1
|
Zhou H, Liu R, Xu Y, Fan J, Liu X, Chen L, Wei Q. Viscoelastic mechanics of living cells. Phys Life Rev 2025; 53:91-116. [PMID: 40043484 DOI: 10.1016/j.plrev.2025.02.004] [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: 02/11/2025] [Accepted: 02/25/2025] [Indexed: 05/18/2025]
Abstract
In cell mechanotransduction, cells respond to external forces or to perceive mechanical properties of their supporting substrates by remodeling themselves. This ability is endowed by modulating cells' viscoelastic properties, which dominates over various complex cellular processes. The viscoelasticity of living cells, a concept adapted from rheology, exhibits substantially spatial and temporal variability. This review aims not only to discuss the rheological properties of cells but also to clarify the complexity of cellular rheology, emphasizing its dependence on both the size scales and time scales of the measurements. Like typical viscoelastic materials, the storage and loss moduli of cells often exhibit robust power-law rheological characteristics with respect to loading frequency. This intrinsic feature is consistent across cell types and is attributed to internal structures, such as cytoskeleton, cortex, cytoplasm and nucleus, all of which contribute to the complexity of cellular rheology. Moreover, the rheological properties of cells are dynamic and play a crucial role in various cellular and tissue functions. In this review, we focus on elucidating time- and size-dependent aspects of cell rheology, the origins of intrinsic rheological properties and how these properties adapt to cellular functions, with the goal of interpretation of rheology into the language of cell biology.
Collapse
Affiliation(s)
- Hui Zhou
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Ruye Liu
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Yizhou Xu
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jierui Fan
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xinyue Liu
- Shanghai Institute of Applied Mathematics and Mechanics, School of Mechanics and Engineering Science, Shanghai University, Shanghai, 200072, China
| | - Longquan Chen
- School of Physics, University of Electronic Science and Technology of China, Chengdu 611731, China.
| | - Qiang Wei
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
2
|
Carrillo JA, Lorenzi T, Macfarlane FR. Spatial Segregation Across Travelling Fronts in Individual-Based and Continuum Models for the Growth of Heterogeneous Cell Populations. Bull Math Biol 2025; 87:77. [PMID: 40388053 PMCID: PMC12089248 DOI: 10.1007/s11538-025-01452-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/15/2025] [Indexed: 05/20/2025]
Abstract
We consider a partial differential equation model for the growth of heterogeneous cell populations subdivided into multiple distinct discrete phenotypes. In this model, cells preferentially move towards regions where they are less compressed, and thus their movement occurs down the gradient of the cellular pressure. The cellular pressure is defined as a weighted sum of the densities (i.e. the volume fractions) of cells with different phenotypes. To translate into mathematical terms the idea that cells with distinct phenotypes have different morphological and mechanical properties, both the cell mobility and the weighted amount the cells contribute to the cellular pressure vary with their phenotype. We formally derive this model as the continuum limit of an on-lattice individual-based model, where cells are represented as single agents undergoing a branching biased random walk corresponding to phenotype-dependent and pressure-regulated cell division, death, and movement. Then, we study travelling wave solutions whereby cells with different phenotypes are spatially segregated across the invading front. Finally, we report on numerical simulations of the two models, demonstrating excellent agreement between them and the travelling wave analysis. The results presented here indicate that inter-cellular variability in mobility can support the maintenance of spatial segregation across invading fronts, whereby cells with a higher mobility drive invasion by occupying regions closer to the front edge.
Collapse
Affiliation(s)
| | - Tommaso Lorenzi
- Department of Mathematical Sciences "G. L. Lagrange", Politecnico di Torino, Turin, Italy
| | - Fiona R Macfarlane
- School of Mathematics and Statistics, University of St Andrews, St Andrews, UK
- Applied BioSimulation, Certara (UK), Sheffield, UK
| |
Collapse
|
3
|
Lv CL, Li B. Interface morphodynamics in living tissues. SOFT MATTER 2025; 21:3670-3687. [PMID: 40226989 DOI: 10.1039/d5sm00145e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Interfaces between distinct tissues or between tissues and environments are common in multicellular organisms. The evolution and stability of these interfaces are essential for tissue development, and their dysfunction can lead to diseases such as cancer. Mounting efforts, either theoretical or experimental, have been devoted to uncovering the morphodynamics of tissue interfaces. Here, we review the recent progress of studies on interface morphodynamics. The regulatory mechanisms governing interface evolution are dissected, with a focus on adhesion, cortical tension, cell activity, extracellular matrix, and microenvironment. We examine the methodologies used to study morphodynamics, emphasizing the characteristics of experimental techniques and theoretical models. Finally, we explore the broader implications of interface morphodynamics in tissue morphogenesis and diseases, offering a comprehensive perspective on this rapidly developing field.
Collapse
Affiliation(s)
- Cheng-Lin Lv
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China.
| | - Bo Li
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China.
- Mechano-X Institute, Department of Engineering Mechanics, Tsinghua University, Beijing, China
- State Key Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, China
| |
Collapse
|
4
|
Li Z, Xiao C, Yang X, Li Z. Progress in the mechanical properties of nanoparticles for tumor-targeting delivery. Chem Soc Rev 2025. [PMID: 40341776 DOI: 10.1039/d3cs00912b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Cancer nanomedicines have attracted significant attention in the past several decades, and the physicochemical properties, such as the size, shape, composition, surface charge, hydrophobicity, and mechanical properties, of nanoparticles have been optimized for potent cancer therapy. Since publishing our 2020 tutorial review "Influence of nanomedicine mechanical properties on tumor targeting delivery" in Chemical Society Reviews, substantial advancements have been made in understanding the role of mechanical properties in cancer nanomedicine. Notably, in vivo transport processes that are dependent on the mechanical properties of nanomedicine, including long circulation, tumor accumulation, and deep penetration, have been extensively studied using various nano-drug delivery systems. These studies have demonstrated that leveraging these mechanical properties can significantly enhance the antitumor efficacy of nanomedicine. In this review, we categorize the advancements in the mechanical properties of cancer nanomedicine into three distinct themes: the interactions between nanoparticles with varied mechanical properties and cells (2002 - present), the impact of these properties on in vivo delivery processes (2007 - present), and the strategic use of mechanical properties to boost cancer therapy (2023 - present). We analyze how different mechanical properties of organic, inorganic, hybrid, and biological nanoparticles affect their delivery processes at the macroscopic level, i.e., in tissues, organs and cells. At the microscopic level, their biological and physical interactions with biological barriers, physiological structures, cell membranes, organelles, and other structures reveal the potential mechanism of nanoparticles' mechanical properties in determining their antitumor efficacy. Furthermore, we address the current challenges and future prospects in the mechanical properties of cancer nanomedicine, as well as the clinical translation potential of nanoparticles with diverse mechanical characteristics.
Collapse
Affiliation(s)
- Zheng Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
| | - Chen Xiao
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
| | - Xiangliang Yang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Zifu Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
5
|
Hilai K, Grubich D, Akrawi M, Zhu H, Zaghloul R, Shi C, Do M, Zhu D, Zhang J. Mechanical Evolution of Metastatic Cancer Cells in 3D Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2403242. [PMID: 40116569 PMCID: PMC12051740 DOI: 10.1002/smll.202403242] [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: 04/22/2024] [Revised: 01/01/2025] [Indexed: 03/23/2025]
Abstract
Cellular biomechanics plays a critical role in cancer metastasis and tumor progression. Existing studies on cancer cell biomechanics are mostly conducted in flat 2D conditions, where cells' behavior can differ considerably from those in 3D physiological environments. Despite great advances in developing 3D in vitro models, probing cellular elasticity in 3D conditions remains a major challenge for existing technologies. In this work, optical Brillouin microscopy is utilized to longitudinally acquire mechanical images of growing cancerous spheroids over the period of 8 days. The dense mechanical mapping from Brillouin microscopy enables us to extract spatially resolved and temporally evolving mechanical features that were previously inaccessible. Using an established machine learning algorithm, it is demonstrated that incorporating these extracted mechanical features significantly improves the classification accuracy of cancer cells, from 74% to 95%. Building on this finding, a deep learning pipeline capable of accurately differentiating cancerous spheroids from normal ones solely using Brillouin images have been developed, suggesting the mechanical features of cancer cells can potentially serve as a new biomarker in cancer classification and detection.
Collapse
Affiliation(s)
- Karlin Hilai
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
| | - Daniil Grubich
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
| | - Marcus Akrawi
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
- Department of Physics and AstronomyWayne State UniversityDetroitMI48201USA
| | - Hui Zhu
- Department of Computer ScienceWayne State UniversityDetroitMI48202USA
| | - Razanne Zaghloul
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
| | - Chenjun Shi
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
- Department of Biomedical EngineeringInstitute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48823USA
| | - Man Do
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
| | - Dongxiao Zhu
- Department of Computer ScienceWayne State UniversityDetroitMI48202USA
| | - Jitao Zhang
- Department of Biomedical EngineeringWayne State UniversityDetroitMI48202USA
- Department of Biomedical EngineeringInstitute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48823USA
| |
Collapse
|
6
|
Chisolm SJ, Guo E, Subramaniam V, Schulze KD, Angelini TE. Elastic modulus versus cell packing density in MDCK epithelial monolayers. J Biomech 2025; 184:112648. [PMID: 40157056 DOI: 10.1016/j.jbiomech.2025.112648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/18/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
The elastic moduli of tissues are connected to their states of health and function. The epithelial monolayer is a simple, minimal, tissue model that is often used to gain understanding of mechanical behavior at the cellular or multi-cellular scale. Here we investigate how the elastic modulus of Madin Darby Canine Kidney (MDCK) cells depends on their packing density. Rather than measuring elasticity at the sub-cellular scale with local probes, we characterize the monolayer at the multi-cellular scale, as one would a thin slab of elastic material. We use a micro-indentation system to apply gentle forces to the apical side of MDCK monolayers, applying a normal force to approximately 100 cells in each experiment. In low-density confluent monolayers, we find that the elastic modulus decreases with increasing cell density. At high densities, the modulus appears to plateau. This finding will help guide our understanding of known collective behaviors in epithelial monolayers and other tissues where variations in cell packing density are correlated with cell motion.
Collapse
Affiliation(s)
- Steven J Chisolm
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32605, United States.
| | - Emily Guo
- Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, United States.
| | - Vignesh Subramaniam
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32605, United States.
| | - Kyle D Schulze
- Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, United States.
| | - Thomas E Angelini
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32605, United States; Department of Materials Science and Engineering, University of Florida, Gainesville, FL 32605, United States; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32605, United States.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Xiu J, Xue R, Duan X, Yao F, Liu X, Meng F, Xiong C, Huang J. Mechanical characterization of nonlinear elasticity of growing intestinal organoids with a microinjection method. Acta Biomater 2025; 196:271-280. [PMID: 40032216 DOI: 10.1016/j.actbio.2025.02.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
Mechanical properties of intestinal organoids are crucial for intestinal development, homeostatic renewal, and pathogenesis. However, characterizing these properties remains challenging. Here, we developed a microinjection-based method to quantify the growth time-dependent nonlinear elasticity of intestinal organoids. With aid of the neo-Hookean hyperelastic constitutive model, we discovered that the global elastic modulus of intestinal organoids increased linearly during the early stages of culture, followed by a sharp rise, indicating a time-dependent nonlinear hardening behaviour during growth. The global modulus of intestinal organoids was found to correlate with the cell phenotype ratio, revealing a significant relationship between mechanical properties and biological phenotypes. Furthermore, we developed a biomechanical model on the basis of the unsteady Bernoulli equation to quantitatively explore the global mechanical responses of intestinal organoids, which showed good agreement with the experimental data. The work not only elucidated the mechanical response and modulus characteristics of small intestinal organoids from a biomechanical perspective, but also presented a new microinjection-based methodology for quantifying the mechanical properties of organoids, offering significant potential for various organoid-related applications. STATEMENT OF SIGNIFICANCE: Mechanical properties of intestinal organoids are essential for intestinal development, homeostatic renewal, and pathogenesis. However, how to quantitatively characterize their global mechanical properties remains challenging. Here, we developed a new microinjection-based experimental platform to quantify spatiotemporal dynamics of mechanical responses and global elasticity of intestinal organoids. Unlike traditional nanoindentation methods, the proposed characterization technique can quantitatively measure the global mechanical properties of organoids, which is crucial for detecting the inherent relationship between the global mechanical properties and the biological phenotypes of organoids. Likewise, it established a methodological foundation for revealing the mechanobiological characteristics associated with the growth and development of various organoids. This can enhance our understanding of mechanobiological mechanisms of organoids and is beneficial for various organoid-related applications.
Collapse
Affiliation(s)
- Jidong Xiu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Rui Xue
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Xiaocen Duan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Fangyun Yao
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Fanlu Meng
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China.
| | - Chunyang Xiong
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
10
|
Brückner DB, Hannezo E. Tissue Active Matter: Integrating Mechanics and Signaling into Dynamical Models. Cold Spring Harb Perspect Biol 2025; 17:a041653. [PMID: 38951023 PMCID: PMC11960702 DOI: 10.1101/cshperspect.a041653] [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: 07/03/2024]
Abstract
The importance of physical forces in the morphogenesis, homeostatic function, and pathological dysfunction of multicellular tissues is being increasingly characterized, both theoretically and experimentally. Analogies between biological systems and inert materials such as foams, gels, and liquid crystals have provided striking insights into the core design principles underlying multicellular organization. However, these connections can seem surprising given that a key feature of multicellular systems is their ability to constantly consume energy, providing an active origin for the forces that they produce. Key emerging questions are, therefore, to understand whether and how this activity grants tissues novel properties that do not have counterparts in classical materials, as well as their consequences for biological function. Here, we review recent discoveries at the intersection of active matter and tissue biology, with an emphasis on how modeling and experiments can be combined to understand the dynamics of multicellular systems. These approaches suggest that a number of key biological tissue-scale phenomena, such as morphogenetic shape changes, collective migration, or fate decisions, share unifying design principles that can be described by physical models of tissue active matter.
Collapse
Affiliation(s)
- David B Brückner
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Edouard Hannezo
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| |
Collapse
|
11
|
Faure LM, Venturini V, Roca-Cusachs P. Cell compression - relevance, mechanotransduction mechanisms and tools. J Cell Sci 2025; 138:jcs263704. [PMID: 40145202 DOI: 10.1242/jcs.263704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025] Open
Abstract
From border cell migration during Drosophila embryogenesis to solid stresses inside tumors, cells are often compressed during physiological and pathological processes, triggering major cell responses. Cell compression can be observed in vivo but also controlled in vitro through tools such as micro-channels or planar confinement assays. Such tools have recently become commercially available, allowing a broad research community to tackle the role of cell compression in a variety of contexts. This has led to the discovery of conserved compression-triggered migration modes, cell fate determinants and mechanosensitive pathways, among others. In this Review, we will first address the different ways in which cells can be compressed and their biological contexts. Then, we will discuss the distinct mechanosensing and mechanotransducing pathways that cells activate in response to compression. Finally, we will describe the different in vitro systems that have been engineered to compress cells.
Collapse
Affiliation(s)
- Laura M Faure
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Valeria Venturini
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- University of Barcelona (UB), 08036 Barcelona, Spain
| |
Collapse
|
12
|
Saraswathibhatla A, Rabbi MF, Varma S, Srivastava V, Ilina O, Alyafei NHK, Hodgson L, Gartner Z, Friedl P, West R, Kim T, Chaudhuri O. Swirling motion of breast cancer cells radially aligns collagen fibers to enable collective invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.635980. [PMID: 39974994 PMCID: PMC11838510 DOI: 10.1101/2025.01.31.635980] [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
In breast cancer (BC), radial alignment of collagen fibers at the tumor-matrix interface facilitates collective invasion of cancer cells into the surrounding stromal matrix, a critical step toward metastasis. Collagen remodeling is driven by proteases and cellular forces, mediated by matrix mechanical plasticity, or irreversible matrix deformation in response to force. However, the specific mechanisms causing collagen radial alignment remain unclear. Here, we study collective invasion of BC tumor spheroids in collagen-rich matrices. Increasing plasticity to BC-relevant ranges facilitates invasion, with increasing stiffness potentiating a transition from single cell to collective invasion. At enhanced plasticity, cells radially align collagen at the tumor-matrix interface prior to invasion. Surprisingly, cells migrate tangentially to the tumor-matrix interface in a swirling-like motion, perpendicular to the direction of alignment. Mechanistically, swirling generates local shear stresses, leading to distally propagating contractile radial stresses due to negative normal stress, an underappreciated property of collagen-rich matrices. These contractile stresses align collagen fibers radially, facilitating collective invasion. The basement membrane (BM), which separates epithelia from stroma in healthy tissues, acts as a mechanical insulator by preventing swirling cells from aligning collagen. Thus, after breaching the BM, swirling of BC cells at the tumor-stroma interface radially aligns collagen to facilitate invasion.
Collapse
|
13
|
Sinha S, Li X, Malmi-Kakkada AN, Thirumalai D. Mechanical feedback links cell division and dynamics in growing cell collectives. SOFT MATTER 2025; 21:1170-1179. [PMID: 39820196 DOI: 10.1039/d4sm01230e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Local stresses in a tissue, a collective property, regulate cell division and apoptosis. In turn, cell growth and division induce active stresses in the tissue. As a consequence, there is a feedback between cell growth and local stresses. However, how the cell dynamics depend on local stress-dependent cell division and the feedback strength is not understood. Here, we probe the consequences of stress-mediated growth and cell division on cell dynamics using agent-based simulations of a two-dimensional growing tissue. We discover a rich dynamical behavior of individual cells, ranging from jamming (mean square displacement, Δ(t) ∼ tα with α less than unity), to hyperdiffusion (α > 2) depending on cell division rate and the strength of the mechanical feedback. Strikingly, Δ(t) is determined by the tissue growth law, which quantifies cell proliferation, measuring the number of cells N(t) as a function of time. The growth law (N(t) ∼ tλ at long times) is regulated by the critical pressure that controls the strength of the mechanical feedback and the ratio between cell division-apoptosis rates. We show that λ ∼ α, which implies that higher growth rate leads to a greater degree of cell migration. The variations in cell motility are linked to the emergence of highly persistent forces extending over several cell cycle times. Our predictions are testable using cell-tracking imaging techniques.
Collapse
Affiliation(s)
- Sumit Sinha
- Department of Physics, University of Texas at Austin, Austin, TX 78712, USA.
| | - Xin Li
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712, USA
| | | | - D Thirumalai
- Department of Physics, University of Texas at Austin, Austin, TX 78712, USA.
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
14
|
Ravichandran A, Mahajan V, van de Kemp T, Taubenberger A, Bray LJ. Phenotypic analysis of complex bioengineered 3D models. Trends Cell Biol 2025:S0962-8924(24)00257-5. [PMID: 39794253 DOI: 10.1016/j.tcb.2024.12.004] [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: 08/07/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025]
Abstract
With advances in underlying technologies such as complex multicellular systems, synthetic materials, and bioengineering techniques, we can now generate in vitro miniaturized human tissues that recapitulate the organotypic features of normal or diseased tissues. Importantly, these 3D culture models have increasingly provided experimental access to diverse and complex tissues architectures and their morphogenic assembly in vitro. This review presents an analytical toolbox for biological researchers using 3D modeling technologies through which they can find a collation of currently available methods to phenotypically assess their 3D models in their normal state as well as their response to therapeutic or pathological agents.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Vaibhav Mahajan
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Tom van de Kemp
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Anna Taubenberger
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Laura J Bray
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.
| |
Collapse
|
15
|
Zhang Y, Qi F, Chen P, Liu BF, Li Y. Spatially defined microenvironment for engineering organoids. BIOPHYSICS REVIEWS 2024; 5:041302. [PMID: 39679203 PMCID: PMC11646138 DOI: 10.1063/5.0198848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/01/2024] [Indexed: 12/17/2024]
Abstract
In the intricately defined spatial microenvironment, a single fertilized egg remarkably develops into a conserved and well-organized multicellular organism. This observation leads us to hypothesize that stem cells or other seed cell types have the potential to construct fully structured and functional tissues or organs, provided the spatial cues are appropriately configured. Current organoid technology, however, largely depends on spontaneous growth and self-organization, lacking systematic guided intervention. As a result, the structures replicated in vitro often emerge in a disordered and sparse manner during growth phases. Although existing organoids have made significant contributions in many aspects, such as advancing our understanding of development and pathogenesis, aiding personalized drug selection, as well as expediting drug development, their potential in creating large-scale implantable tissue or organ constructs, and constructing multicomponent microphysiological systems, together with functioning at metabolic levels remains underutilized. Recent discoveries have demonstrated that the spatial definition of growth factors not only induces directional growth and migration of organoids but also leads to the formation of assembloids with multiple regional identities. This opens new avenues for the innovative engineering of higher-order organoids. Concurrently, the spatial organization of other microenvironmental cues, such as physical stresses, mechanical loads, and material composition, has been minimally explored. This review delves into the burgeoning field of organoid engineering with a focus on potential spatial microenvironmental control. It offers insight into the molecular principles, expected outcomes, and potential applications, envisioning a future perspective in this domain.
Collapse
Affiliation(s)
- Yilan Zhang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fukang Qi
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
16
|
Da Silva André G, Labouesse C. Mechanobiology of 3D cell confinement and extracellular crowding. Biophys Rev 2024; 16:833-849. [PMID: 39830117 PMCID: PMC11735831 DOI: 10.1007/s12551-024-01244-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/30/2024] [Indexed: 01/22/2025] Open
Abstract
Cells and tissues are often under some level of confinement, imposed by the microenvironment and neighboring cells, meaning that there are limitations to cell size, volume changes, and fluid exchanges. 3D cell culture, increasingly used for both single cells and organoids, inherently impose levels of confinement absent in 2D systems. It is thus key to understand how different levels of confinement influences cell survival, cell function, and cell fate. It is well known that the mechanical properties of the microenvironment, such as stiffness and stress relaxation, are important in activating mechanosensitive pathways, and these are responsive to confinement conditions. In this review, we look at how low, intermediate, and high levels of confinement modulate the activation of known mechanobiology pathways, in single cells, organoids, and tumor spheroids, with a specific focus on 3D confinement in microwells, elastic, or viscoelastic scaffolds. In addition, a confining microenvironment can drastically limit cellular communication in both healthy and diseased tissues, due to extracellular crowding. We discuss potential implications of extracellular crowding on molecular transport, extracellular matrix deposition, and fluid transport. Understanding how cells sense and respond to various levels of confinement should inform the design of 3D engineered matrices that recapitulate the physical properties of tissues.
Collapse
Affiliation(s)
- Gabriela Da Silva André
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Céline Labouesse
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
17
|
Shan H, Chen M, Zhao S, Wei X, Zheng M, Li Y, Lin Q, Jiang Z, Chen Z, Fei C, Li Z, Chen Z, Chen X. Acoustic virtual 3D scaffold for direct-interacting tumor organoid-immune cell coculture systems. SCIENCE ADVANCES 2024; 10:eadr4831. [PMID: 39576870 PMCID: PMC11584020 DOI: 10.1126/sciadv.adr4831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024]
Abstract
Three-dimensional (3D) cell culture has revolutionized life sciences, particularly in organoid technologies. Traditional bioscaffold materials, however, complicate the detachment of tumor organoids and hamper the routine use of organoid-immune cell cocultures. Here, we show an acoustic virtual 3D scaffold (AV-Scaf) method to achieve 3D tumor organoid culture, enabling a direct-interacting tumor organoid-immune cell coculture system. The self-organization process of tumor cells is facilitated by a vortex acoustic field, which enables the cell bioassembly and ion channel activation. This approach can significantly enhance the influx of calcium ions, thereby accelerating intercellular interactions of cellular assemblies. We established scaffold-free melanoma and breast cancer organoids using AV-Scaf and cocultured melanoma organoids with T cells. We found that our coculture system resulted in a high activation state of T cells, characterized by notable up-regulation of granzyme B (2.82 to 17.5%) and interferon-γ (1.36 to 16%). AV-Scaf offers an efficient method for tumor organoid-immune cell studies, advancing cancer research and immunotherapy development.
Collapse
Affiliation(s)
- Han Shan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Maike Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| | - Xiongwei Wei
- The School of Integrated Circuit, Xidian University, Xi'an 710071, China
| | - Mingde Zheng
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Yixin Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| | - Qibo Lin
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Zixi Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| | - Ziyan Chen
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Chunlong Fei
- The School of Integrated Circuit, Xidian University, Xi'an 710071, China
| | - Zhaoxi Li
- The School of Integrated Circuit, Xidian University, Xi'an 710071, China
| | - Zeyu Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| |
Collapse
|
18
|
Yang H, Meyer F, Huang S, Yang L, Lungu C, Olayioye MA, Buehler MJ, Guo M. Learning collective cell migratory dynamics from a static snapshot with graph neural networks. ARXIV 2024:arXiv:2401.12196v3. [PMID: 38344226 PMCID: PMC10854275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Multicellular self-assembly into functional structures is a dynamic process that is critical in the development and diseases, including embryo development, organ formation, tumor invasion, and others. Being able to infer collective cell migratory dynamics from their static configuration is valuable for both understanding and predicting these complex processes. However, the identification of structural features that can indicate multicellular motion has been difficult, and existing metrics largely rely on physical instincts. Here we show that using a graph neural network (GNN), the motion of multicellular collectives can be inferred from a static snapshot of cell positions, in both experimental and synthetic datasets.
Collapse
Affiliation(s)
- Haiqian Yang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Florian Meyer
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Shaoxun Huang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Liu Yang
- Department of Computer Sciences, University of Wisconsin - Madison, Madison, WI 53706, USA
| | - Cristiana Lungu
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Monilola A. Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Markus J. Buehler
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Laboratory for Atomistic and Molecular Mechanics (LAMM), Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
- Center for Computational Science and Engineering, Schwarzman College of Computing, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| |
Collapse
|
19
|
Ye J, Li F, Hua T, Ma K, Wang J, Zhao Z, Yang Z, Luo C, Jia R, Li Y, Hao M, Wu J, Lu M, Yuan Z, Zhang J, Chen J. Liver mechanosignaling as a natural anti-hepatitis B virus mechanism. Nat Commun 2024; 15:8375. [PMID: 39333106 PMCID: PMC11437074 DOI: 10.1038/s41467-024-52718-3] [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: 06/19/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
The mechanisms underlying the natural control of hepatitis B virus (HBV) infection have long been an intriguing question. Given the wide physiological range of liver stiffness and the growing attention to the role of mechanical microenvironment in homeostasis and diseases, we investigated how physical matrix cues impact HBV replication. High matrix stiffness significantly inhibited HBV replication and activated YAP in primary hepatocyte culture system, a key molecule in mechanosignaling. YAP activation notably suppressed HBV transcription and antigen expression. Several YAP-induced genes exhibited strong anti-HBV effects. Single-cell analysis of liver tissue from male individuals with active HBV replication revealed a strong significant negative correlation between YAP signature activation and HBV transcript levels. Intraperitoneal administration of YAP small molecule agonist potently controls HBV in male mouse models. These findings unveil a mechanism that involves the mechanical environment of hepatocytes and YAP to clear hepatotropic viral infection in the liver, providing new perspectives for HBV cure studies and antiviral development.
Collapse
Affiliation(s)
- Jianyu Ye
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Fahong Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Ting Hua
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Kewei Ma
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Jinyu Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Zixin Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Zhongning Yang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Chen Luo
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Ruohan Jia
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Menghan Hao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China
| | - Jian Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China.
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China.
| | - Jiming Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China.
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jieliang Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Huashan Hospital, Shanghai Medical College Fudan University, Shanghai, China.
- Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Diseases and Biosecurity, Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Fudan University, Shanghai, China.
| |
Collapse
|
20
|
Habli Z, Zantout A, Al-Haj N, Saab R, El-Sabban M, Khraiche ML. Single-Cell Fluidic Force Spectroscopy Reveals Dynamic Mechanical Fingerprints of Malignancy in Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50147-50159. [PMID: 39105773 PMCID: PMC11440459 DOI: 10.1021/acsami.4c06335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
The interplay between cancer cell physical characteristics and metastatic potential highlights the significance of cancer cell mechanobiology. Using fluidic-based single-cell force spectroscopy (SCFS), quartz crystal microbalance with dissipation (QCM-D), and a model of cells with a spectrum of metastatic potential, we track the progression of biomechanics across the metastatic states by measuring cell-substrate and cell-to-cell adhesion forces, cell spring constant, cell height, and cell viscoelasticity. Compared to highly metastatic cells, cells in the lower spectrum of metastatic ability are found to be systematically stiffer, less viscoelastic, and larger. These mechanical transformations in cells within a cluster correlate with cells' metastatic potential but are significantly absent in single cells. Additionally, the response to chemotherapy is found to be highly dependent on cell viscoelastic properties in terms of both response time and magnitude. Shifts in cell softness and elasticity might serve as mechanoadaptive mechanisms during cancer cell metastasis, contributing to our understanding of metastasis and the effectiveness of potential therapeutic interventions.
Collapse
Affiliation(s)
- Zeina Habli
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Ahmad Zantout
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Nadine Al-Haj
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Raya Saab
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Massoud L Khraiche
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| |
Collapse
|
21
|
Mowla A, Hepburn MS, Li J, Vahala D, Amos SE, Hirvonen LM, Sanderson RW, Wijesinghe P, Maher S, Choi YS, Kennedy BF. Multimodal mechano-microscopy reveals mechanical phenotypes of breast cancer spheroids in three dimensions. APL Bioeng 2024; 8:036113. [PMID: 39257700 PMCID: PMC11387014 DOI: 10.1063/5.0213077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/01/2024] [Indexed: 09/12/2024] Open
Abstract
Cancer cell invasion relies on an equilibrium between cell deformability and the biophysical constraints imposed by the extracellular matrix (ECM). However, there is little consensus on the nature of the local biomechanical alterations in cancer cell dissemination in the context of three-dimensional (3D) tumor microenvironments (TMEs). While the shortcomings of two-dimensional (2D) models in replicating in situ cell behavior are well known, 3D TME models remain underutilized because contemporary mechanical quantification tools are limited to surface measurements. Here, we overcome this major challenge by quantifying local mechanics of cancer cell spheroids in 3D TMEs. We achieve this using multimodal mechano-microscopy, integrating optical coherence microscopy-based elasticity imaging with confocal fluorescence microscopy. We observe that non-metastatic cancer spheroids show no invasion while showing increased peripheral cell elasticity in both stiff and soft environments. Metastatic cancer spheroids, however, show ECM-mediated softening in a stiff microenvironment and, in a soft environment, initiate cell invasion with peripheral softening associated with early metastatic dissemination. This exemplar of live-cell 3D mechanotyping supports that invasion increases cell deformability in a 3D context, illustrating the power of multimodal mechano-microscopy for quantitative mechanobiology in situ.
Collapse
Affiliation(s)
| | | | | | - Danielle Vahala
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Sebastian E Amos
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Liisa M Hirvonen
- Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, Perth, WA 6009, Australia
| | | | - Philip Wijesinghe
- Centre of Biophotonics, SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Samuel Maher
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | | |
Collapse
|
22
|
Burgstaller A, Piernitzki N, Küchler N, Koch M, Kister T, Eichler H, Kraus T, Schwarz EC, Dustin ML, Lautenschläger F, Staufer O. Soft Synthetic Cells with Mobile Membrane Ligands for Ex Vivo Expansion of Therapy-Relevant T Cell Phenotypes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401844. [PMID: 38751204 DOI: 10.1002/smll.202401844] [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: 03/07/2024] [Revised: 04/29/2024] [Indexed: 10/01/2024]
Abstract
The expansion of T cells ex vivo is crucial for effective immunotherapy but currently limited by a lack of expansion approaches that closely mimic in vivo T cell activation. Taking inspiration from bottom-up synthetic biology, a new synthetic cell technology is introduced based on dispersed liquid-liquid phase-separated droplet-supported lipid bilayers (dsLBs) with tunable biochemical and biophysical characteristics, as artificial antigen presenting cells (aAPCs) for ex vivo T cell expansion. These findings obtained with the dsLB technology reveal three key insights: first, introducing laterally mobile stimulatory ligands on soft aAPCs promotes expansion of IL-4/IL-10 secreting regulatory CD8+ T cells, with a PD-1 negative phenotype, less prone to immune suppression. Second, it is demonstrated that lateral ligand mobility can mask differential T cell activation observed on substrates of varying stiffness. Third, dsLBs are applied to reveal a mechanosensitive component in bispecific Her2/CD3 T cell engager-mediated T cell activation. Based on these three insights, lateral ligand mobility, alongside receptor- and mechanosignaling, is proposed to be considered as a third crucial dimension for the design of ex vivo T cell expansion technologies.
Collapse
Affiliation(s)
- Anna Burgstaller
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Campus E8 1, 66123, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Campus Saarland, 66123, Saarbrücken, Germany
| | - Nils Piernitzki
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Campus E8 1, 66123, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Campus Saarland, 66123, Saarbrücken, Germany
| | - Nadja Küchler
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Building 48, 66421, Homburg, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Thomas Kister
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Hermann Eichler
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University and Saarland University Medical Center, Homburg, Germany
| | - Tobias Kraus
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Saarland University, Colloid and Interface Chemistry, 66123, Saarbrücken, Germany
| | - Eva C Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Building 48, 66421, Homburg, Germany
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology and, Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Franziska Lautenschläger
- Center for Biophysics, Saarland University, Campus Saarland, 66123, Saarbrücken, Germany
- Experimental Physics, Faculty of Natural Science and Technology, Saarland University, Campus Saarbrücken, 66123, Saarbrücken, Germany
| | - Oskar Staufer
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Campus E8 1, 66123, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Campus Saarland, 66123, Saarbrücken, Germany
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology and, Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Max Planck Bristol Centre for Minimal Biology, Cantock's Close, Bristol, BS8 1TS, UK
| |
Collapse
|
23
|
Zhang W, Hu Y, Feng P, Li Z, Zhang H, Zhang B, Xu D, Qi J, Wang H, Xu L, Li Z, Xia M, Li J, Chai R, Tian L. Structural Color Colloidal Photonic Crystals for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403173. [PMID: 39083316 PMCID: PMC11423208 DOI: 10.1002/advs.202403173] [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: 03/26/2024] [Revised: 07/10/2024] [Indexed: 09/26/2024]
Abstract
Photonic crystals are a new class of optical microstructure materials characterized by a dielectric constant that varies periodically with space and features a photonic bandgap. Inspired by natural photonic crystals such as butterfly scales, a series of artificial photonic crystals are developed for use in integrated photonic platforms, biosensing, communication, and other fields. Among them, colloidal photonic crystals (CPCs) have gained widespread attention due to their excellent optical properties and advantages, such as ease of preparation and functionalization. This work reviews the classification and self-assembly principles of CPCs, details some of the latest biomedical applications of large-area, high-quality CPCs prepared using advanced self-assembly methods, summarizes the existing challenges in CPC construction and application, and anticipates future development directions and optimization strategy. With further advancements, CPCs are expected to play a more critical role in biosensors, drug delivery, cell research, and other fields, bringing significant benefits to biomedical research and clinical practice.
Collapse
Affiliation(s)
- Wenhui Zhang
- School of Design and Arts, Beijing Institute of Technology, Beijing, 100081, China
| | - Yangnan Hu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Pan Feng
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Zhe Li
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Hui Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Bin Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Dongyu Xu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jieyu Qi
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Huan Wang
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ming Xia
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jilai Li
- Department of Neurology, Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, 100049, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China
| | - Lei Tian
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| |
Collapse
|
24
|
Boot RC, van der Net A, Gogou C, Mehta P, Meijer DH, Koenderink GH, Boukany PE. Cell spheroid viscoelasticity is deformation-dependent. Sci Rep 2024; 14:20013. [PMID: 39198595 PMCID: PMC11358509 DOI: 10.1038/s41598-024-70759-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
Tissue surface tension influences cell sorting and tissue fusion. Earlier mechanical studies suggest that multicellular spheroids actively reinforce their surface tension with applied force. Here we study this open question through high-throughput microfluidic micropipette aspiration measurements on cell spheroids to identify the role of force duration and spheroid deformability. In particular, we aspirate spheroid protrusions of mice fibroblast NIH3T3 and human embryonic HEK293T homogeneous cell spheroids into micron-sized capillaries for different pressures and monitor their viscoelastic creep behavior. We find that larger spheroid deformations lead to faster cellular retraction once the pressure is released, regardless of the applied force. Additionally, less deformable NIH3T3 cell spheroids with an increased expression level of alpha-smooth muscle actin, a cytoskeletal protein upregulating cellular contractility, also demonstrate slower cellular retraction after pressure release for smaller spheroid deformations. Moreover, HEK293T cell spheroids only display cellular retraction at larger pressures with larger spheroid deformations, despite an additional increase in viscosity at these larger pressures. These new insights demonstrate that spheroid viscoelasticity is deformation-dependent and challenge whether surface tension truly reinforces at larger aspiration pressures.
Collapse
Affiliation(s)
- Ruben C Boot
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629, HZ, The Netherlands
| | - Anouk van der Net
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, 2629, HZ, The Netherlands
| | - Christos Gogou
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, 2629, HZ, The Netherlands
| | - Pranav Mehta
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629, HZ, The Netherlands
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, 2333, ZA, The Netherlands
| | - Dimphna H Meijer
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, 2629, HZ, The Netherlands
| | - Gijsje H Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, 2629, HZ, The Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629, HZ, The Netherlands.
| |
Collapse
|
25
|
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
|
26
|
Liu X, Guo R, Li D, Wang Y, Ning J, Yang S, Yang J. Homotypic cell-in-cell structure as a novel prognostic predictor in non-small cell lung cancer and frequently localized at the invasive front. Sci Rep 2024; 14:18952. [PMID: 39147858 PMCID: PMC11327305 DOI: 10.1038/s41598-024-69833-2] [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: 06/11/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Homotypic cell-in-cell structures (hoCICs) are associated with tumor proliferation, invasion, and metastasis and is considered a promising prognostic marker in various cancers. However, the role of hoCICs in non-small cell lung cancer (NSCLC) remains unclear. Tumor tissue sections were obtained from 411 NSCLC patients. We analyzed the relationship between clinicopathological variables and the number of hoCICs. LASSO and multivariate Cox regression analysis were employed to identify prognostic factors for NSCLC. The impact of hoCICs on overall survival (OS) and disease-free survival (DFS) was assessed using the Kaplan-Meier curves and log-rank test. Prognostic models for OS and DFS were developed and validated using the C-index, time-dependent area under the curve (AUC), net reclassification improvement (NRI), integrated discrimination improvement (IDI), calibration curves and decision curve analysis (DCA). Among the cohort, 56% of patients had hoCICs while 44% did not. Notably, hoCICs were primarily found at the tumor invasion front. Male gender, smoking, squamous cell carcinoma, low differentiation, tumor size ≥ 3 cm, advanced TNM stage, lymph node metastasis, pleural invasion, vascular invasion, necrosis, P53 mutation, and high expression of Ki-67 were identified as relative risk factors for hoCICs. Furthermore, hoCICs was found to be a significant prognostic factor for both OS and DFS, with higher frequencies of hoCICs correlating with poorer outcomes. We constructed nomograms for predicting 1-, 3-, and 5-year OS and DFS based on hoCICs, and the calibration curves showed good agreement between the predicted and actual outcomes. The results of the C-index, time-dependent AUC, NRI, IDI, and DCA analyses demonstrated that incorporating hoCICs into the prognostic model significantly enhanced its predictive power and clinical applicability. HoCICs indicated independent perdictive value for OS and DFS in patients with NSCLC. Furthermore, the frequent localization of hoCICs at the tumor invasion front suggested a strong association between hoCICs and tumor invasion as well as metastasis.
Collapse
Affiliation(s)
- Xiaona Liu
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, ShaanXi, China
| | - Rui Guo
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, ShaanXi, China
| | - Dongxuan Li
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, ShaanXi, China
| | - Ya'nan Wang
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, ShaanXi, China
| | - Jingya Ning
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, ShaanXi, China
| | - Shuanying Yang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, ShaanXi, China.
| | - Jun Yang
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, ShaanXi, China.
| |
Collapse
|
27
|
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
|
28
|
Zhang J, Mosier JA, Wu Y, Waddle L, Taufalele PV, Wang W, Sun H, Reinhart‐King CA. Cellular Energy Cycle Mediates an Advection-Like Forward Cell Flow to Support Collective Invasion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400719. [PMID: 39189477 PMCID: PMC11348062 DOI: 10.1002/advs.202400719] [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: 01/20/2024] [Revised: 05/25/2024] [Indexed: 08/28/2024]
Abstract
Collective cell migration is a model for nonequilibrium biological dynamics, which is important for morphogenesis, pattern formation, and cancer metastasis. The current understanding of cellular collective dynamics is based primarily on cells moving within a 2D epithelial monolayer. However, solid tumors often invade surrounding tissues in the form of a stream-like 3D structure, and how biophysical cues are integrated at the cellular level to give rise to this collective streaming remains unclear. Here, it is shown that cell cycle-mediated bioenergetics drive a forward advective flow of cells and energy to the front to support 3D collective invasion. The cell division cycle mediates a corresponding energy cycle such that cellular adenosine triphosphate (ATP) energy peaks just before division. A reaction-advection-diffusion (RAD) type model coupled with experimental measurements further indicates that most cells enter an active division cycle at rear positions during 3D streaming. Once the cells progress to a later stage toward division, the high intracellular energy allows them to preferentially stream toward the tip and become leader cells. This energy-driven cellular flow may be a fundamental characteristic of 3D collective dynamics based on thermodynamic principles important for not only cancer invasion but also tissue morphogenesis.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Biomedical EngineeringVanderbilt University2301 Vanderbilt PlaceNashvilleTN37235USA
- Department of Biomedical EngineeringUniversity of Arkansas790 W. Dickson StFayettevilleAR72701USA
| | - Jenna A. Mosier
- Department of Biomedical EngineeringVanderbilt University2301 Vanderbilt PlaceNashvilleTN37235USA
| | - Yusheng Wu
- Department of Biomedical EngineeringVanderbilt University2301 Vanderbilt PlaceNashvilleTN37235USA
| | - Logan Waddle
- Department of Biomedical EngineeringUniversity of Arkansas790 W. Dickson StFayettevilleAR72701USA
| | - Paul V. Taufalele
- Department of Biomedical EngineeringVanderbilt University2301 Vanderbilt PlaceNashvilleTN37235USA
| | - Wenjun Wang
- Department of Biomedical EngineeringVanderbilt University2301 Vanderbilt PlaceNashvilleTN37235USA
| | - Heng Sun
- Department of Biomedical EngineeringVanderbilt University2301 Vanderbilt PlaceNashvilleTN37235USA
| | - Cynthia A. Reinhart‐King
- Department of Biomedical EngineeringVanderbilt University2301 Vanderbilt PlaceNashvilleTN37235USA
| |
Collapse
|
29
|
Meng J, Xu X, Jiang C, Xia P, Xu P, Tian L, Xu Y, Li D, Tan Y, Ji B. Tensile force field plays a crucial role in local invasion of tumor cells through a mechano-chemical coupling mechanism. SOFT MATTER 2024. [PMID: 39027971 DOI: 10.1039/d4sm00335g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cancer metastasis starts from early local invasion, during which tumor cells detach from the primary tumor, penetrate the extracellular matrix (ECM), and then invade neighboring tissues. However, the cellular mechanics in the detaching and penetrating processes have not been fully understood, and the underlying mechanisms that influence cell polarization and migration in the 3D matrix during tumor invasion remain largely unknown. In this study, we employed a dual tumor-spheroid model to investigate the cellular mechanisms of the tumor invasion. Our results revealed that the tensional force field developed by the active contraction of cells and tissues played a pivotal role in tumor invasion, acting as the driving force for remodeling the collagen fibers during the invasion process. The remodeled collagen fibers promoted cell polarization and migration because of the stiffening of the fiber matrix. The aligned fibers facilitated tumor cell invasion and directed migration from one spheroid to the other. Inhibiting/shielding the cellular contractility abolished matrix remodeling and re-alignment and significantly decreased tumor cell invasion. By developing a coarse-grained cell model that considers the mutual interaction between cells and fibers, we predicted the tensional force field in the fiber network and the associated cell polarization and cell-matrix interaction during cell invasion, which revealed a mechano-chemical coupling mechanism at the cellular level of the tumor invasion process. Our study highlights the roles of cellular mechanics at the early stage of tumor metastasis and may provide new therapeutic strategies for cancer therapy.
Collapse
Affiliation(s)
- Jianfeng Meng
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xiangyu Xu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chaohui Jiang
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Peng Xia
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Science Institute, Zhejiang University, Hangzhou 310058, China
| | - Pengfei Xu
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Liangfei Tian
- MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yingke Xu
- MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Dechang Li
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
| | - Youhua Tan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Baohua Ji
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
30
|
Arora P, Sadhukhan S, Nandi SK, Bi D, Sood AK, Ganapathy R. A shape-driven reentrant jamming transition in confluent monolayers of synthetic cell-mimics. Nat Commun 2024; 15:5645. [PMID: 38969629 PMCID: PMC11226658 DOI: 10.1038/s41467-024-49044-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/21/2024] [Indexed: 07/07/2024] Open
Abstract
Many critical biological processes, like wound healing, require densely packed cell monolayers/tissues to transition from a jammed solid-like to a fluid-like state. Although numerical studies anticipate changes in the cell shape alone can lead to unjamming, experimental support for this prediction is not definitive because, in living systems, fluidization due to density changes cannot be ruled out. Additionally, a cell's ability to modulate its motility only compounds difficulties since even in assemblies of rigid active particles, changing the nature of self-propulsion has non-trivial effects on the dynamics. Here, we design and assemble a monolayer of synthetic cell-mimics and examine their collective behaviour. By systematically increasing the persistence time of self-propulsion, we discovered a cell shape-driven, density-independent, re-entrant jamming transition. Notably, we observed cell shape and shape variability were mutually constrained in the confluent limit and followed the same universal scaling as that observed in confluent epithelia. Dynamical heterogeneities, however, did not conform to this scaling, with the fast cells showing suppressed shape variability, which our simulations revealed is due to a transient confinement effect of these cells by their slower neighbors. Our experiments unequivocally establish a morphodynamic link, demonstrating that geometric constraints alone can dictate epithelial jamming/unjamming.
Collapse
Affiliation(s)
- Pragya Arora
- Chemistry and Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, India.
| | - Souvik Sadhukhan
- Tata Institute of Fundamental Research, Hyderabad, 500046, India
| | | | - Dapeng Bi
- Department of Physics, Northeastern University, Boston, MA, 02115, USA
| | - A K Sood
- Department of Physics, Indian Institute of Science, Bangalore, 560012, India
- International Centre for Materials Science, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, India
| | - Rajesh Ganapathy
- International Centre for Materials Science, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, India.
- School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, India.
| |
Collapse
|
31
|
Hilai K, Grubich D, Akrawi M, Zhu H, Zaghloul R, Shi C, Do M, Zhu D, Zhang J. Mechanical evolution of metastatic cancer cells in three-dimensional microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601015. [PMID: 39005477 PMCID: PMC11244934 DOI: 10.1101/2024.06.27.601015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Cellular biomechanics plays critical roles in cancer metastasis and tumor progression. Existing studies on cancer cell biomechanics are mostly conducted in flat 2D conditions, where cells' behavior can differ considerably from those in 3D physiological environments. Despite great advances in developing 3D in vitro models, probing cellular elasticity in 3D conditions remains a major challenge for existing technologies. In this work, we utilize optical Brillouin microscopy to longitudinally acquire mechanical images of growing cancerous spheroids over the period of eight days. The dense mechanical mapping from Brillouin microscopy enables us to extract spatially resolved and temporally evolving mechanical features that were previously inaccessible. Using an established machine learning algorithm, we demonstrate that incorporating these extracted mechanical features significantly improves the classification accuracy of cancer cells, from 74% to 95%. Building on this finding, we have developed a deep learning pipeline capable of accurately differentiating cancerous spheroids from normal ones solely using Brillouin images, suggesting the mechanical features of cancer cells could potentially serve as a new biomarker in cancer classification and detection.
Collapse
Affiliation(s)
- Karlin Hilai
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| | - Daniil Grubich
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| | - Marcus Akrawi
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| | - Hui Zhu
- Department of Computer Science, Wayne State University, Detroit, MI, 48202, USA
| | - Razanne Zaghloul
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| | - Chenjun Shi
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| | - Man Do
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| | - Dongxiao Zhu
- Department of Computer Science, Wayne State University, Detroit, MI, 48202, USA
| | - Jitao Zhang
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
32
|
Chang Z, Li LY, Shi ZJ, Liu W, Xu GK. Beyond stiffness: Multiscale viscoelastic features as biomechanical markers for assessing cell types and states. Biophys J 2024; 123:1869-1881. [PMID: 38835167 PMCID: PMC11267428 DOI: 10.1016/j.bpj.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/14/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024] Open
Abstract
Cell mechanics are pivotal in regulating cellular activities, diseases progression, and cancer development. However, the understanding of how cellular viscoelastic properties vary in physiological and pathological stimuli remains scarce. Here, we develop a hybrid self-similar hierarchical theory-microrheology approach to accurately and efficiently characterize cellular viscoelasticity. Focusing on two key cell types associated with livers fibrosis-the capillarized liver sinusoidal endothelial cells and activated hepatic stellate cells-we uncover a universal two-stage power-law rheology characterized by two distinct exponents, αshort and αlong. The mechanical profiles derived from both exponents exhibit significant potential for discriminating among diverse cells. This finding suggests a potential common dynamic creep characteristic across biological systems, extending our earlier observations in soft tissues. Using a tailored hierarchical model for cellular mechanical structures, we discern significant variations in the viscoelastic properties and their distribution profiles across different cell types and states from the cytoplasm (elastic stiffness E1 and viscosity η), to a single cytoskeleton fiber (elastic stiffness E2), and then to the cell level (transverse expansion stiffness E3). Importantly, we construct a logistic-regression-based machine-learning model using the dynamic parameters that outperforms conventional cell-stiffness-based classifiers in assessing cell states, achieving an area under the curve of 97% vs. 78%. Our findings not only advance a robust framework for monitoring intricate cell dynamics but also highlight the crucial role of cellular viscoelasticity in discerning cell states across a spectrum of liver diseases and prognosis, offering new avenues for developing diagnostic and therapeutic strategies based on cellular viscoelasticity.
Collapse
Affiliation(s)
- Zhuo Chang
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Li-Ya Li
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhi-Jun Shi
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Wenjia Liu
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Guang-Kui Xu
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
33
|
Xie J, Huck WTS, Bao M. Unveiling the Intricate Connection: Cell Volume as a Key Regulator of Mechanotransduction. Annu Rev Biophys 2024; 53:299-317. [PMID: 38424091 DOI: 10.1146/annurev-biophys-030822-035656] [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: 03/02/2024]
Abstract
The volumes of living cells undergo dynamic changes to maintain the cells' structural and functional integrity in many physiological processes. Minor fluctuations in cell volume can serve as intrinsic signals that play a crucial role in cell fate determination during mechanotransduction. In this review, we discuss the variability of cell volume and its role in vivo, along with an overview of the mechanisms governing cell volume regulation. Additionally, we provide insights into the current approaches used to control cell volume in vitro. Furthermore, we summarize the biological implications of cell volume regulation and discuss recent advances in understanding the fundamental relationship between cell volume and mechanotransduction. Finally, we delve into the potential underlying mechanisms, including intracellular macromolecular crowding and cellular mechanics, that govern the global regulation of cell fate in response to changes in cell volume. By exploring the intricate interplay between cell volume and mechanotransduction, we underscore the importance of considering cell volume as a fundamental signaling cue to unravel the basic principles of mechanotransduction. Additionally, we propose future research directions that can extend our current understanding of cell volume in mechanotransduction. Overall, this review highlights the significance of considering cell volume as a fundamental signal in understanding the basic principles in mechanotransduction and points out the possibility of controlling cell volume to control cell fate, mitigate disease-related damage, and facilitate the healing of damaged tissues.
Collapse
Affiliation(s)
- Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands;
| | - Min Bao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China;
| |
Collapse
|
34
|
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
|
35
|
Ti F, Yu C, Li M, Liu S, Lu TJ, Chen X. Cross-scale mechanobiological regulation of cylindrical compressible liquid inclusion via coating. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2024; 36:395101. [PMID: 38906135 DOI: 10.1088/1361-648x/ad5ace] [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: 02/05/2024] [Accepted: 06/21/2024] [Indexed: 06/23/2024]
Abstract
The double-bag theory in modern anatomy suggests that structures with coatings are commonly found in human body at various length scales, such as osteocyte processes covered by pericellular matrix and bones covered by muscle tissue. To understand the mechanical behaviors and physiological responses of such biological structures, we develop an analytical model to quantify surface effects on the deformation of a coated cylindrical compressible liquid inclusion in an elastic matrix subjected to remote loading. Our analytical solution reveals that coating can either amplify or attenuate the volumetric strain of the inclusion, depending on the relative elastic moduli of inclusion, coating, and matrix. For illustration, we utilize this solution to explore amplification/attenuation of volumetric strain in musculoskeletal systems, nerve cells, and vascular tissues. We demonstrate that coating often plays a crucial role in mechanical regulation of the development and repair of human tissues and cells. Our model provides qualitative analysis of cross-scale mechanical response of coated liquid inclusions, helpful for constructing mechanical microenvironment of cells.
Collapse
Affiliation(s)
- Fei Ti
- State Key Laboratory of Mechanics and Control for Aerospace Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, People's Republic of China
- MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, People's Republic of China
| | - Chenlei Yu
- State Key Laboratory for Strength and Vibration of Mechanical Structures, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Moxiao Li
- State Key Laboratory of Mechanics and Control for Aerospace Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, People's Republic of China
- MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, People's Republic of China
| | - Shaobao Liu
- State Key Laboratory of Mechanics and Control for Aerospace Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, People's Republic of China
- MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, People's Republic of China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control for Aerospace Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, People's Republic of China
- MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, People's Republic of China
| | - Xin Chen
- Xi'an Modern Chemistry Research Institute, Xi'an 710065, People's Republic of China
| |
Collapse
|
36
|
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
|
37
|
Prasanna CVS, Jolly MK, Bhat R. Spatial heterogeneity in tumor adhesion qualifies collective cell invasion. Biophys J 2024; 123:1635-1647. [PMID: 38725244 PMCID: PMC11214055 DOI: 10.1016/j.bpj.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/12/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024] Open
Abstract
Collective cell invasion (CCI), a canon of most invasive solid tumors, is an emergent property of the interactions between cancer cells and their surrounding extracellular matrix (ECM). However, tumor populations invariably consist of cells expressing variable levels of adhesive proteins that mediate such interactions, disallowing an intuitive understanding of how tumor invasiveness at a multicellular scale is influenced by spatial heterogeneity of cell-cell and cell-ECM adhesion. Here, we have used a Cellular Potts model-based multiscale computational framework that is constructed on the histopathological principles of glandular cancers. In earlier efforts on homogenous cancer cell populations, this framework revealed the relative ranges of interactions, including cell-cell and cell-ECM adhesion that drove collective, dispersed, and mixed multimodal invasion. Here, we constitute a tumor core of two separate cell subsets showing distinct intra- and inter-subset cell-cell or cell-ECM adhesion strengths. These two subsets of cells are arranged to varying extents of spatial intermingling, which we call the heterogeneity index (HI). We observe that low and high inter-subset cell adhesion favors invasion of high-HI and low-HI intermingled populations with distinct intra-subset cell-cell adhesion strengths, respectively. In addition, for explored values of cell-ECM adhesion strengths, populations with high HI values collectively invade better than those with lower HI values. We then asked how spatial invasion is regulated by progressively intermingled cellular subsets that are epithelial, i.e., showed high cell-cell but poor cell-ECM adhesion, and mesenchymal, i.e., with reversed adhesion strengths to the former. Here too, inter-subset adhesion plays an important role in contextualizing the proportionate relationship between HI and invasion. An exception to this relationship is seen for cases of heterogeneous cell-ECM adhesion where sub-maximal HI patterns with higher outer localization of cells with stronger ECM adhesion collectively invade better than their relatively higher-HI counterparts. Our simulations also reveal how adhesion heterogeneity qualifies collective invasion, when either cell-cell or cell-ECM adhesion type is varied but results in an invasive dispersion when both adhesion types are simultaneously altered.
Collapse
Affiliation(s)
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore, India.
| | - Ramray Bhat
- Department of Bioengineering, Indian Institute of Science, Bangalore, India; Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
38
|
Shen Y, Ori-McKenney KM. Microtubule-associated protein MAP7 promotes tubulin posttranslational modifications and cargo transport to enable osmotic adaptation. Dev Cell 2024; 59:1553-1570.e7. [PMID: 38574732 PMCID: PMC11187767 DOI: 10.1016/j.devcel.2024.03.022] [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: 06/23/2023] [Revised: 09/11/2023] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
Cells remodel their cytoskeletal networks to adapt to their environment. Here, we analyze the mechanisms utilized by the cell to tailor its microtubule landscape in response to changes in osmolarity that alter macromolecular crowding. By integrating live-cell imaging, ex vivo enzymatic assays, and in vitro reconstitution, we probe the impact of cytoplasmic density on microtubule-associated proteins (MAPs) and tubulin posttranslational modifications (PTMs). We find that human epithelial cells respond to fluctuations in cytoplasmic density by modulating microtubule acetylation, detyrosination, or MAP7 association without differentially affecting polyglutamylation, tyrosination, or MAP4 association. These MAP-PTM combinations alter intracellular cargo transport, enabling the cell to respond to osmotic challenges. We further dissect the molecular mechanisms governing tubulin PTM specification and find that MAP7 promotes acetylation and inhibits detyrosination. Our data identify MAP7 in modulating the tubulin code, resulting in microtubule cytoskeleton remodeling and alteration of intracellular transport as an integrated mechanism of cellular adaptation.
Collapse
Affiliation(s)
- Yusheng Shen
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Kassandra M Ori-McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
39
|
Chen F, Li X, Guo W, Wang Y, Guo M, Shum HC. Size Scaling of Condensates in Multicomponent Phase Separation. J Am Chem Soc 2024; 146:16000-16009. [PMID: 38809420 DOI: 10.1021/jacs.4c02906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Constant proportionalities between cells and their intracellular organelles have been widely observed in various types of cells, known as intracellular size scaling. However, the mechanism underlying the size scaling and its modulation by environmental factors in multicomponent systems remain poorly understood. Here, we study the size scaling of membrane-less condensates using microdroplet-encapsulated minimalistic condensates formed by droplet microfluidics and mean-field theory. We demonstrate that the size scaling of condensates is an inherent characteristic of liquid-liquid phase separation. This concept is supported by experiments showing the occurrence of size scaling phenomena in various condensate systems and a generic lever rule acquired from mean-field theory. Moreover, it is found that the condensate-to-microdroplet scaling ratio can be affected by the solute and salt concentrations, with good agreement between experiments and predictions by theory. Notably, we identify a noise buffering mechanism whereby condensates composed of large macromolecules effectively maintain constant volumes and counteract concentration fluctuations of small molecules. This mechanism is achieved through the dynamic rearrangement of small molecules in and out of membrane-free interfaces. Our work provides crucial insights into understanding mechanistic principles that govern the size of cells and intracellular organelles as well as associated biological functions.
Collapse
Affiliation(s)
- Feipeng Chen
- Department of Mechanical Engineering, the University of Hong Kong, Pokfulam Road, Hong Kong (SAR) 999077, China
| | - Xiufeng Li
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong (SAR) 999077, China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Wei Guo
- Department of Mechanical Engineering, the University of Hong Kong, Pokfulam Road, Hong Kong (SAR) 999077, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong (SAR) 999077, China
| | - Yuchao Wang
- Department of Mechanical Engineering, the University of Hong Kong, Pokfulam Road, Hong Kong (SAR) 999077, China
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Ho Cheung Shum
- Department of Mechanical Engineering, the University of Hong Kong, Pokfulam Road, Hong Kong (SAR) 999077, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong (SAR) 999077, China
| |
Collapse
|
40
|
Vos BE, Muenker TM, Betz T. Characterizing intracellular mechanics via optical tweezers-based microrheology. Curr Opin Cell Biol 2024; 88:102374. [PMID: 38824902 DOI: 10.1016/j.ceb.2024.102374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/24/2024] [Accepted: 05/03/2024] [Indexed: 06/04/2024]
Abstract
Intracellular organization is a highly regulated homeostatic state maintained to ensure eukaryotic cells' correct and efficient functioning. Thanks to decades of research, vast knowledge of the proteins involved in intracellular transport and organization has been acquired. However, how these influence and potentially regulate the intracellular mechanical properties of the cell is largely unknown. There is a deep knowledge gap between the understanding of cortical mechanics, which is accessible by a series of experimental tools, and the intracellular situation that has been largely neglected due to the difficulty of performing intracellular mechanics measurements. Recently, tools required for such quantitative and localized analysis of intracellular mechanics have been introduced. Here, we review how these approaches and the resulting viscoelastic models lead the way to a full mechanical description of the cytoplasm, which is instrumental for a quantitative characterization of the intracellular life of cells.
Collapse
Affiliation(s)
- Bart E Vos
- Third Institute of Physics, Georg August University, Göttingen, Germany
| | - Till M Muenker
- Third Institute of Physics, Georg August University, Göttingen, Germany
| | - Timo Betz
- Third Institute of Physics, Georg August University, Göttingen, Germany; Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), Georg August University, Göttingen, Germany.
| |
Collapse
|
41
|
Ang I, Yousafzai MS, Yadav V, Mohler K, Rinehart J, Bouklas N, Murrell M. Elastocapillary effects determine early matrix deformation by glioblastoma cell spheroids. APL Bioeng 2024; 8:026109. [PMID: 38706957 PMCID: PMC11069407 DOI: 10.1063/5.0191765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/12/2024] [Indexed: 05/07/2024] Open
Abstract
During cancer pathogenesis, cell-generated mechanical stresses lead to dramatic alterations in the mechanical and organizational properties of the extracellular matrix (ECM). To date, contraction of the ECM is largely attributed to local mechanical stresses generated during cell invasion, but the impact of "elastocapillary" effects from surface tension on the tumor periphery has not been examined. Here, we embed glioblastoma cell spheroids within collagen gels, as a model of tumors within the ECM. We then modulate the surface tension of the spheroids, such that the spheroid contracts or expands. Surprisingly, in both cases, at the far-field, the ECM is contracted toward the spheroids prior to cellular migration from the spheroid into the ECM. Through computational simulation, we demonstrate that contraction of the ECM arises from a balance of spheroid surface tension, cell-ECM interactions, and time-dependent, poroelastic effects of the gel. This leads to the accumulation of ECM near the periphery of the spheroid and the contraction of the ECM without regard to the expansion or contraction of the spheroid. These results highlight the role of tissue-level surface stresses and fluid flow within the ECM in the regulation of cell-ECM interactions.
Collapse
Affiliation(s)
- Ida Ang
- Department of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York 14853, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Chang J, Saraswathibhatla A, Song Z, Varma S, Sanchez C, Alyafei NHK, Indana D, Slyman R, Srivastava S, Liu K, Bassik MC, Marinkovich MP, Hodgson L, Shenoy V, West RB, Chaudhuri O. Cell volume expansion and local contractility drive collective invasion of the basement membrane in breast cancer. NATURE MATERIALS 2024; 23:711-722. [PMID: 37957268 PMCID: PMC11185842 DOI: 10.1038/s41563-023-01716-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 10/05/2023] [Indexed: 11/15/2023]
Abstract
Breast cancer becomes invasive when carcinoma cells invade through the basement membrane (BM)-a nanoporous layer of matrix that physically separates the primary tumour from the stroma. Single cells can invade through nanoporous three-dimensional matrices due to protease-mediated degradation or force-mediated widening of pores via invadopodial protrusions. However, how multiple cells collectively invade through the physiological BM, as they do during breast cancer progression, remains unclear. Here we developed a three-dimensional in vitro model of collective invasion of the BM during breast cancer. We show that cells utilize both proteases and forces-but not invadopodia-to breach the BM. Forces are generated from a combination of global cell volume expansion, which stretches the BM, and local contractile forces that act in the plane of the BM to breach it, allowing invasion. These results uncover a mechanism by which cells collectively interact to overcome a critical barrier to metastasis.
Collapse
Affiliation(s)
- Julie Chang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Zhaoqiang Song
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sushama Varma
- Department of Pathology, Stanford University Medical Center, Palo Alto, CA, USA
| | - Colline Sanchez
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Dhiraj Indana
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Raleigh Slyman
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Sucheta Srivastava
- Department of Pathology, Stanford University Medical Center, Palo Alto, CA, USA
| | - Katherine Liu
- Department of Genetics, Stanford University Medical Center, Palo Alto, CA, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University Medical Center, Palo Alto, CA, USA
| | - M Peter Marinkovich
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Dermatology Service, VA Medical Center, Palo Alto, CA, USA
| | - Louis Hodgson
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Vivek Shenoy
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert B West
- Department of Pathology, Stanford University Medical Center, Palo Alto, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
| |
Collapse
|
43
|
Tang W, Huang J, Pegoraro AF, Zhang JH, Tang Y, Bi D, Kotton DN, Guo M. Nuclear size-regulated emergence of topological packing order on growing human lung alveolospheres. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589951. [PMID: 38659777 PMCID: PMC11042317 DOI: 10.1101/2024.04.17.589951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Within multicellular living systems, cells coordinate their positions with spatiotemporal accuracy to form various structures, setting the clock to control developmental processes and trigger maturation. These arrangements can be regulated by tissue topology, biochemical cues, as well as mechanical perturbations. However, the fundamental rules of how local cell packing order is regulated in forming three-dimensional (3D) multicellular architectures remain unclear. Furthermore, how cellular coordination evolves during developmental processes, and whether this cell patterning behavior is indicative of more complex biological functions, is largely unknown. Here, using human lung alveolospheres as a model system, by combining experiments and numerical simulations, we find that, surprisingly, cell packing behavior on alveolospheres resembles hard-disk packing but with increased randomness; the stiffer cell nuclei act as the hard disks surrounded by deformable cell bodies. Interestingly, we observe the emergence of topological packing order during alveolosphere growth, as a result of increasing nucleus-to-cell size ratio. Specifically, we find more hexagon-concentrated cellular packing with increasing bond orientational order, indicating a topological gas-to-liquid transition. Additionally, by osmotically changing the compactness of cells on alveolospheres, we observe that the variations in packing order align with the change of nucleus-to-cell size ratio. Together, our findings reveal the underlying rules of cell coordination and topological phases during human lung alveolosphere growth. These static packing characteristics are consistent with cell dynamics, together suggesting that better cellular packing stabilizes local cell neighborhoods and may regulate more complex biological functions such as organ development and cellular maturation.
Collapse
|
44
|
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
|
45
|
Jacob DR, Guiblet WM, Mamayusupova H, Shtumpf M, Ciuta I, Ruje L, Gretton S, Bikova M, Correa C, Dellow E, Agrawal SP, Shafiei N, Drobysevskaja A, Armstrong CM, Lam JDG, Vainshtein Y, Clarkson CT, Thorn GJ, Sohn K, Pradeepa MM, Chandrasekharan S, Brooke GN, Klenova E, Zhurkin VB, Teif VB. Nucleosome reorganisation in breast cancer tissues. Clin Epigenetics 2024; 16:50. [PMID: 38561804 PMCID: PMC10986098 DOI: 10.1186/s13148-024-01656-4] [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: 12/29/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Nucleosome repositioning in cancer is believed to cause many changes in genome organisation and gene expression. Understanding these changes is important to elucidate fundamental aspects of cancer. It is also important for medical diagnostics based on cell-free DNA (cfDNA), which originates from genomic DNA regions protected from digestion by nucleosomes. RESULTS We have generated high-resolution nucleosome maps in paired tumour and normal tissues from the same breast cancer patients using MNase-assisted histone H3 ChIP-seq and compared them with the corresponding cfDNA from blood plasma. This analysis has detected single-nucleosome repositioning at key regulatory regions in a patient-specific manner and common cancer-specific patterns across patients. The nucleosomes gained in tumour versus normal tissue were particularly informative of cancer pathways, with ~ 20-fold enrichment at CpG islands, a large fraction of which marked promoters of genes encoding DNA-binding proteins. The tumour tissues were characterised by a 5-10 bp decrease in the average distance between nucleosomes (nucleosome repeat length, NRL), which is qualitatively similar to the differences between pluripotent and differentiated cells. This effect was correlated with gene activity, differential DNA methylation and changes in local occupancy of linker histone variants H1.4 and H1X. CONCLUSIONS Our study offers a novel resource of high-resolution nucleosome maps in breast cancer patients and reports for the first time the effect of systematic decrease of NRL in paired tumour versus normal breast tissues from the same patient. Our findings provide a new mechanistic understanding of nucleosome repositioning in tumour tissues that can be valuable for patient diagnostics, stratification and monitoring.
Collapse
Affiliation(s)
- Divya R Jacob
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Wilfried M Guiblet
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hulkar Mamayusupova
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Mariya Shtumpf
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Isabella Ciuta
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Luminita Ruje
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Svetlana Gretton
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
- School of Engineering, Arts, Science and Technology, University of Suffolk, James Hehir Building, University Avenue, Ipswich, Suffolk, IP3 0FS, UK
| | - Milena Bikova
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Clark Correa
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Emily Dellow
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Shivam P Agrawal
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Navid Shafiei
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | | | - Chris M Armstrong
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Jonathan D G Lam
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Yevhen Vainshtein
- Fraunhofer-Institut für Grenzflächen- und Bioverfahrenstechnik IGB, Nobelstraße 12, 70569, Stuttgart, Germany
| | - Christopher T Clarkson
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
- University College London, Gower St, Bloomsbury, London, WC1E 6BT, UK
| | - Graeme J Thorn
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Kai Sohn
- Fraunhofer-Institut für Grenzflächen- und Bioverfahrenstechnik IGB, Nobelstraße 12, 70569, Stuttgart, Germany
| | - Madapura M Pradeepa
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Sankaran Chandrasekharan
- Colchester General Hospital, East Suffolk and North Essex NHS Foundation Trust, Turner Road, Colchester, CO4 5JL, UK
| | - Greg N Brooke
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Elena Klenova
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Victor B Zhurkin
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Vladimir B Teif
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK.
| |
Collapse
|
46
|
Wang J, Zhang B, Chen X, Xin Y, Li K, Zhang C, Tang K, Tan Y. Cell mechanics regulate the migration and invasion of hepatocellular carcinoma cells via JNK signaling. Acta Biomater 2024; 176:321-333. [PMID: 38272199 DOI: 10.1016/j.actbio.2024.01.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Hepatocellular carcinoma (HCC) cells, especially those with metastatic competence, show reduced stiffness compared to the non-malignant counterparts. However, it is still unclear whether and how the mechanics of HCC cells influence their migration and invasion. This study reports that HCC cells with enhanced motility show reduced mechanical stiffness and cytoskeleton, suggesting the inverse correlation between cellular stiffness and motility. Through pharmacologic and genetic approaches, inhibiting actomyosin activity reduces HCC cellular stiffness but promotes their migration and invasion, while activating it increases cell stiffness but impairs cell motility. Actomyosin regulates cell motility through the influence on cellular stiffness. Mechanistically, weakening/strengthening cells inhibits/promotes c-Jun N terminal kinase (JNK) phosphorylation, activation/inhibition of which rescues the effects of cell mechanics on their migration and invasion. Further, HCC cancer stem cells (CSCs) exhibit higher motility but lower stiffness than control cells. Increasing CSC stiffness weakens migration and invasion through the activation of JNK signaling. In conclusion, our findings unveil a new regulatory role of actomyosin-mediated cellular mechanics in tumor cell motility and present new evidence to support that tumor cell softening may be one driving force for HCC metastasis. STATEMENT OF SIGNIFICANCE: Tumor cells progressively become softened during metastasis and low cell stiffness is associated with high metastatic potential. However, it remains unclear whether tumor cell softening is a by-product of or a driving force for tumor progression. This work reports that the stiffness of hepatocellular carcinoma cells is linked to their migration and invasion. Importantly, tumor cell softening promotes migration and invasion, while cell stiffening impairs the mobility. Weakening/strengthening cells inhibits/promotes JNK phosphorylation, activation/inhibition of which rescues the effects of cell mechanics on their migration and invasion ability. Further, stiffening liver cancer stem cells attenuates their motility through activating JNK signaling. In summary, our study uncovers a previously unappreciated role of tumor cell mechanics in migration and invasion and implicates the therapeutic potential of cell mechanics in the mechanotargeting of metastasis.
Collapse
Affiliation(s)
- Junfan Wang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Bai Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Xi Chen
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Cunyu Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Kai Tang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China.
| |
Collapse
|
47
|
Yanamandra AK, Zhang J, Montalvo G, Zhou X, Biedenweg D, Zhao R, Sharma S, Hoth M, Lautenschläger F, Otto O, Del Campo A, Qu B. PIEZO1-mediated mechanosensing governs NK-cell killing efficiency and infiltration in three-dimensional matrices. Eur J Immunol 2024; 54:e2350693. [PMID: 38279603 DOI: 10.1002/eji.202350693] [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: 07/31/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/28/2024]
Abstract
Natural killer (NK) cells play a vital role in eliminating tumorigenic cells. Efficient locating and killing of target cells in complex three-dimensional (3D) environments are critical for their functions under physiological conditions. However, the role of mechanosensing in regulating NK-cell killing efficiency in physiologically relevant scenarios is poorly understood. Here, we report that the responsiveness of NK cells is regulated by tumor cell stiffness. NK-cell killing efficiency in 3D is impaired against softened tumor cells, whereas it is enhanced against stiffened tumor cells. Notably, the durations required for NK-cell killing and detachment are significantly shortened for stiffened tumor cells. Furthermore, we have identified PIEZO1 as the predominantly expressed mechanosensitive ion channel among the examined candidates in NK cells. Perturbation of PIEZO1 abolishes stiffness-dependent NK-cell responsiveness, significantly impairs the killing efficiency of NK cells in 3D, and substantially reduces NK-cell infiltration into 3D collagen matrices. Conversely, PIEZO1 activation enhances NK killing efficiency as well as infiltration. In conclusion, our findings demonstrate that PIEZO1-mediated mechanosensing is crucial for NK killing functions, highlighting the role of mechanosensing in NK-cell killing efficiency under 3D physiological conditions and the influence of environmental physical cues on NK-cell functions.
Collapse
Affiliation(s)
- Archana K Yanamandra
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Jingnan Zhang
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Galia Montalvo
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Saarbrücken, Germany
| | - Xiangda Zhou
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Doreen Biedenweg
- Institute of Physics, University of Greifswald, Greifswald, Germany
| | - Renping Zhao
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Shulagna Sharma
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Franziska Lautenschläger
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
- Center for Biophysics, Saarland University, Saarbrücken, Germany
| | - Oliver Otto
- Institute of Physics, University of Greifswald, Greifswald, Germany
| | - Aránzazu Del Campo
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
- Chemistry Department, Saarland University, Saarbrücken, Germany
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
48
|
Luu N, Zhang S, Lam RHW, Chen W. Mechanical Constraints in Tumor Guide Emergent Spatial Patterns of Glioblastoma Cancer Stem Cells. MECHANOBIOLOGY IN MEDICINE 2024; 2:100027. [PMID: 38770108 PMCID: PMC11105673 DOI: 10.1016/j.mbm.2023.100027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The mechanical constraints in the overcrowding glioblastoma (GBM) microenvironment have been implicated in the regulation of tumor heterogeneity and disease progression. Especially, such mechanical cues can alter cellular DNA transcription and give rise to a subpopulation of tumor cells called cancer stem cells (CSCs). These CSCs with stem-like properties are critical drivers of tumorigenesis, metastasis, and treatment resistance. Yet, the biophysical and molecular machinery underlying the emergence of CSCs in tumor remained unexplored. This work employed a two-dimensional micropatterned multicellular model to examine the impact of mechanical constraints arisen from geometric confinement on the emergence and spatial patterning of CSCs in GBM tumor. Our study identified distinct spatial distributions of GBM CSCs in different geometric patterns, where CSCs mostly emerged in the peripheral regions. The spatial pattern of CSCs was found to correspond to the gradients of mechanical stresses resulted from the interplay between the cell-ECM and cell-cell interactions within the confined environment. Further mechanistic study highlighted a Piezo1-RhoA-focal adhesion signaling axis in regulating GBM cell mechanosensing and the subsequent CSC phenotypic transformation. These findings provide new insights into the biophysical origin of the unique spatial pattern of CSCs in GBM tumor and offer potential avenues for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Ngoc Luu
- Department of Biomedical Engineering, New York University, Brooklyn, NY, USA
| | - Shuhao Zhang
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, USA
| | - Raymond H. W. Lam
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Hong Kong SAR, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Weiqiang Chen
- Department of Biomedical Engineering, New York University, Brooklyn, NY, USA
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, USA
| |
Collapse
|
49
|
Azam I, Benson JD. Multiscale transport and 4D time-lapse imaging in precision-cut liver slices (PCLS). PeerJ 2024; 12:e16994. [PMID: 38426134 PMCID: PMC10903333 DOI: 10.7717/peerj.16994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Background Monitoring cellular processes across different levels of complexity, from the cellular to the tissue scale, is important for understanding tissue structure and function. However, it is challenging to monitor and estimate these structural and dynamic interactions within three-dimensional (3D) tissue models. Objective The aim of this study was to design a method for imaging, tracking, and quantifying 3D changes in cell morphology (shape and size) within liver tissue, specifically a precision-cut liver slice (PCLS). A PCLS is a 3D model of the liver that allows the study of the structure and function of liver cells in their native microenvironment. Methods Here, we present a method for imaging liver tissue during anisosmotic exposure in a multispectral four-dimensional manner. Three metrics of tissue morphology were measured to quantify the effects of osmotic stress on liver tissue. We estimated the changes in the volume of whole precision cut liver slices, quantified the changes in nuclei position, and calculated the changes in volumetric responses of tissue-embedded cells. Results During equilibration with cell-membrane-permeating and non-permeating solutes, the whole tissue experiences shrinkage and expansion. As nuclei showed a change in position and directional displacement under osmotic stress, we demonstrate that nuclei could be used as a probe to measure local osmotic and mechanical stress. Moreover, we demonstrate that cells change their volume within tissue slices as a result of osmotic perturbation and that this change in volume is dependent on the position of the cell within the tissue and the duration of the exposure. Conclusion The results of this study have implications for a better understanding of multiscale transport, mechanobiology, and triggered biological responses within complex biological structures.
Collapse
Affiliation(s)
- Iqra Azam
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - James D. Benson
- Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
50
|
Runel G, Lopez-Ramirez N, Barbollat-Boutrand L, Cario M, Durand S, Grimont M, Schartl M, Dalle S, Caramel J, Chlasta J, Masse I. Cancer Cell Biomechanical Properties Accompany Tspan8-Dependent Cutaneous Melanoma Invasion. Cancers (Basel) 2024; 16:694. [PMID: 38398085 PMCID: PMC10887418 DOI: 10.3390/cancers16040694] [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: 12/17/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The intrinsic biomechanical properties of cancer cells remain poorly understood. To decipher whether cell stiffness modulation could increase melanoma cells' invasive capacity, we performed both in vitro and in vivo experiments exploring cell stiffness by atomic force microscopy (AFM). We correlated stiffness properties with cell morphology adaptation and the molecular mechanisms underlying epithelial-to-mesenchymal (EMT)-like phenotype switching. We found that melanoma cell stiffness reduction was systematically associated with the acquisition of invasive properties in cutaneous melanoma cell lines, human skin reconstructs, and Medaka fish developing spontaneous MAP-kinase-induced melanomas. We observed a systematic correlation of stiffness modulation with cell morphological changes towards mesenchymal characteristic gains. We accordingly found that inducing melanoma EMT switching by overexpressing the ZEB1 transcription factor, a major regulator of melanoma cell plasticity, was sufficient to decrease cell stiffness and transcriptionally induce tetraspanin-8-mediated dermal invasion. Moreover, ZEB1 expression correlated with Tspan8 expression in patient melanoma lesions. Our data suggest that intrinsic cell stiffness could be a highly relevant marker for human cutaneous melanoma development.
Collapse
Affiliation(s)
- Gaël Runel
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
- BioMeca, 60F, Bioserra 2, Av. Rockefeller, 69008 Lyon, France
| | - Noémie Lopez-Ramirez
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Laetitia Barbollat-Boutrand
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Muriel Cario
- National Reference Center for Rare Skin Disease, Department of Dermatology, University Hospital, INSERM 1035, 33000 Bordeaux, France
- AquiDerm, University Bordeaux, 33076 Bordeaux, France
| | - Simon Durand
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Maxime Grimont
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Manfred Schartl
- Developmental Biochemistry, University of Würzburg, 97074 Würzburg, Germany
- Xiphophorus Genetic Stock Center, Texas State University, San Marcos, TX 78666, USA
| | - Stéphane Dalle
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
- Dermatology Department, Hôpital Universitaire Lyon Sud, Hospices Civils de Lyon, 69495 Pierre-Bénite, France
| | - Julie Caramel
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Julien Chlasta
- BioMeca, 60F, Bioserra 2, Av. Rockefeller, 69008 Lyon, France
| | - Ingrid Masse
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| |
Collapse
|