role in regulating blood flow by constricting and dilating vessels. However, the biophysical mechanisms through which pericytes transduce microenvironmental chemical and mechanical cues to mediate vessel diameter, thereby impacting oxygen and nutrient delivery, remain largely unknown. This knowledge gap is clinically relevant as numerous diseases are associated with the aberrant contraction of pericytes, which are unusually susceptible to injury. Here, we report the development of a high-throughput hydrogel-based pericyte contraction cytometer that quantifies single-cell contraction forces from murine and human pericytes in different microvascular microenvironments and in the presence of competing vasoconstricting and vasodilating stimuli. We further show that murine pericyte survival in hypoxia is mediated by the mechanical microenvironment and that, paradoxically, pre-treating pericytes to reduce contraction increases hypoxic cell death. Moreover, using the contraction cytometer as a drug-screening tool, we found that cofilin-1 could be applied extracellularly to release murine pericytes from hypoxia-induced contractile rigor mortis and, therefore, may represent a novel approach for mitigating the long-lasting decrease in blood flow that occurs after hypoxic injury.
Collapse
Affiliation(s)
| | - Ignas Gaska
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, Georgia 30322, USA
| | | | | | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, Georgia 30322, USA
| | | | - David R. Myers
- Author to whom correspondence should be addressed:. Tel.: 404-727-0401
| |
Collapse
|
28
|
Hansen E, Rolling C, Wang M, Holaska JM. Emerin deficiency drives MCF7 cells to an invasive phenotype. Sci Rep 2024; 14:19998. [PMID: 39198511 PMCID: PMC11358522 DOI: 10.1038/s41598-024-70752-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
During metastasis, cancer cells traverse the vasculature by squeezing through very small gaps in the endothelium. Thus, nuclei in metastatic cancer cells must become more malleable to move through these gaps. Our lab showed invasive breast cancer cells have 50% less emerin protein resulting in smaller, misshapen nuclei, and higher metastasis rates than non-cancerous controls. Thus, emerin deficiency was predicted to cause increased nuclear compliance, cell migration, and metastasis. We tested this hypothesis by downregulating emerin in noninvasive MCF7 cells and found emerin knockdown causes smaller, dysmorphic nuclei, resulting in increased impeded cell migration. Emerin reduction in invasive breast cancer cells showed similar results. Supporting the clinical relevance of emerin reduction in cancer progression, our analysis of 192 breast cancer patient samples showed emerin expression inversely correlates with cancer invasiveness. We conclude emerin loss is an important driver of invasive transformation and has utility as a biomarker for tumor progression.
Collapse
Affiliation(s)
- Emily Hansen
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, MEB 534, 401 South Broadway, Camden, NJ, 08103, USA
- Molecular and Cell Biology and Neuroscience Program, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Stratford, NJ, 08084, USA
| | - Christal Rolling
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, MEB 534, 401 South Broadway, Camden, NJ, 08103, USA
- Molecular and Cell Biology and Neuroscience Program, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Stratford, NJ, 08084, USA
| | - Matthew Wang
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, MEB 534, 401 South Broadway, Camden, NJ, 08103, USA
- Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, 08084, USA
| | - James M Holaska
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, MEB 534, 401 South Broadway, Camden, NJ, 08103, USA.
- Molecular and Cell Biology and Neuroscience Program, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Stratford, NJ, 08084, USA.
| |
Collapse
|
29
|
Hansen E, Rolling C, Wang M, Holaska JM. Emerin deficiency drives MCF7 cells to an invasive phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.21.581379. [PMID: 38712242 PMCID: PMC11071294 DOI: 10.1101/2024.02.21.581379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
During metastasis, cancer cells traverse the vasculature by squeezing through very small gaps in the endothelium. Thus, nuclei in metastatic cancer cells must become more malleable to move through these gaps. Our lab showed invasive breast cancer cells have 50% less emerin protein resulting in smaller, misshapen nuclei, and higher metastasis rates than non-cancerous controls. Thus, emerin deficiency was predicted to cause increased nuclear compliance, cell migration, and metastasis. We tested this hypothesis by downregulating emerin in noninvasive MCF7 cells and found emerin knockdown causes smaller, dysmorphic nuclei, resulting in increased impeded cell migration. Emerin reduction in invasive breast cancer cells showed similar results. Supporting the clinical relevance of emerin reduction in cancer progression, our analysis of 192 breast cancer patient samples showed emerin expression inversely correlates with cancer invasiveness. We conclude emerin loss is an important driver of invasive transformation and has utility as a biomarker for tumor progression.
Collapse
Affiliation(s)
- Emily Hansen
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ
- Molecular and Cell Biology and Neuroscience Program, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Stratford, NJ
| | - Christal Rolling
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ
- Molecular and Cell Biology and Neuroscience Program, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Stratford, NJ
| | - Matthew Wang
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ
- Rowan-Virtua School of Osteopathic Medicine
| | - James M. Holaska
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ
- Molecular and Cell Biology and Neuroscience Program, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Stratford, NJ
| |
Collapse
|
30
|
Gasser E, Su E, Vaidžiulytė K, Abbade N, Cognart H, Manneville JB, Viovy JL, Piel M, Pierga JY, Terao K, Villard C. Deformation under flow and morphological recovery of cancer cells. LAB ON A CHIP 2024; 24:3930-3944. [PMID: 38993177 PMCID: PMC11302772 DOI: 10.1039/d4lc00246f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024]
Abstract
The metastatic cascade includes a blood circulation step for cells detached from the primary tumor. This stage involves significant shear stress as well as large and fast deformation as the cells circulate through the microvasculature. These mechanical stimuli are well reproduced in microfluidic devices. However, the recovery dynamics after deformation is also pivotal to understand how a cell can pass through the multiple capillary constrictions encountered during a single hemodynamic cycle. The microfluidic system developed in this work allows single cell recovery to be studied under flow-free conditions following pressure-actuated cell deformation inside constricted microchannels. We used three breast cancer cell lines - namely MCF-7, SK-BR3 and MDA-MB231 - as cellular models representative of different cancer phenotypes. Changing the size of the constriction allows exploration of moderate to strong deformation regimes, the latter being associated with the formation of plasma membrane blebs. In the regime of moderate deformation, all cell types display a fast elastic recovery behavior followed by a slower viscoelastic regime, well described by a double exponential decay. Among the three cell types, cells of the mesenchymal phenotype, i.e. the MDA-MB231 cells, are softer and the most fluid-like, in agreement with previous studies. Our main finding here is that the fast elastic recovery regime revealed by our novel microfluidic system is under the control of cell contractility ensured by the integrity of the cell cortex. Our results suggest that the cell cortex plays a major role in the transit of circulating tumor cells by allowing their fast morphological recovery after deformation in blood capillaries.
Collapse
Affiliation(s)
- Emile Gasser
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
- Laboratoire Interdisciplinaire des Energies de Demain, CNRS UMR 8236, Université Paris Cité, F-75013, Paris, France.
| | - Emilie Su
- Laboratoire Interdisciplinaire des Energies de Demain, CNRS UMR 8236, Université Paris Cité, F-75013, Paris, France.
- Laboratoire Matière et Systèmes Complexes (MSC), CNRS UMR 7057, Université Paris Cité, 10 Rue Alice Domon et Léonie Duquet, F-75013 Paris, France
| | - Kotryna Vaidžiulytė
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS UMR144, Université PSL, F-75005 Paris, France
| | - Nassiba Abbade
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS UMR144, Université PSL, F-75005 Paris, France
| | - Hamizah Cognart
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
| | - Jean-Baptiste Manneville
- Laboratoire Matière et Systèmes Complexes (MSC), CNRS UMR 7057, Université Paris Cité, 10 Rue Alice Domon et Léonie Duquet, F-75013 Paris, France
| | - Jean-Louis Viovy
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS UMR144, Université PSL, F-75005 Paris, France
| | - Jean-Yves Pierga
- Département d'Oncologie Médicale de l'Institut Curie et Université Paris Cité, France
| | - Kyohei Terao
- Nano-Micro Structure Device Integrated Research Center, Kagawa University, 2217-20 Hayashi-cho, Takamatsu 761-0396, Japan.
| | - Catherine Villard
- Laboratoire Interdisciplinaire des Energies de Demain, CNRS UMR 8236, Université Paris Cité, F-75013, Paris, France.
| |
Collapse
|
31
|
Iványi GT, Nemes B, Gróf I, Fekete T, Kubacková J, Tomori Z, Bánó G, Vizsnyiczai G, Kelemen L. Optically Actuated Soft Microrobot Family for Single-Cell Manipulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401115. [PMID: 38814436 DOI: 10.1002/adma.202401115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/17/2024] [Indexed: 05/31/2024]
Abstract
Precisely controlled manipulation of nonadherent single cells is often a pre-requisite for their detailed investigation. Optical trapping provides a versatile means for positioning cells with submicrometer precision or measuring forces with femto-Newton resolution. A variant of the technique, called indirect optical trapping, enables single-cell manipulation with no photodamage and superior spatial control and stability by relying on optically trapped microtools biochemically bound to the cell. High-resolution 3D lithography enables to prepare such cell manipulators with any predefined shape, greatly extending the number of achievable manipulation tasks. Here, it is presented for the first time a novel family of cell manipulators that are deformable by optical tweezers and rely on their elasticity to hold cells. This provides a more straightforward approach to indirect optical trapping by avoiding biochemical functionalization for cell attachment, and consequently by enabling the manipulated cells to be released at any time. Using the photoresist Ormocomp, the deformations achievable with optical forces in the tens of pN range and present three modes of single-cell manipulation as examples to showcase the possible applications such soft microrobotic tools can offer are characterized. The applications describe here include cell collection, 3D cell imaging, and spatially and temporally controlled cell-cell interaction.
Collapse
Affiliation(s)
- Gergely T Iványi
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, Szeged, 6720, Hungary
| | - Botond Nemes
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Ilona Gróf
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Tamás Fekete
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Jana Kubacková
- Department of Biophysics, Institute of Experimental Physics SAS, Watsonova 47, Košice, 04001, Slovakia
| | - Zoltán Tomori
- Department of Biophysics, Institute of Experimental Physics SAS, Watsonova 47, Košice, 04001, Slovakia
| | - Gregor Bánó
- Department of Biophysics, Faculty of Science, P. J. Šafárik University in Košice, Jesenná 5, Košice, 04154, Slovakia
| | - Gaszton Vizsnyiczai
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
- Department of Biotechnology, University of Szeged, Szeged, 6720, Hungary
| | - Lóránd Kelemen
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
| |
Collapse
|
32
|
Gan Q, Ge Z, Wang X, Dai S, Li N, Wang J, Liu L, Yu H. Stiffness-Tunable Substrate Fabrication by DMD-Based Optical Projection Lithography for Cancer Cell Invasion Studies. IEEE Trans Biomed Eng 2024; 71:2201-2210. [PMID: 38345950 DOI: 10.1109/tbme.2024.3364971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
OBJECTIVE Cancer cell invasion is a critical cause of fatality in cancer patients. Physiologically relevant tumor models play a key role in revealing the mechanisms underlying the invasive behavior of cancer cells. However, most existing models only consider interactions between cells and extracellular matrix (ECM) components while neglecting the role of matrix stiffness in tumor invasion. Here, we propose an effective approach that can construct stiffness-tunable substrates using digital mirror device (DMD)-based optical projection lithography to explore the invasion behavior of cancer cells. The printability, mechanical properties, and cell viability of three-dimensional (3D) models can be tuned by the concentration of prepolymer and the exposure time. The invasion trajectories of gastric cancer cells in tumor models of different stiffness were automatically detected and tracked in real-time using a deep learning algorithm. The results show that tumor models of different mechanical stiffness can yield distinct regulatory effects. Moreover, owing to the biophysical characteristics of the 3D in vitro model, different cellular substructures of cancer cells were induced. The proposed tunable substrate construction method can be used to build various microstructures to achieve simulation of cancer invasion and antitumor screening, which has great potential in promoting personalized therapy.
Collapse
|
33
|
Urbanska M, Guck J. Single-Cell Mechanics: Structural Determinants and Functional Relevance. Annu Rev Biophys 2024; 53:367-395. [PMID: 38382116 DOI: 10.1146/annurev-biophys-030822-030629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
The mechanical phenotype of a cell determines its ability to deform under force and is therefore relevant to cellular functions that require changes in cell shape, such as migration or circulation through the microvasculature. On the practical level, the mechanical phenotype can be used as a global readout of the cell's functional state, a marker for disease diagnostics, or an input for tissue modeling. We focus our review on the current knowledge of structural components that contribute to the determination of the cellular mechanical properties and highlight the physiological processes in which the mechanical phenotype of the cells is of critical relevance. The ongoing efforts to understand how to efficiently measure and control the mechanical properties of cells will define the progress in the field and drive mechanical phenotyping toward clinical applications.
Collapse
Affiliation(s)
- Marta Urbanska
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
34
|
Chen Y, Ni C, Zhang X, Ni Z, Xiang N. High-Throughput Sorting and Single-Cell Mechanotyping by Hydrodynamic Sorting-Mechanotyping Cytometry. SMALL METHODS 2024; 8:e2301195. [PMID: 38213022 DOI: 10.1002/smtd.202301195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/28/2023] [Indexed: 01/13/2024]
Abstract
The existence of many background blood cells hinders the accurate identification of circulating tumor cells (CTCs) in the blood of cancer patients. To unlock this limitation, a hydrodynamic sorting-mechanotyping cytometry (HSMC) integrated with a sorting-concentration chip and a detection chip is proposed for simultaneously achieving the high-throughput cell sorting and the multi-parameter mechanotyping of the sorted tumor cells. The HSMC adopts the spiral inertial microfluidics for label-free sorting of cells in a high-throughput manner, allowing the efficient enrichment of tumor cells from the large background blood cells. Then, the sorted cells are concentrated by the concentration unit and finally passed through the detection unit for hydrodynamic deformation. The HSMC has a high throughput for sorting and detection and can successfully reveal the differences in the cellular mechanical properties. After characterizing and optimizing the single chips, the identification of white blood cells (WBCs) and three types of tumor cells (A549, MCF-7, and MDA-MB-231 cells) is successfully achieved. The identification accuracies for WBCs and different tumor cells are all larger than 94%, while the highest identification accuracy is up to 99.2%. This study envisions that the HSMC will offer an avenue for the analysis of single cell intrinsic mechanics in clinical medicine.
Collapse
Affiliation(s)
- Yao Chen
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China
| | - Chen Ni
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China
| | - Xiaozhe Zhang
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China
| | - Zhonghua Ni
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China
| | - Nan Xiang
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China
| |
Collapse
|
35
|
Golo M, Newman PLH, Kempe D, Biro M. Mechanoimmunology in the solid tumor microenvironment. Biochem Soc Trans 2024; 52:1489-1502. [PMID: 38856041 DOI: 10.1042/bst20231427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
The tumor microenvironment (TME) is a complex and dynamic ecosystem that adjoins the cancer cells within solid tumors and comprises distinct components such as extracellular matrix, stromal and immune cells, blood vessels, and an abundance of signaling molecules. In recent years, the mechanical properties of the TME have emerged as critical determinants of tumor progression and therapeutic response. Aberrant mechanical cues, including altered tissue architecture and stiffness, contribute to tumor progression, metastasis, and resistance to treatment. Moreover, burgeoning immunotherapies hold great promise for harnessing the immune system to target and eliminate solid malignancies; however, their success is hindered by the hostile mechanical landscape of the TME, which can impede immune cell infiltration, function, and persistence. Consequently, understanding TME mechanoimmunology - the interplay between mechanical forces and immune cell behavior - is essential for developing effective solid cancer therapies. Here, we review the role of TME mechanics in tumor immunology, focusing on recent therapeutic interventions aimed at modulating the mechanical properties of the TME to potentiate T cell immunotherapies, and innovative assays tailored to evaluate their clinical efficacy.
Collapse
Affiliation(s)
- Matteo Golo
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter L H Newman
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Daryan Kempe
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Maté Biro
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
36
|
Pouraria H, Houston JP. Elasticity of Carrier Fluid: A Key Factor Affecting Mechanical Phenotyping in Deformability Cytometry. MICROMACHINES 2024; 15:822. [PMID: 39064333 PMCID: PMC11278870 DOI: 10.3390/mi15070822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024]
Abstract
Recently, microfluidics deformability cytometry has emerged as a powerful tool for high-throughput mechanical phenotyping of large populations of cells. These methods characterize cells by their mechanical fingerprints by exerting hydrodynamic forces and monitoring the resulting deformation. These devices have shown great promise for label-free cytometry, yet there is a critical need to improve their accuracy and reconcile any discrepancies with other methods, such as atomic force microscopy. In this study, we employ computational fluid dynamics simulations and uncover how the elasticity of frequently used carrier fluids, such as methylcellulose dissolved in phosphate-buffered saline, is significantly influential to the resulting cellular deformation. We conducted CFD simulations conventionally used within the deformability cytometry field, which neglect fluid elasticity. Subsequently, we incorporated a more comprehensive model that simulates the viscoelastic nature of the carrier fluid. A comparison of the predicted stresses between these two approaches underscores the significance of the emerging elastic stresses in addition to the well-recognized viscous stresses along the channel. Furthermore, we utilize a two-phase flow model to predict the deformation of a promyelocyte (i.e., HL-60 cell type) within a hydrodynamic constriction channel. The obtained results highlight a substantial impact of the elasticity of carrier fluid on cellular deformation and raise questions about the accuracy of mechanical property estimates derived by neglecting elastic stresses.
Collapse
Affiliation(s)
| | - Jessica P. Houston
- Department of Chemical and Materials Engineering, New Mexico State University, Las Cruces, NM 88003, USA;
| |
Collapse
|
37
|
Smith IM, Ursitti JA, Majeti P, Givpoor N, Stemberger MB, Hengen A, Banerjee S, Stains J, Martin SS, Ward C, Stroka KM. High throughput cell mechanotyping of cell response to cytoskeletal modulations using a microfluidic cell deformation system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599307. [PMID: 38948841 PMCID: PMC11212920 DOI: 10.1101/2024.06.17.599307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Cellular mechanical properties influence cellular functions across pathological and physiological systems. The observation of these mechanical properties is limited in part by methods with a low throughput of acquisition or with low accessibility. To overcome these limitations, we have designed, developed, validated, and optimized a microfluidic cellular deformation system (MCDS) capable of mechanotyping suspended cells on a population level at a high throughput rate of ∼300 cells pers second. The MCDS provides researchers with a viable method for efficiently quantifying cellular mechanical properties towards defining prognostic implications of mechanical changes in pathology or screening drugs to modulate cytoskeletal integrity.
Collapse
|
38
|
Möckel C, Beck T, Kaliman S, Abuhattum S, Kim K, Kolb J, Wehner D, Zaburdaev V, Guck J. Estimation of the mass density of biological matter from refractive index measurements. BIOPHYSICAL REPORTS 2024; 4:100156. [PMID: 38718671 PMCID: PMC11090064 DOI: 10.1016/j.bpr.2024.100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/04/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024]
Abstract
The quantification of physical properties of biological matter gives rise to novel ways of understanding functional mechanisms. One of the basic biophysical properties is the mass density (MD). It affects the dynamics in sub-cellular compartments and plays a major role in defining the opto-acoustical properties of cells and tissues. As such, the MD can be connected to the refractive index (RI) via the well known Lorentz-Lorenz relation, which takes into account the polarizability of matter. However, computing the MD based on RI measurements poses a challenge, as it requires detailed knowledge of the biochemical composition of the sample. Here we propose a methodology on how to account for assumptions about the biochemical composition of the sample and respective RI measurements. To this aim, we employ the Biot mixing rule of RIs alongside the assumption of volume additivity to find an approximate relation of MD and RI. We use Monte-Carlo simulations and Gaussian propagation of uncertainty to obtain approximate analytical solutions for the respective uncertainties of MD and RI. We validate this approach by applying it to a set of well-characterized complex mixtures given by bovine milk and intralipid emulsion and employ it to estimate the MD of living zebrafish (Danio rerio) larvae trunk tissue. Our results illustrate the importance of implementing this methodology not only for MD estimations but for many other related biophysical problems, such as mechanical measurements using Brillouin microscopy and transient optical coherence elastography.
Collapse
Affiliation(s)
- Conrad Möckel
- Max Planck Institute for the Science of Light, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Timon Beck
- Max Planck Institute for the Science of Light, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Sara Kaliman
- Max Planck Institute for the Science of Light, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Shada Abuhattum
- Max Planck Institute for the Science of Light, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Kyoohyun Kim
- Max Planck Institute for the Science of Light, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Julia Kolb
- Max Planck Institute for the Science of Light, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniel Wehner
- Max Planck Institute for the Science of Light, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Vasily Zaburdaev
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany; Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
39
|
Chapman M, Rajagopal V, Stewart A, Collins DJ. Critical review of single-cell mechanotyping approaches for biomedical applications. LAB ON A CHIP 2024; 24:3036-3063. [PMID: 38804123 DOI: 10.1039/d3lc00978e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Accurate mechanical measurements of cells has the potential to improve diagnostics, therapeutics and advance understanding of disease mechanisms, where high-resolution mechanical information can be measured by deforming individual cells. Here we evaluate recently developed techniques for measuring cell-scale stiffness properties; while many such techniques have been developed, much of the work examining single-cell stiffness is impacted by difficulties in standardization and comparability, giving rise to large variations in reported mechanical moduli. We highlight the role of underlying mechanical theories driving this variability, and note opportunities to develop novel mechanotyping devices and theoretical models that facilitate convenient and accurate mechanical characterisation. Moreover, many high-throughput approaches are confounded by factors including cell size, surface friction, natural population heterogeneity and convolution of elastic and viscous contributions to cell deformability. We nevertheless identify key approaches based on deformability cytometry as a promising direction for further development, where both high-throughput and accurate single-cell resolutions can be realized.
Collapse
Affiliation(s)
- Max Chapman
- Department of Biomedical Engineering, University of Melbourne, Melbourne, Victoria, Australia.
| | - Vijay Rajagopal
- Department of Biomedical Engineering, University of Melbourne, Melbourne, Victoria, Australia.
| | - Alastair Stewart
- ARC Centre for Personalised Therapeutics Technologies, The University of Melbourne, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, Australia
| | - David J Collins
- Department of Biomedical Engineering, University of Melbourne, Melbourne, Victoria, Australia.
- Graeme Clarke Institute University of Melbourne Parkville, Victoria 3052, Australia
| |
Collapse
|
40
|
Radman BA, Alhameed AMM, Shu G, Yin G, Wang M. Cellular elasticity in cancer: a review of altered biomechanical features. J Mater Chem B 2024; 12:5299-5324. [PMID: 38742281 DOI: 10.1039/d4tb00328d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
A large number of studies have shown that changes in biomechanical characteristics are an important indicator of tumor transformation in normal cells. Elastic deformation is one of the more studied biomechanical features of tumor cells, which plays an important role in tumourigenesis and development. Altered cell elasticity often brings many indications. This manuscript reviews the effects of altered cellular elasticity on cell characteristics, including adhesion viscosity, migration, proliferation, and differentiation elasticity and stiffness. Also, the physical factors that may affect cell elasticity, such as temperature, cell height, cell-viscosity, and aging, are summarized. Then, the effects of cell-matrix, cytoskeleton, in vitro culture medium, and cell-substrate with different three-dimensional structures on cell elasticity during cell tumorigenesis are outlined. Importantly, we summarize the current signaling pathways that may affect cellular elasticity, as well as tests for cellular elastic deformation. Finally, we summarize current hybrid materials: polymer-polymer, protein-protein, and protein-polymer hybrids, also, nano-delivery strategies that target cellular resilience and cases that are at least in clinical phase 1 trials. Overall, the behavior of cancer cell elasticity is modulated by biological, chemical, and physical changes, which in turn have the potential to alter cellular elasticity, and this may be an encouraging prediction for the future discovery of cancer therapies.
Collapse
Affiliation(s)
- Bakeel A Radman
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.
- Department of Biology, College of Science and Education, Albaydha University, Yemen
| | | | - Guang Shu
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, 410013, China
- China-Africa Research Center of Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, 410013, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Maonan Wang
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.
| |
Collapse
|
41
|
Chojowski R, Schwarz US, Ziebert F. The role of the nucleus for cell mechanics: an elastic phase field approach. SOFT MATTER 2024; 20:4488-4503. [PMID: 38804018 DOI: 10.1039/d4sm00345d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The nucleus of eukaryotic cells typically makes up around 30% of the cell volume and has significantly different mechanics, which can make it effectively up to ten times stiffer than the surrounding cytoplasm. Therefore it is an important element for cell mechanics, but a quantitative understanding of its mechanical role during whole cell dynamics is largely missing. Here we demonstrate that elastic phase fields can be used to describe dynamical cell processes in adhesive or confining environments in which the nucleus acts as a stiff inclusion in an elastic cytoplasm. We first introduce and verify our computational method and then study several prevalent cell-mechanical measurement methods. For cells on adhesive patterns, we find that nuclear stress is shielded by the adhesive pattern. For cell compression between two parallel plates, we obtain force-compression curves that allow us to extract an effective modulus for the cell-nucleus composite. For micropipette aspiration, the effect of the nucleus on the effective modulus is found to be much weaker, highlighting the complicated interplay between extracellular geometry and cell mechanics that is captured by our approach. We also show that our phase field approach can be used to investigate the effects of Kelvin-Voigt-type viscoelasticity and cortical tension.
Collapse
Affiliation(s)
- Robert Chojowski
- Institute for Theoretical Physics, Heidelberg University, Philosophenweg 19, 69120 Heidelberg, Germany.
- BioQuant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Ulrich S Schwarz
- Institute for Theoretical Physics, Heidelberg University, Philosophenweg 19, 69120 Heidelberg, Germany.
- BioQuant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Falko Ziebert
- Institute for Theoretical Physics, Heidelberg University, Philosophenweg 19, 69120 Heidelberg, Germany.
- BioQuant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| |
Collapse
|
42
|
Hou X, Ouyang J, Tang L, Wu P, Deng X, Yan Q, Shi L, Fan S, Fan C, Guo C, Liao Q, Li Y, Xiong W, Li G, Zeng Z, Wang F. KCNK1 promotes proliferation and metastasis of breast cancer cells by activating lactate dehydrogenase A (LDHA) and up-regulating H3K18 lactylation. PLoS Biol 2024; 22:e3002666. [PMID: 38905316 PMCID: PMC11192366 DOI: 10.1371/journal.pbio.3002666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 05/07/2024] [Indexed: 06/23/2024] Open
Abstract
Breast cancer is the most prevalent malignancy and the most significant contributor to mortality in female oncology patients. Potassium Two Pore Domain Channel Subfamily K Member 1 (KCNK1) is differentially expressed in a variety of tumors, but the mechanism of its function in breast cancer is unknown. In this study, we found for the first time that KCNK1 was significantly up-regulated in human breast cancer and was correlated with poor prognosis in breast cancer patients. KCNK1 promoted breast cancer proliferation, invasion, and metastasis in vitro and vivo. Further studies unexpectedly revealed that KCNK1 increased the glycolysis and lactate production in breast cancer cells by binding to and activating lactate dehydrogenase A (LDHA), which promoted histones lysine lactylation to induce the expression of a series of downstream genes and LDHA itself. Notably, increased expression of LDHA served as a vicious positive feedback to reduce tumor cell stiffness and adhesion, which eventually resulted in the proliferation, invasion, and metastasis of breast cancer. In conclusion, our results suggest that KCNK1 may serve as a potential breast cancer biomarker, and deeper insight into the cancer-promoting mechanism of KCNK1 may uncover a novel therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Xiangchan Hou
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jiawei Ouyang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Le Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Pan Wu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiangying Deng
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Shi
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunmei Fan
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Can Guo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Medicine, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Fuyan Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| |
Collapse
|
43
|
Rahmati N, Keshavarz Motamed P, Maftoon N. Numerical study of ultra-large von Willebrand factor multimers in coagulopathy. Biomech Model Mechanobiol 2024; 23:737-756. [PMID: 38217745 DOI: 10.1007/s10237-023-01803-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/30/2023] [Indexed: 01/15/2024]
Abstract
An excessive von Willebrand factor (VWF) secretion, coupled with a moderate to severe deficiency of ADAMTS13 activity, serves as a linking mechanism between inflammation to thrombosis. The former facilitates platelet adhesion to the vessel wall and the latter is required to cleave VWF multimers. As a result, the ultra-large VWF (UL-VWF) multimers released by Weibel-Palade bodies remain uncleaved. In this study, using a computational model based on first principles, we quantitatively show how the uncleaved UL-VWF multimers interact with the blood cells to initiate microthrombosis. We observed that platelets first adhere to unfolded and stretched uncleaved UL-VWF multimers anchored to the microvessel wall. By the end of this initial adhesion phase, the UL-VWF multimers and platelets make a mesh-like trap in which the red blood cells increasingly accumulate to initiate a gradually growing microthrombosis. Although high-shear rate and blood flow velocity are required to activate platelets and unfold the UL-VWFs, during the initial adhesion phase, the blood velocity drastically drops after thrombosis, and as a result, the wall shear stress is elevated near UL-VWF roots, and the pressure drops up to 6 times of the healthy condition. As the time passes, these trends progressively continue until the microthrombosis fully develops and the effective size of the microthrombosis and these flow quantities remain almost constant. Our findings quantitatively demonstrate the potential role of UL-VWF in coagulopathy.
Collapse
Affiliation(s)
- Nahid Rahmati
- Department of Systems Design Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
- Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON, Canada
| | - Pouyan Keshavarz Motamed
- Department of Systems Design Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
- Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON, Canada
| | - Nima Maftoon
- Department of Systems Design Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada.
- Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
44
|
Rahmati N, Maftoon N. Computational analysis of cancer cell adhesion in curved vessels affected by wall shear stress for prediction of metastatic spreading. Front Bioeng Biotechnol 2024; 12:1393413. [PMID: 38860135 PMCID: PMC11163055 DOI: 10.3389/fbioe.2024.1393413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/19/2024] [Indexed: 06/12/2024] Open
Abstract
Introduction: The dynamics of circulating tumor cells (CTCs) within blood vessels play a pivotal role in predicting metastatic spreading of cancer within the body. However, the limited understanding and method to quantitatively investigate the influence of vascular architecture on CTC dynamics hinders our ability to predict metastatic process effectively. To address this limitation, the present study was conducted to investigate the influence of blood vessel tortuosity on the behaviour of CTCs, focusing specifically on establishing methods and examining the role of shear stress in CTC-vessel wall interactions and its subsequent impact on metastasis. Methods: We computationally simulated CTC behaviour under various shear stress conditions induced by vessel tortuosity. Our computational model, based on the lattice Boltzmann method (LBM) and a coarse-grained spectrin-link membrane model, efficiently simulates blood plasma dynamics and CTC deformability. The model incorporates fluid-structure interactions and receptor-ligand interactions crucial for CTC adhesion using the immersed boundary method (IBM). Results: Our findings reveal that uniform shear stress in straight vessels leads to predictable CTC-vessel interactions, whereas in curved vessels, asymmetrical flow patterns and altered shear stress create distinct adhesion dynamics, potentially influencing CTC extravasation. Quantitative analysis shows a 25% decrease in the wall shear stress in low-shear regions and a 58.5% increase in the high-shear region. We observed high-shear regions in curved vessels to be potential sites for increased CTC adhesion and extravasation, facilitated by elevated endothelial expression of adhesion molecules. This phenomenon correlates with the increased number of adhesion bonds, which rises to approximately 40 in high-shear regions, compared to around 12 for straight vessels and approximately 5-6 in low-shear regions. The findings also indicate an optimal cellular stiffness necessary for successful CTC extravasation in curved vessels. Discussion: By the quantitative assessment of the risk of CTC extravasation as a function of vessel tortuosity, our study offers a novel tool for the prediction of metastasis risk to support the development of personalized therapeutic interventions based on individual vascular characteristics and tumor cell properties.
Collapse
Affiliation(s)
- Nahid Rahmati
- Department of Systems Design Engineering, University of Waterloo, Waterloo, ON, Canada
- Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON, Canada
| | - Nima Maftoon
- Department of Systems Design Engineering, University of Waterloo, Waterloo, ON, Canada
- Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
45
|
Hartmann B, Fleischhauer L, Nicolau M, Jensen THL, Taran FA, Clausen-Schaumann H, Reuten R. Profiling native pulmonary basement membrane stiffness using atomic force microscopy. Nat Protoc 2024; 19:1498-1528. [PMID: 38429517 DOI: 10.1038/s41596-024-00955-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 11/27/2023] [Indexed: 03/03/2024]
Abstract
Mammalian cells sense and react to the mechanics of their immediate microenvironment. Therefore, the characterization of the biomechanical properties of tissues with high spatial resolution provides valuable insights into a broad variety of developmental, homeostatic and pathological processes within living organisms. The biomechanical properties of the basement membrane (BM), an extracellular matrix (ECM) substructure measuring only ∼100-400 nm across, are, among other things, pivotal to tumor progression and metastasis formation. Although the precise assignment of the Young's modulus E of such a thin ECM substructure especially in between two cell layers is still challenging, biomechanical data of the BM can provide information of eminent diagnostic potential. Here we present a detailed protocol to quantify the elastic modulus of the BM in murine and human lung tissue, which is one of the major organs prone to metastasis. This protocol describes a streamlined workflow to determine the Young's modulus E of the BM between the endothelial and epithelial cell layers shaping the alveolar wall in lung tissues using atomic force microscopy (AFM). Our step-by-step protocol provides instructions for murine and human lung tissue extraction, inflation of these tissues with cryogenic cutting medium, freezing and cryosectioning of the tissue samples, and AFM force-map recording. In addition, it guides the reader through a semi-automatic data analysis procedure to identify the pulmonary BM and extract its Young's modulus E using an in-house tailored user-friendly AFM data analysis software, the Center for Applied Tissue Engineering and Regenerative Medicine processing toolbox, which enables automatic loading of the recorded force maps, conversion of the force versus piezo-extension curves to force versus indentation curves, calculation of Young's moduli and generation of Young's modulus maps, where the pulmonary BM can be identified using a semi-automatic spatial filtering tool. The entire protocol takes 1-2 d.
Collapse
Affiliation(s)
- Bastian Hartmann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Lutz Fleischhauer
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Monica Nicolau
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Thomas Hartvig Lindkær Jensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | - Florin-Andrei Taran
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Hauke Clausen-Schaumann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany.
- Center for Nanoscience, Munich, Germany.
| | - Raphael Reuten
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany.
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
46
|
Zhu L, Wang H. Cholesterol-regulated cellular stiffness may enhance evasion of NK cell-mediated cytotoxicity in gastric cancer stem cells. FEBS Open Bio 2024; 14:855-866. [PMID: 38494433 PMCID: PMC11073496 DOI: 10.1002/2211-5463.13793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/14/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024] Open
Abstract
Gastric cancer has a high rate of recurrence, and as such, immunotherapy strategies are being investigated as a potential therapeutic strategy. Although the involvement of immune checkpoints in immunotherapy is well studied, biomechanical cues, such as target cell stiffness, have not yet been subject to the same level of investigation. Changes in the cholesterol content of the cell membrane directly influence tumor cell stiffness. Here, we investigated the effect of cholesterol on NK cell-mediated killing of gastric cancer stem-like cells. We report that surviving tumor cells with stem-like properties elevated cholesterol metabolism to evade NK cell cytotoxicity. Inhibition of cholesterol metabolism enhances NK cell-mediated killing of gastric cancer stem-like cells, highlighting a potential avenue for improving immunotherapy efficacy. This study suggests a possible effect of cancer cell stiffness on immune evasion and offers insights into enhancing immunotherapeutic strategies against tumors.
Collapse
Affiliation(s)
- Lijuan Zhu
- Department of Radiation Oncology (II)The First Affiliated Hospital of Jinzhou Medical UniversityChina
| | - Hongjin Wang
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Jinzhou Medical UniversityChina
| |
Collapse
|
47
|
Reichel F, Goswami R, Girardo S, Guck J. High-throughput viscoelastic characterization of cells in hyperbolic microchannels. LAB ON A CHIP 2024; 24:2440-2453. [PMID: 38600866 DOI: 10.1039/d3lc01061a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Extensive research has demonstrated the potential of cell viscoelastic properties as intrinsic indicators of cell state, functionality, and disease. For this, several microfluidic techniques have been developed to measure cell viscoelasticity with high-throughput. However, current microchannel designs introduce complex stress distributions on cells, leading to inaccuracies in determining the stress-strain relationship and, consequently, the viscoelastic properties. Here, we introduce a novel approach using hyperbolic microchannels that enable precise measurements under a constant extensional stress and offer a straightforward stress-strain relationship, while operating at a measurement rate of up to 100 cells per second. We quantified the stresses acting in the channels using mechanical calibration particles made from polyacrylamide (PAAm) and found that the measurement buffer, a solution of methyl cellulose and phosphate buffered saline, shows strain-thickening following a power law up to 200 s-1. By measuring oil droplets with varying viscosities, we successfully detected changes in the relaxation times of the droplets and our approach could be used to get the interfacial tension and viscosity of liquid-liquid droplet systems from the same measurement. We further applied this methodology to PAAm microgel beads, demonstrating the accurate recovery of Young's moduli and the near-ideal elastic behavior of the beads. To explore the influence of altered cell viscoelasticity, we treated HL60 human leukemia cells with latrunculin B and nocodazole, resulting in clear changes in cell stiffness while relaxation times were only minimally affected. In conclusion, our approach offers a streamlined and time-efficient solution for assessing the viscoelastic properties of large cell populations and other microscale soft particles.
Collapse
Affiliation(s)
- Felix Reichel
- Max Planck Institute for the Science of Light, Erlangen, Germany.
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ruchi Goswami
- Max Planck Institute for the Science of Light, Erlangen, Germany.
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Salvatore Girardo
- Max Planck Institute for the Science of Light, Erlangen, Germany.
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany.
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
48
|
Conti S, Venturini V, Cañellas-Socias A, Cortina C, Abenza JF, Stephan-Otto Attolini C, Middendorp Guerra E, Xu CK, Li JH, Rossetti L, Stassi G, Roca-Cusachs P, Diz-Muñoz A, Ruprecht V, Guck J, Batlle E, Labernadie A, Trepat X. Membrane to cortex attachment determines different mechanical phenotypes in LGR5+ and LGR5- colorectal cancer cells. Nat Commun 2024; 15:3363. [PMID: 38637494 PMCID: PMC11026456 DOI: 10.1038/s41467-024-47227-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/26/2024] [Indexed: 04/20/2024] Open
Abstract
Colorectal cancer (CRC) tumors are composed of heterogeneous and plastic cell populations, including a pool of cancer stem cells that express LGR5. Whether these distinct cell populations display different mechanical properties, and how these properties might contribute to metastasis is poorly understood. Using CRC patient derived organoids (PDOs), we find that compared to LGR5- cells, LGR5+ cancer stem cells are stiffer, adhere better to the extracellular matrix (ECM), move slower both as single cells and clusters, display higher nuclear YAP, show a higher survival rate in response to mechanical confinement, and form larger transendothelial gaps. These differences are largely explained by the downregulation of the membrane to cortex attachment proteins Ezrin/Radixin/Moesin (ERMs) in the LGR5+ cells. By analyzing single cell RNA-sequencing (scRNA-seq) expression patterns from a patient cohort, we show that this downregulation is a robust signature of colorectal tumors. Our results show that LGR5- cells display a mechanically dynamic phenotype suitable for dissemination from the primary tumor whereas LGR5+ cells display a mechanically stable and resilient phenotype suitable for extravasation and metastatic growth.
Collapse
Affiliation(s)
- Sefora Conti
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Valeria Venturini
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Adrià Cañellas-Socias
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomedica en Red de Cancer (CIBERONC), Barcelona, Spain
| | - Carme Cortina
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomedica en Red de Cancer (CIBERONC), Barcelona, Spain
| | - Juan F Abenza
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Emily Middendorp Guerra
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomedica en Red de Cancer (CIBERONC), Barcelona, Spain
| | - Catherine K Xu
- Max Planck Institute for the Science of Light, Erlangen, Germany
| | - Jia Hui Li
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Leone Rossetti
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Giorgio Stassi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Facultat de Medicina, University of Barcelona (UB), Barcelona, Spain
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Verena Ruprecht
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany
- Department of Physics, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Max-Planck Zentrum für Physik und Medizin, Erlangen, Germany
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomedica en Red de Cancer (CIBERONC), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| | - Anna Labernadie
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Principe Felipe (CIPF), Valencia, Spain.
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.
- Facultat de Medicina, University of Barcelona (UB), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain.
| |
Collapse
|
49
|
Coker ZN, Troyanova-Wood M, Steelman ZA, Ibey BL, Bixler JN, Scully MO, Yakovlev VV. Brillouin microscopy monitors rapid responses in subcellular compartments. PHOTONIX 2024; 5:9. [PMID: 38618142 PMCID: PMC11006764 DOI: 10.1186/s43074-024-00123-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/12/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024]
Abstract
Measurements and imaging of the mechanical response of biological cells are critical for understanding the mechanisms of many diseases, and for fundamental studies of energy, signal and force transduction. The recent emergence of Brillouin microscopy as a powerful non-contact, label-free way to non-invasively and non-destructively assess local viscoelastic properties provides an opportunity to expand the scope of biomechanical research to the sub-cellular level. Brillouin spectroscopy has recently been validated through static measurements of cell viscoelastic properties, however, fast (sub-second) measurements of sub-cellular cytomechanical changes have yet to be reported. In this report, we utilize a custom multimodal spectroscopy system to monitor for the very first time the rapid viscoelastic response of cells and subcellular structures to a short-duration electrical impulse. The cytomechanical response of three subcellular structures - cytoplasm, nucleoplasm, and nucleoli - were monitored, showing distinct mechanical changes despite an identical stimulus. Through this pioneering transformative study, we demonstrate the capability of Brillouin spectroscopy to measure rapid, real-time biomechanical changes within distinct subcellular compartments. Our results support the promising future of Brillouin spectroscopy within the broad scope of cellular biomechanics.
Collapse
Affiliation(s)
- Zachary N. Coker
- Department of Physics & Astronomy, Texas A&M University, 4242 TAMU, College Station, TX 77843 USA
- SAIC, Fort Sam Houston, TX 78234 USA
| | | | - Zachary A. Steelman
- Air Force Research Laboratory, JBSA Fort Sam Houston, Fort Sam Houston, TX 78234 USA
| | - Bennett L. Ibey
- Air Force Research Laboratory, JBSA Fort Sam Houston, Fort Sam Houston, TX 78234 USA
| | - Joel N. Bixler
- Air Force Research Laboratory, JBSA Fort Sam Houston, Fort Sam Houston, TX 78234 USA
| | - Marlan O. Scully
- Department of Physics & Astronomy, Texas A&M University, 4242 TAMU, College Station, TX 77843 USA
- Institute for Quantum Science and Engineering, Texas A&M University, College Station, TX 77843 USA
| | - Vladislav V. Yakovlev
- Department of Physics & Astronomy, Texas A&M University, 4242 TAMU, College Station, TX 77843 USA
- Institute for Quantum Science and Engineering, Texas A&M University, College Station, TX 77843 USA
- Department of Biomedical Engineering, Texas A&M University, 3120 TAMU, 101 Bizzell Street, College Station, TX 77843 USA
| |
Collapse
|
50
|
Combriat T, Olsen PA, Låstad SB, Malthe-Sørenssen A, Krauss S, Dysthe DK. Acoustic Wave-Induced Stroboscopic Optical Mechanotyping of Adherent Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307929. [PMID: 38417124 DOI: 10.1002/advs.202307929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/02/2024] [Indexed: 03/01/2024]
Abstract
In this study, a novel, high content technique using a cylindrical acoustic transducer, stroboscopic fast imaging, and homodyne detection to recover the mechanical properties (dynamic shear modulus) of living adherent cells at low ultrasonic frequencies is presented. By analyzing the micro-oscillations of cells, whole populations are simultaneously mechanotyped with sub-cellular resolution. The technique can be combined with standard fluorescence imaging allowing to further cross-correlate biological and mechanical information. The potential of the technique is demonstrated by mechanotyping co-cultures of different cell types with significantly different mechanical properties.
Collapse
Affiliation(s)
- Thomas Combriat
- Njord Centre, Department of Physics, University of Oslo, P.O. Box 1048 Blindern, Oslo, 0316, Norway
- Hybrid Technology Hub, University of Oslo, Institute of Basic Medical Sciences P.O. Box 1110 Blindern, Oslo, 0317, Norway
- Center for Computing in Science Education, University of Oslo, P.O. Box 1048 Blindern, Oslo, 0316, Norway
| | - Petter Angell Olsen
- Hybrid Technology Hub, University of Oslo, Institute of Basic Medical Sciences P.O. Box 1110 Blindern, Oslo, 0317, Norway
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950, Nydalen, Oslo, 0424, Norway
| | - Silja Borring Låstad
- Njord Centre, Department of Physics, University of Oslo, P.O. Box 1048 Blindern, Oslo, 0316, Norway
| | - Anders Malthe-Sørenssen
- Njord Centre, Department of Physics, University of Oslo, P.O. Box 1048 Blindern, Oslo, 0316, Norway
- Center for Computing in Science Education, University of Oslo, P.O. Box 1048 Blindern, Oslo, 0316, Norway
| | - Stefan Krauss
- Hybrid Technology Hub, University of Oslo, Institute of Basic Medical Sciences P.O. Box 1110 Blindern, Oslo, 0317, Norway
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950, Nydalen, Oslo, 0424, Norway
| | - Dag Kristian Dysthe
- Njord Centre, Department of Physics, University of Oslo, P.O. Box 1048 Blindern, Oslo, 0316, Norway
| |
Collapse
|