1
|
Wang H, Liu R, Yu Y, Xue H, Shen R, Zhang Y, Ding J. Effects of cell shape and nucleus shape on epithelial-mesenchymal transition revealed using chimeric micropatterns. Biomaterials 2025; 317:123013. [PMID: 39733514 DOI: 10.1016/j.biomaterials.2024.123013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/16/2024] [Accepted: 12/13/2024] [Indexed: 12/31/2024]
Abstract
Epithelial-mesenchymal transition (EMT) is a key phenotypic switch in cancer metastasis, leading to fatal consequences for patients. Under geometric constraints, the morphology of cancer cells changes in both cellular and subcellular levels, whose effects on EMT are, however, not fully understood. Herein, we designed and fabricated chimeric micropatterns of polystyrene (PS) with adhesion contrast to reveal the impacts of cell shapes and nuclear shapes on EMT in a decoupled way. Cell elongation was modulated via microwell aspect ratios (ARs), and nuclear deformation was generated through a micropillar array in the microwell. Human non-small cell lung cancer cells (A549) were cultured on the quasi-three dimensional micropatterned surfaces, and transforming growth factor-β1 (TGF-β1) was added to induce EMT. We found that chimeric micropatterns upregulated EMT with an increase of cellular AR and nuclear indentation under given TGF-β1. The subsequent assessment of the contractility and oriented assembly of microfilaments elucidated the key role of mechanotransduction in cell elongation and EMT, as proved by myosin inhibition, while it was obstructed by micropillars in the chimeric micropattern. Hence, the micropillar array possessed a nonmonotonic influence, enhancing the EMT of cells with AR of 1, but hindering the EMT with an impact more significant on microwells with large ARs due to the impeded cytoskeleton assembly. This fundamental research has illustrated the complex of cellular and subcellular geometries on cell behaviors including phenotype transition in cancer metastasis.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Ruili Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Yue Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Hongrui Xue
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Runjia Shen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Yanshuang Zhang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
2
|
Rodríguez-Montaño ÓL, Santoro L, Vaiani L, Lamberti L, Uva AE, Boccaccio A. Cell adhesion on substrates with variable curvature: Effects on genetic transcription processes. Comput Biol Med 2025; 189:109917. [PMID: 40023074 DOI: 10.1016/j.compbiomed.2025.109917] [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: 10/18/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Several studies suggest that changes in nuclear morphology due to forces and deformations as result of cell adhesion on biological substrates can induce molecular streaming through nuclear pore openings and alter chromatin structure. The condensed state of chromatin hinders transcription and replication, while its decompaction, induced by adhesion, plays a key role in differentiation. However, assessing nuclear stress/strain in vivo remains challenging, and the impact of substrate curvature on nuclear mechanics and chromatin structures is still unclear. In this study, we developed an axisymmetric finite element model of a mesenchymal stem cell adhering to substrates with different curvatures to analyze nuclear stress distribution and identify locations where adhesion-induced gene expression may occur. Results reveal a nuclear stress field with principal stresses in radial and circumferential directions, leading to chromatin decondensation and nuclear pore opening. The predicted forces acting on chromatin fibers, estimated and compared with experimental data, remain slightly below 5 pN-the threshold at which internucleosomal attraction is disrupted, triggering chromatin condensation-decondensation transition-. During early spreading, nuclear forces achieved through adhesion on convex substrates approach this threshold more closely than in concave or flat cases. These findings provide insights for tissue engineering and regenerative medicine, where early control of stem cell fate through substrate design is crucial. Understanding how mesenchymal stem cells respond to substrate curvature could lead to improved biomaterial surface topographies for guiding cell behavior. Tailoring curvature and mechanical properties may enhance early lineage commitment, optimizing regenerative strategies for tissue repair and organ regeneration.
Collapse
Affiliation(s)
| | - Lorenzo Santoro
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy
| | - Lorenzo Vaiani
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy
| | - Luciano Lamberti
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy
| | - Antonio E Uva
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy
| | - Antonio Boccaccio
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Bari, 70125, Italy.
| |
Collapse
|
3
|
Huang Y, Yang F, Liu C, Wang J, Wang Y, Song G, Wang Z. Mechanical Analysis of Phellinus Linteus-Induced Apoptosis of Hepatoma Cells. Microsc Res Tech 2025; 88:1491-1500. [PMID: 39806945 DOI: 10.1002/jemt.24804] [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: 06/17/2024] [Revised: 10/23/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025]
Abstract
Liver cancer is prevalent with the third highest mortality rate globally. The biomechanical properties of cancer cells play a crucial role in their proliferation and differentiation. Studying the morphological and mechanical properties of individual living cells can be helpful for early diagnosis of cancers. Herein, atomic force microscopy (AFM) was used to investigate the effects of Phellinus linteus on hepatocyte cells (HL-7702) and hepatocellular carcinoma cells (SMCC-7721) in terms of morphological and mechanical changes at the nanoscale. The water extract of Phellinus linteus (PLWE) resulted in increased height and surface roughness of SMCC-7721 cells. Also, the PLWE-treated showed that the average adhesion decreased by 1.69 nN and the average Young's modulus increased by 0.379 kPa. Additionally, the SMCC-7721 cells treated with PLWE showed clearly reduced activity compared with HL-7702 cells. This study suggested that Phellinus Linteus could be a potential candidate for selective anti-cancer therapy, providing a new avenue for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yuxi Huang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Fan Yang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Chuanzhi Liu
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Jianfei Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Ying Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Guicai Song
- College of Physics, Changchun University of Science and Technology, Changchun, China
| | - Zuobin Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
- JR3CN & IRAC, University of Bedfordshire, Luton, UK
| |
Collapse
|
4
|
Vinayak V, Basir R, Golloshi R, Toth J, Sant'Anna L, Lakadamyali M, McCord RP, Shenoy VB. Polymer model integrates imaging and sequencing to reveal how nanoscale heterochromatin domains influence gene expression. Nat Commun 2025; 16:3816. [PMID: 40268925 PMCID: PMC12019571 DOI: 10.1038/s41467-025-59001-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: 09/27/2024] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
Chromatin organization regulates gene expression, with nanoscale heterochromatin domains playing a fundamental role. Their size varies with microenvironmental stiffness and epigenetic interventions, but how these factors regulate their formation and influence transcription remains unclear. To address this, we developed a sequencing-informed copolymer model that simulates chromatin evolution through diffusion and active epigenetic reactions. Our model predicts the formation of nanoscale heterochromatin domains and quantifies how domain size scales with epigenetic reaction rates, showing that epigenetic and compaction changes primarily occur at domain boundaries. We validated these predictions via Hi-C and super-resolution imaging of hyperacetylated melanoma cells and identified differential expression of metastasis-related genes through RNA-seq. We validated our findings in hMSCs, where epigenetic reaction rates respond to microenvironmental stiffness. Conclusively, our simulations reveal that heterochromatin domain boundaries regulate gene expression and epigenetic memory. These findings demonstrate how external cues drive chromatin organization and transcriptional memory in development and disease.
Collapse
Affiliation(s)
- Vinayak Vinayak
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ramin Basir
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rosela Golloshi
- Departments of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Giovanis Institute for Translational Cell Biology, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Joshua Toth
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Lucas Sant'Anna
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Melike Lakadamyali
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Patton McCord
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, USA
| | - Vivek B Shenoy
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
McKee B, Abolghasemzade S, Wang TC, Harsh K, Kaur S, Blanchard R, Menon KB, Mohajeri M, Dickinson RB, Lele TP. Excess surface area of the nuclear lamina enables unhindered cell migration through constrictions. SCIENCE ADVANCES 2025; 11:eads6573. [PMID: 40153505 PMCID: PMC11952090 DOI: 10.1126/sciadv.ads6573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/25/2025] [Indexed: 03/30/2025]
Abstract
Cell migration through narrow spaces is essential in wound healing and metastatic spread of cancer. Cells must deform the large nucleus to fit through constricting channels. To understand the role of the nuclear lamina in limiting cell migration through constrictions, we imaged it in cells migrating through periodic constricting channels in a microdevice. The lamina underwent cycles of wrinkling and smoothing as the nucleus changed from an irregular, rounded shape in the wide channel regions between constrictions to a smooth, hourglass shape as the nucleus passed through the center of a constriction. The laminar surface area of nuclei within constrictions was measured to be at or above the computationally predicted threshold area for the nuclear volume. The channels excluded control nuclei that had insufficient excess surface area, but not nuclei lacking lamin A/C. Thus, the excess surface area of the nuclear lamina enables cell migration through constricting channels.
Collapse
Affiliation(s)
- Brendan McKee
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Samere Abolghasemzade
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Ting-Ching Wang
- McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Kajol Harsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Simran Kaur
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Ryan Blanchard
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Krishna Belraj Menon
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
- Indian Institute of Science Education and Research, Pune, Maharashtra 411008, India
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Richard B. Dickinson
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Tanmay P. Lele
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
- McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA
- Department of Translational Medical Sciences, Texas A&M University, College Station, TX 77843, USA
- Texas A&M University School of Engineering Medicine, 1020 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
6
|
Hong Y, Peng X, Yu H, Jafari M, Shakiba D, Huang Y, Qu C, Melika EE, Tawadros AK, Mujahid A, Huang YY, Sandler JA, Pryse KM, Sacks JM, Elson EL, Genin GM, Alisafaei F. Cell-matrix feedback controls stretch-induced cellular memory and fibroblast activation. Proc Natl Acad Sci U S A 2025; 122:e2322762122. [PMID: 40100625 PMCID: PMC11962495 DOI: 10.1073/pnas.2322762122] [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: 12/31/2023] [Accepted: 01/30/2025] [Indexed: 03/20/2025] Open
Abstract
Mechanical stretch can activate long-lived changes in fibroblasts, increasing their contractility and initiating phenotypic transformations. This activation, critical to wound healing and procedures such as skin grafting, increases with mechanical stimulus for cells cultured in two-dimensional but is highly variable in cells in three-dimensional (3D) tissue. Here, we show that static mechanical stretch of cells in 3D tissues can either increase or decrease fibroblast activation depending upon recursive cell-extracellular matrix (ECM) feedback and demonstrate control of this activation through integrated in vitro and mathematical models. ECM viscoelasticity, signaling dynamics, and cell mechanics combine to yield a predictable, but nonmonotonic, relationship between mechanical stretch and long-term cell activation. Results demonstrate that feedback between cells and ECM determine how cells retain memory of mechanical stretch and have direct implications for improving outcomes in skin grafting procedures.
Collapse
Affiliation(s)
- Yuan Hong
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
| | - Xiangjun Peng
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO63130
| | - Haomin Yu
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
| | - Mohammad Jafari
- NSF Science and Technology Center for Engineering Mechanobiology, Newark, NJ07102
- Department of Mechanical Engineering, New Jersey Institute of Technology, Newark, NJ07102
| | - Delaram Shakiba
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Yuxuan Huang
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO63130
| | - Chengqing Qu
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
| | - Ermia E. Melika
- NSF Science and Technology Center for Engineering Mechanobiology, Newark, NJ07102
- Department of Mechanical Engineering, New Jersey Institute of Technology, Newark, NJ07102
| | - Andrew K. Tawadros
- NSF Science and Technology Center for Engineering Mechanobiology, Newark, NJ07102
- Department of Mechanical Engineering, New Jersey Institute of Technology, Newark, NJ07102
| | - Aliza Mujahid
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- NSF Science and Technology Center for Engineering Mechanobiology, Newark, NJ07102
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ07102
| | - Yin-Yuan Huang
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO63130
| | - Jacob A. Sandler
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
| | - Kenneth M. Pryse
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
| | - Justin M. Sacks
- Division of Plastic and Reconstructive Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO63110
| | - Elliot L. Elson
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis School of Medicine, St. Louis, MO63110
| | - Guy M. Genin
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, MO63130
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO63130
| | - Farid Alisafaei
- NSF Science and Technology Center for Engineering Mechanobiology, Newark, NJ07102
- Department of Mechanical Engineering, New Jersey Institute of Technology, Newark, NJ07102
| |
Collapse
|
7
|
Alisafaei F, Shakiba D, Hong Y, Ramahdita G, Huang Y, Iannucci LE, Davidson MD, Jafari M, Qian J, Qu C, Ju D, Flory DR, Huang YY, Gupta P, Jiang S, Mujahid A, Singamaneni S, Pryse KM, Chao PHG, Burdick JA, Lake SP, Elson EL, Huebsch N, Shenoy VB, Genin GM. Tension anisotropy drives fibroblast phenotypic transition by self-reinforcing cell-extracellular matrix mechanical feedback. NATURE MATERIALS 2025:10.1038/s41563-025-02162-5. [PMID: 40128624 DOI: 10.1038/s41563-025-02162-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 01/28/2025] [Indexed: 03/26/2025]
Abstract
Mechanical factors such as stress in the extracellular environment affect the phenotypic commitment of cells. Stress fields experienced by cells in tissues are multiaxial, but how cells integrate such information is largely unknown. Here we report that the anisotropy of stress fields is a critical factor triggering a phenotypic transition in fibroblast cells, outweighing the role of stress amplitude, a factor previously described to modulate such a transition. Combining experimental and computational approaches, we identified a self-reinforcing mechanism in which cellular protrusions interact with collagen fibres to establish tension anisotropy. This anisotropy, in turn, stabilizes the protrusions and enhances their contractile forces. Disruption of this self-reinforcing process, either by reducing tension anisotropy or by inhibiting contractile protrusions, prevents the phenotypic conversion of fibroblasts to contractile myofibroblasts. Overall, our findings support stress anisotropy as a factor modulating cellular responses, expanding our understanding of the role of mechanical forces in biological processes.
Collapse
Affiliation(s)
- Farid Alisafaei
- NSF Science and Technology Center for Engineering Mechanobiology, Newark, NJ, USA
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Delaram Shakiba
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Yuan Hong
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Ghiska Ramahdita
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Yuxuan Huang
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Leanne E Iannucci
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Matthew D Davidson
- NSF Science and Technology Center for Engineering Mechanobiology, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Mohammad Jafari
- NSF Science and Technology Center for Engineering Mechanobiology, Newark, NJ, USA
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Jin Qian
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Chengqing Qu
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - David Ju
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Dashiell R Flory
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Yin-Yuan Huang
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Prashant Gupta
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Shumeng Jiang
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Aliza Mujahid
- NSF Science and Technology Center for Engineering Mechanobiology, Newark, NJ, USA
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Srikanth Singamaneni
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Kenneth M Pryse
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Pen-Hsiu Grace Chao
- Department of Biomedical Engineering, School of Engineering and School of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jason A Burdick
- NSF Science and Technology Center for Engineering Mechanobiology, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Spencer P Lake
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Elliot L Elson
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nathaniel Huebsch
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA.
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA.
| | - Vivek B Shenoy
- NSF Science and Technology Center for Engineering Mechanobiology, Philadelphia, PA, USA.
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.
| | - Guy M Genin
- NSF Science and Technology Center for Engineering Mechanobiology, Saint Louis, MO, USA.
- Department of Mechanical Engineering & Materials Science, Washington University in Saint Louis, Saint Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA.
| |
Collapse
|
8
|
Peng X, Huang Y, Kong W, Du Y, Elson EL, Feng XQ, Genin GM. Critical Cell Spacing Drives Phase Transition in Matrix-Mediated Tissue Condensation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.05.622090. [PMID: 40166242 PMCID: PMC11956908 DOI: 10.1101/2024.11.05.622090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Biological tissues exhibit phase transitions governed by mechanical feedback between cells and their extracellular matrix (ECM). We demonstrate through bio-chemo-mechanical modeling that this emergent behavior arises from competing physical effects: increasing matrix stiffness enhances individual cell activation while simultaneously weakening long-range mechanical communication. This competition establishes a critical cell spacing threshold (80-160 µ m) that precisely matches experimental observations across diverse cell types and collagen densities. Our model reveals that the critical stretch ratio at which fibrous networks transition from compliant to strain-stiffening governs this threshold through the formation of tension bands between neighboring cells. These tension bands create a mechanical percolation network that drives the collective phase transition in tissue behavior. Our model explains how fibrous architecture controls emergent mechanical properties in biological systems and offers insight into both the physics of fiber-reinforced composite materials under active stress, and into potential mechanical interventions for fibrotic disorders.
Collapse
|
9
|
Tang K, Zheng Y, Hu G, Xin Y, Li K, Zhang C, Chen X, Zhang B, Li X, Hu B, Jia Q, Zheng YP, Yang M, Tan Y. Local soft niches in mechanically heterogeneous primary tumors promote brain metastasis via mechanotransduction-mediated HDAC3 activity. SCIENCE ADVANCES 2025; 11:eadq2881. [PMID: 40009679 PMCID: PMC11864190 DOI: 10.1126/sciadv.adq2881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025]
Abstract
Tumor cells with organ-specific metastasis traits arise in primary lesions with substantial variations of local niche mechanics owing to intratumoral heterogeneity. However, the roles of mechanically heterogeneous primary tumor microenvironment in metastatic organotropism remain an enigma. This study reports that persistent priming in soft but not stiff niches that mimic primary tumor mechanical heterogeneity induces transcriptional reprogramming reminiscent of neuron and promotes the acquisition of brain metastatic potential. Soft-primed cells generate brain metastases in vivo through enhanced transendothelial migration across blood-brain barrier and brain colonization, which is further supported by the findings that tumor cells residing in local soft niches of primary xenografts exhibit brain metastatic tropism. Mechanistically, soft niches suppress cytoskeleton-nucleus-mediated mechanotransduction, which promotes histone deacetylase 3 activity. Inhibiting histone deacetylase 3 abolishes niche softness-induced brain metastatic ability. Collectively, this study uncovers a previously unappreciated role of local niche softness within primary tumors in brain metastasis, highlighting the significance of primary tumor mechanical heterogeneity in metastatic organotropism.
Collapse
Affiliation(s)
- Kai Tang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Yufan Zheng
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Guanshuo Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Cunyu Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Xi Chen
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Bai Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Xueyi Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Bing Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Qiong Jia
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, 210006
| | - Yong-ping Zheng
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| |
Collapse
|
10
|
Li H, Harvey DH, Dai J, Swingle SP, Compton AM, Sugali CK, Dhamodaran K, Yao J, Lin TY, Sulchek T, Kim T, Ethier CR, Mao W. Characterization, Enrichment, and Computational Modeling of Cross-Linked Actin Networks in Transformed Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2025; 66:65. [PMID: 40009371 PMCID: PMC11878246 DOI: 10.1167/iovs.66.2.65] [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: 08/17/2024] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Purpose Cross-linked actin networks (CLANs) are prevalent in the glaucomatous trabecular meshwork (TM). We previously developed the GTM3L cell line, which spontaneously forms fluorescently labeled CLANs, by transducing GTM3, a transformed glaucomatous TM cell line, with a lentivirus expressing the LifeAct-GFP fusion protein. Here, we determined if LifeAct-GFP viral copy numbers are associated with CLANs, developed approaches to increase CLAN incidence, and computationally studied the biomechanical properties of CLAN-containing GTM3L cells. Methods GTM3L cells were fluorescently sorted for viral copy number analysis to determine whether increased CLAN incidence was associated with copy number. CLAN incidence was increased by combining (1) differential adhesion sorting, (2) cell deswelling, and (3) cell stiffness selection. GTM3L cells were cultured on glass or soft hydrogels for stiffness measurement by atomic force microscopy. Computational models studied the biomechanical properties of CLANs. Results GTM3L cells had one LifeAct-GFP viral copy/cell on average, and viral copy number or LifeAct-GFP expression level did not associate with CLAN incidence rate. However, CLAN rate was increased from -0.28% to -50% by combining the three enrichment methods noted above. Further, GTM3L cells formed more CLANs on a stiff versus a soft substrate. Computational modeling predicted that CLANs contribute to higher cell stiffness, including increased resistance of the nucleus to tensile stress when CLANs are physically linked to the nucleus. Conclusions It is possible to greatly enhance CLAN incidence in GTM3L cells. CLANs are mechanosensitive structures that affect cell biomechanical properties. Further research is needed to determine the biomechanics, mechanobiology, and etiology of CLANs in the TM.
Collapse
Affiliation(s)
- Haiyan Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, Georgia, United States
| | - Devon H. Harvey
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana, United States
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana, United States
| | - Jiannong Dai
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana, United States
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana, United States
| | - Steven P. Swingle
- Department of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Anthony M. Compton
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, Georgia, United States
| | - Chenna Kesavulu Sugali
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana, United States
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana, United States
| | - Kamesh Dhamodaran
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana, United States
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana, United States
| | - Jing Yao
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, United States
| | - Tsai-Yu Lin
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana, United States
| | - Todd Sulchek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, Georgia, United States
- Department of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Taeyoon Kim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States
| | - C. Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, Georgia, United States
- Department of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Weiming Mao
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana, United States
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana, United States
- Department of Biochemistry & Molecular Biology, Indiana University, Indianapolis, Indiana, United States
| |
Collapse
|
11
|
Shin SJ, Bayarkhangai B, Tsogtbaatar K, Yuxuan M, Kim S, Kim Y, Taghizadeh A, Kim D, Kim D, Lee J, Hyun J, Kim H. Matrix-Rigidity Cooperates With Biochemical Cues in M2 Macrophage Activation Through Increased Nuclear Deformation and Chromatin Accessibility. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403409. [PMID: 39828979 PMCID: PMC11848612 DOI: 10.1002/advs.202403409] [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: 04/01/2024] [Revised: 06/19/2024] [Indexed: 01/22/2025]
Abstract
Macrophages encounter a myriad of biochemical and mechanical stimuli across various tissues and pathological contexts. Notably, matrix rigidity has emerged as a pivotal regulator of macrophage activation through mechanotransduction. However, the precise mechanisms underlying the interplay between mechanical and biochemical cues within the nuclear milieu remain elusive. Here We elucidate how the increased matrix rigidity drives macrophages to amplify alternatively-activated (M2 phenotype) signaling cooperatively with biochemical cues (e.g., IL4/13) through altered nuclear mechanics. We demonstrate that reconstructed podosome-like F-actins and contractility induce nucleus deformation, opening nuclear pores, which facilitates nuclear translocation of the key transcription factor STAT6. Furthermore, the altered nuclear mechanics increases chromatin accessibility induced by H3K9 methylation, particularly of M2-associated gene promoters. These cooperative events of the mechano-chemical signaling at the cytoskeletal-to-nuclear domains facilitate M2 transcriptional activation and cellular functions. We further evidence the rigidity-primed M2 macrophages are immunosuppressive and accumulated within stiffened tumors in patients. This study proposes a mechanism by which matrix mechanics crosstalks with biochemical signals to potentiate macrophage activation through nuclear mechanosensing and chromatin modifications, offering insights into macrophage mechanobiology and its therapeutic modulations.
Collapse
Affiliation(s)
- Seung Jae Shin
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
| | - Buuvee Bayarkhangai
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Khaliunsarnai Tsogtbaatar
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Meng Yuxuan
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Sang‐Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of ChemistryCollege of Science & TechnologyDankook UniversityCheonan31116Republic of Korea
| | - Yong‐Jae Kim
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Ali Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Daesan Kim
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Dong‐Hwee Kim
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- Department of Integrative Energy EngineeringCollege of EngineeringKorea UniversitySeoul02841Republic of Korea
- Biomedical Research CenterKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Jung‐Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
- UCL Eastman‐Korea Dental Medicine Innovation CentreDankook UniversityCheonan31116Republic of Korea
- Department of Biomaterials ScienceSchool of DentistryDankook UniversityCheonan31116Republic of Korea
- Cell & Matter InstituteDankook UniversityCheonan31116Republic of Korea
| | - Jeongeun Hyun
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
- UCL Eastman‐Korea Dental Medicine Innovation CentreDankook UniversityCheonan31116Republic of Korea
- Department of Regenerative Dental MedicineSchool of DentistryDankook UniversityCheonan31116Republic of Korea
| | - Hae‐Won Kim
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
- Mechanobiology Dental Medicine Research CenterDankook UniversityCheonan31116Republic of Korea
- Department of Nanobiomedical Sciences & BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
- UCL Eastman‐Korea Dental Medicine Innovation CentreDankook UniversityCheonan31116Republic of Korea
- Cell & Matter InstituteDankook UniversityCheonan31116Republic of Korea
- Department of Regenerative Dental MedicineSchool of DentistryDankook UniversityCheonan31116Republic of Korea
| |
Collapse
|
12
|
Zhou W, Lin J, Wang Q, Wang X, Yao X, Yan Y, Sun W, Zhu Q, Zhang X, Wang X, Ji B, Ouyang H. Chromatin-site-specific accessibility: A microtopography-regulated door into the stem cell fate. Cell Rep 2025; 44:115106. [PMID: 39723890 DOI: 10.1016/j.celrep.2024.115106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 11/12/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Biomaterials that mimic extracellular matrix topography are crucial in tissue engineering. Previous research indicates that certain biomimetic topography can guide stem cells toward multiple specific lineages. However, the mechanisms by which topographic cues direct stem cell differentiation remain unclear. Here, we demonstrate that microtopography influences nuclear tension in mesenchymal stem cells (MSCs), shaping chromatin accessibility and determining lineage commitment. On aligned substrates, MSCs exhibit high cytoskeletal tension along the fiber direction, creating anisotropic nuclear stress that opens chromatin sites for neurogenic, myogenic, and tenogenic genes via transcription factors like Nuclear receptor TLX (TLX). In contrast, random substrates induce isotropic nuclear stress, promoting chromatin accessibility for osteogenic and chondrogenic genes through Runt-related transcription factors (RUNX). Our findings reveal that aligned and random microtopographies direct site-specific chromatin stretch and lineage-specific gene expression, priming MSCs for distinct lineages. This study introduces a novel framework for understanding how topographic cues govern cell fate in tissue repair and regeneration.
Collapse
Affiliation(s)
- Wenyan Zhou
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; School of Medicine, Taizhou University, Taizhou, Zhejiang Province 318000, China
| | - Junxin Lin
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; School of Medicine, Taizhou University, Taizhou, Zhejiang Province 318000, China
| | - Qianchun Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325000, China
| | - Xianliu Wang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 200051, China
| | - Xudong Yao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province 322000, China
| | - Yiyang Yan
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang Province 314400, China
| | - Wei Sun
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang Province 314400, China
| | - Qiuwen Zhu
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Xiaoan Zhang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Xiaozhao Wang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China
| | - Baohua Ji
- Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, Zhejiang Province 310027, China; Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310027, China.
| | - Hongwei Ouyang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 311121, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang Province 314400, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang Province 310058, China.
| |
Collapse
|
13
|
Tam SW, Cheung AKL, Qin P, Zhang S, Huang Z, Yung KKL. Extracellular Silica Nanomatrices Promote In Vitro Maturation of Anti-tumor Dendritic Cells via Activation of Focal Adhesion Kinase. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2314358. [PMID: 39268785 PMCID: PMC11733713 DOI: 10.1002/adma.202314358] [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: 12/30/2023] [Revised: 08/12/2024] [Indexed: 09/15/2024]
Abstract
The efficacy of dendritic cell (DC)-based cancer vaccines is critically determined by the functionalities of in vitro maturated DCs. The maturation of DCs typically relies on chemicals that are cytotoxic or hinder the ability of DCs to efficiently activate the antigen-specific cytotoxic T-lymphocytes (CTLs) against tumors. Herein, the maturation chemicals are replaced with extracellular silica nanomatrices, fabricated by glancing angle deposition, to promote in vitro maturation of murine bone marrow-derived DCs (mBMDCs). The extracellular nanomatrices composed of silica nanozigzags (NZs) enable the generation of mature mBMDCs with upregulated levels of co-stimulatory molecules, C-C chemokine receptor type-7, X-C motif chemokine recetpor-1, DC-specific ICAM-3 grabbing nonintegrin, and enhanced endocytic capacity. The in vitro maturation is partially governed by focal adhesion kinase (FAK) that is mechanically activated in the curved cell adhesions formed at the DC-NZ interfaces. The NZ-maturated mBMDCs can prime the antigen-specific CTLs into programmed cell death protein-1 (PD-1)lowCD44high memory phenotypes in vitro and suppress the growth of tumors in vivo. Meanwhile, the NZ-mediated beneficial effects are also observed in human monocyte-derived DCs. This work demonstrates that the silica NZs promote the anti-tumor capacity of in vitro maturated DCs via the mechanoactivation of FAK, supporting the potential of silica NZs being a promising biomaterial for cancer immunotherapy.
Collapse
Affiliation(s)
- Sze Wah Tam
- Department of BiologyHong Kong Baptist UniversityKowloonHong Kong SARChina
- Golden Meditech Center for NeuroRegeneration SciencesHong Kong Baptist UniversityKowloonHong Kong SARChina
| | | | - Ping Qin
- Department of BiologyHong Kong Baptist UniversityKowloonHong Kong SARChina
- Golden Meditech Center for NeuroRegeneration SciencesHong Kong Baptist UniversityKowloonHong Kong SARChina
| | - Shiqing Zhang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentJinan UniversityGuangzhou510632China
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug ResearchCollege of PharmacyJinan UniversityGuangzhou510632China
| | - Zhifeng Huang
- Department of ChemistryThe Chinese University of Hong Kong (CUHK)Shatin, N.T.Hong Kong SARChina
- Shenzhen Research Institute of CUHKNo. 10, 2nd Yuexing Road, NanshanShenzhenGuangdong518057China
| | - Ken Kin Lam Yung
- Department of Science and Environmental Studiesthe Education University of Hong KongN.T.Hong Kong SARChina
| |
Collapse
|
14
|
Wang TC, Abolghasemzade S, McKee BP, Singh I, Pendyala K, Mohajeri M, Patel H, Shaji A, Kersey AL, Harsh K, Kaur S, Dollahon CR, Chukkapalli S, Lele PP, Conway DE, Gaharwar AK, Dickinson RB, Lele TP. Matrix stiffness drives drop like nuclear deformation and lamin A/C tension-dependent YAP nuclear localization. Nat Commun 2024; 15:10151. [PMID: 39578439 PMCID: PMC11584751 DOI: 10.1038/s41467-024-54577-4] [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: 03/21/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Extracellular matrix (ECM) stiffness influences cancer cell fate by altering gene expression. Previous studies suggest that stiffness-induced nuclear deformation may regulate gene expression through YAP nuclear localization. We investigated the role of the nuclear lamina in this process. We show that the nuclear lamina exhibits mechanical threshold behavior: once unwrinkled, the nuclear lamina is inextensible. A computational model predicts that the unwrinkled lamina is under tension, which is confirmed using a lamin tension sensor. Laminar unwrinkling is caused by nuclear flattening during cell spreading on stiff ECM. Knockdown of lamin A/C eliminates nuclear surface tension and decreases nuclear YAP localization. These findings show that nuclear deformation in cells conforms to the nuclear drop model and reveal a role for lamin A/C tension in controlling YAP localization in cancer cells.
Collapse
Affiliation(s)
- Ting-Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Samere Abolghasemzade
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Brendan P McKee
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Ishita Singh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kavya Pendyala
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Hailee Patel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Aakansha Shaji
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Anna L Kersey
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kajol Harsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Simran Kaur
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Christina R Dollahon
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Sasanka Chukkapalli
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Pushkar P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA
| | - Richard B Dickinson
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Tanmay P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA.
| |
Collapse
|
15
|
Bakht SM, Pardo A, Gomez‐Florit M, Caballero D, Kundu SC, Reis RL, Domingues RMA, Gomes ME. Human Tendon-on-Chip: Unveiling the Effect of Core Compartment-T Cell Spatiotemporal Crosstalk at the Onset of Tendon Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401170. [PMID: 39258510 PMCID: PMC11538684 DOI: 10.1002/advs.202401170] [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: 01/31/2024] [Revised: 06/27/2024] [Indexed: 09/12/2024]
Abstract
The lack of representative in vitro models recapitulating human tendon (patho)physiology is among the major factors hindering consistent progress in the knowledge-based development of adequate therapies for tendinopathy.Here, an organotypic 3D tendon-on-chip model is designed that allows studying the spatiotemporal dynamics of its cellular and molecular mechanisms.Combining the synergistic effects of a bioactive hydrogel matrix with the biophysical cues of magnetic microfibers directly aligned on the microfluidic chip, it is possible to recreate the anisotropic architecture, cell patterns, and phenotype of tendon intrinsic (core) compartment. When incorporated with vascular-like vessels emulating the interface between its intrinsic-extrinsic compartments, crosstalk with endothelial cells are found to drive stromal tenocytes toward a reparative profile. This platform is further used to study adaptive immune cell responses at the onset of tissue inflammation, focusing on interactions between tendon compartment tenocytes and circulating T cells.The proinflammatory signature resulting from this intra/inter-cellular communication induces the recruitment of T cells into the inflamed core compartment and confirms the involvement of this cellular crosstalk in positive feedback loops leading to the amplification of tendon inflammation.Overall, the developed 3D tendon-on-chip provides a powerful new tool enabling mechanistic studies on the pathogenesis of tendinopathy as well as for assessing new therapies.
Collapse
Affiliation(s)
- Syeda M. Bakht
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/GuimarãesPortugal
| | - Alberto Pardo
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/GuimarãesPortugal
- Colloids and Polymers Physics GroupParticle Physics DepartmentMaterials Institute (iMATUS)and Health Research Institute (IDIS)University of Santiago de CompostelaSantiago de Compostela15782Spain
| | | | - David Caballero
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/GuimarãesPortugal
| | - Subhas C. Kundu
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/GuimarãesPortugal
| | - Rui L. Reis
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/GuimarãesPortugal
| | - Rui M. A. Domingues
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/GuimarãesPortugal
| | - Manuela E. Gomes
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/GuimarãesPortugal
- School of Medicine and Biomedical Sciences (ICBAS), Unit for Multidisciplinary Research in Biomedicine (UMIB)University of PortoRua Jorge Viterbo Ferreira 228Porto4050‐313 PortoPortugal
| |
Collapse
|
16
|
Swoger M, Thanh MTH, Byfield FJ, Dam V, Williamson J, Frank B, Hehnly H, Conway D, Patteson AE. Vimentin molecular linkages with nesprin-3 enhance nuclear deformations by cell geometric constraints. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.621001. [PMID: 39554181 PMCID: PMC11565891 DOI: 10.1101/2024.10.29.621001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The nucleus is the organelle of the cell responsible for controlling protein expression, which has a direct effect on cellular biological functions. Here we show that the cytoskeletal protein vimentin plays an important role in increasing cell-generated forces transmitted to the cell nucleus, resulting in increased nuclear deformations in strongly polarized cells. Using micropatterned substrates to geometrically control cell shape in wild-type and vimentin-null cells, we show vimentin increases polarization and deformation of the cell nucleus. Loss of nesprin-3, which physically couples vimentin to the nuclear envelope, phenotypically copies the loss of vimentin, suggesting vimentin transmits forces to the cell nucleus through direct molecular linkages. Use of a fluorescence resonance energy transfer (FRET) sensor that binds to the nuclear envelope through lamin-A/C suggests vimentin increases the tension on the nuclear envelope. Our results indicate that nuclear shape and deformation can be modified by the vimentin cytoskeleton and its specific crosslinks to the cell nucleus.
Collapse
|
17
|
Pavlov DA, Heffler J, Suay-Corredera C, Dehghany M, Shen KM, Zuela-Sopilniak N, Randell R, Uchida K, Jain R, Shenoy V, Lammerding J, Prosser B. Microtubule forces drive nuclear damage in LMNA cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.10.579774. [PMID: 38948795 PMCID: PMC11212868 DOI: 10.1101/2024.02.10.579774] [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
Nuclear homeostasis requires a balance of forces between the cytoskeleton and nucleus. Mutations in the LMNA gene, which encodes the nuclear envelope proteins lamin A/C, disrupt this balance by weakening the nuclear lamina. This results in nuclear damage in contractile tissues and ultimately muscle disease. Intriguingly, disrupting the LINC complex that connects the cytoskeleton to the nucleus has emerged as a promising strategy to ameliorate LMNA-associated cardiomyopathy. Yet how LINC complex disruption protects the cardiomyocyte nucleus remains unclear. To address this, we developed an assay to quantify the coupling of cardiomyocyte contraction to nuclear deformation and interrogated its dependence on the nuclear lamina and LINC complex. We found that, surprisingly, the LINC complex was mostly dispensable for transferring contractile strain to the nucleus, and that increased nuclear strain in lamin A/C-deficient cardiomyocytes was not rescued by LINC complex disruption. Instead, LINC complex disruption eliminated the cage of microtubules encircling the nucleus. Disrupting microtubules was sufficient to prevent nuclear damage and rescue cardiac function induced by lamin A/C deficiency. We computationally simulated the stress fields surrounding cardiomyocyte nuclei and show how microtubule forces generate local vulnerabilities that damage lamin A/C-deficient nuclei. Our work pinpoints localized, microtubule-dependent force transmission through the LINC complex as a pathological driver and therapeutic target for LMNA-cardiomyopathy.
Collapse
Affiliation(s)
- Daria Amiad Pavlov
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania
| | - Julie Heffler
- Weill Institute for Cell and Molecular Biology & Meinig School of Biomedical Engineering, Cornell University
| | - Carmen Suay-Corredera
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania
| | - Mohammad Dehghany
- Department of Materials Science and Engineering, Center for Engineering Mechanobiology, University of Pennsylvania
| | - Kaitlyn M. Shen
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania
| | - Noam Zuela-Sopilniak
- Weill Institute for Cell and Molecular Biology & Meinig School of Biomedical Engineering, Cornell University
| | - Rani Randell
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania
| | - Keita Uchida
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania
| | - Rajan Jain
- Departments of Medicine and Cell and Developmental Biology, Penn Cardiovascular Institute, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania
| | - Vivek Shenoy
- Department of Materials Science and Engineering, Center for Engineering Mechanobiology, University of Pennsylvania
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology & Meinig School of Biomedical Engineering, Cornell University
| | - Benjamin Prosser
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
18
|
Dickinson RB, Abolghasemzade S, Lele TP. Rethinking nuclear shaping: insights from the nuclear drop model. SOFT MATTER 2024; 20:7558-7565. [PMID: 39105242 PMCID: PMC11446230 DOI: 10.1039/d4sm00683f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Changes in the nuclear shape caused by cellular shape changes are generally assumed to reflect an elastic deformation from a spherical nuclear shape. Recent evidence, however, suggests that the nuclear lamina, which forms the outer nuclear surface together with the nuclear envelope, possesses more area than that of a sphere of the same volume. This excess area manifests as folds/wrinkles in the nuclear surface in rounded cells and allows facile nuclear flattening during cell spreading without any changes in nuclear volume or surface area. When the lamina becomes smooth and taut, it is inextensible, and supports a surface tension. At this point, it is possible to mathematically calculate the limiting nuclear shape purely based on geometric considerations. In this paper, we provide a commentary on the "nuclear drop model" which seeks to integrate the above features. We outline its testable physical properties and explore its biological implications.
Collapse
Affiliation(s)
- Richard B Dickinson
- Department of Chemical Engineering, University of Florida, 1030 Center Drive, Gainesville, FL, 32611, USA.
| | - Samere Abolghasemzade
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St., College Station, TX, 77843, USA.
| | - Tanmay P Lele
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St., College Station, TX, 77843, USA.
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, 3122 TAMU, College Station, TX, 77843, USA
- Department of Translational Medical Sciences, Texas A&M University, 2121 W Holcombe St., Houston, TX, 77030, USA
| |
Collapse
|
19
|
Gupta S, Swoger M, Saldanha R, Schwarz JM, Patteson AE. Reorganizing chromatin by cellular deformation. Curr Opin Cell Biol 2024; 90:102408. [PMID: 39121805 PMCID: PMC11392642 DOI: 10.1016/j.ceb.2024.102408] [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: 01/14/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024]
Abstract
Biologists have the capability to edit a genome at the nanometer scale and then observe whether or not the edit affects the structure of a developing organ or organism at the centimeter scale. Our understanding of the underlying mechanisms driving this emergent phenomenon from a multiscale perspective remains incomplete. This review focuses predominantly on recent experimental developments in uncovering the mechanical interplay between the chromatin and cell scale since mechanics plays a major role in determining nuclear, cellular, and tissue structure. Here, we discuss the generation and transmission of forces through the cytoskeleton, affecting chromatin diffusivity and organization. Decoding such pieces of these multiscale connections lays the groundwork for solving the genotype-to-phenotype puzzle in biology.
Collapse
Affiliation(s)
- Sarthak Gupta
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY, USA; Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
| | - Maxx Swoger
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - Renita Saldanha
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY, USA
| | - J M Schwarz
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY, USA; Indian Creek Farm, Ithaca, NY, USA
| | - Alison E Patteson
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY, USA.
| |
Collapse
|
20
|
Wang H, Weng X, Chen Y, Mao S, Gao Y, Wu Q, Huang Y, Guan X, Xu Z, Lai Y. Biomimetic concentric microgrooved titanium surfaces influence bone marrow-derived mesenchymal stem cell osteogenic differentiation via H3K4 trimethylation epigenetic regulation. Dent Mater J 2024; 43:683-692. [PMID: 39135261 DOI: 10.4012/dmj.2023-327] [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: 10/01/2024]
Abstract
Material surface micromorphology can modulate cellular behavior and promote osteogenic differentiation through cytoskeletal rearrangement. Bone reconstruction requires precise regulation of gene expression in cells, a process governed by epigenetic mechanisms such as histone modifications, DNA methylation, and chromatin remodeling. We constructed osteon-mimetic concentric microgrooved titanium surfaces with different groove sizes and cultured bone marrow-derived mesenchymal stem cells (BMSCs) on the material surfaces to study how they regulate cell biological behavior and osteogenic differentiation through epigenetics. We found that the cells arranged in concentric circles along the concentric structure in the experimental group, and the concentric microgrooved surface did not inhibit cell proliferation. The results of a series of osteogenic differentiation experiments showed that the concentric microgrooves facilitated calcium deposition and promoted osteogenic differentiation of the BMSCs. Concentric microgrooved titanium surfaces that were 30 μm wide and 10 μm deep promoted osteogenic differentiation of BMSC by increasing WDR5 expression via H3K4 trimethylation upregulation.
Collapse
Affiliation(s)
- Hong Wang
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
- Stomatological Hospital of Xiamen Medical college
| | - Xinze Weng
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Yan Chen
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Shunjie Mao
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Yuerong Gao
- Department of Stomatology of The Third Affiliated Hospital of Xi'an Medical University
| | - Qinglin Wu
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Yanling Huang
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Xin Guan
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Zhiqiang Xu
- Department of Stomatology, Affiliated Hospital of Putian University
| | - Yingzhen Lai
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| |
Collapse
|
21
|
Mubarok W, Elvitigala KCML, Nakaya H, Hotta T, Sakai S. Cell Cycle Modulation through Physical Confinement in Micrometer-Thick Hydrogel Sheaths. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:18717-18726. [PMID: 39166379 DOI: 10.1021/acs.langmuir.4c02434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Recently, surface engineering of the cell membrane with biomaterials has attracted great attention for various biomedical applications. In this study, we investigated the possibility of modulating cell cycle progression using alginate and gelatin-based hydrogel sheaths with a thickness of ∼1 μm. The hydrogel sheath was formed on cell surfaces through cross-linking catalyzed by horseradish peroxidase immobilized on the cell surface. The hydrogel sheath did not decrease the viability (>95% during 2 days of culture) of the human cervical carcinoma cell line (HeLa) expressing the fluorescent ubiquitination-based cell cycle indicator 2 (HeLa/Fucci2). Coating the HeLa/Fucci2 cells with the hydrogel sheath resulted in a cell cycle arrest in the G2/M phase, which can be caused by the reduced F-actin formation. As a result of this cell cycle arrest, an inhibition of cell growth was observed in the HeLa/Fucci2 cells. Taken together, our results demonstrate that the hydrogel sheath coating on the cell surface is a feasible approach to modulating cell cycle progression.
Collapse
Affiliation(s)
- Wildan Mubarok
- Division of Chemical Engineering, Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, Osaka 560-8531, Japan
| | - Kelum Chamara Manoj Lakmal Elvitigala
- Division of Chemical Engineering, Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, Osaka 560-8531, Japan
| | - Hiroto Nakaya
- Graduate School of Frontier Bioscience, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tomoki Hotta
- Division of Chemical Engineering, Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, Osaka 560-8531, Japan
| | - Shinji Sakai
- Division of Chemical Engineering, Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, Osaka 560-8531, Japan
| |
Collapse
|
22
|
Li YM, Ji Y, Meng YX, Kim YJ, Lee H, Kurian AG, Park JH, Yoon JY, Knowles JC, Choi Y, Kim YS, Yoon BE, Singh RK, Lee HH, Kim HW, Lee JH. Neural Tissue-Like, not Supraphysiological, Electrical Conductivity Stimulates Neuronal Lineage Specification through Calcium Signaling and Epigenetic Modification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400586. [PMID: 38984490 PMCID: PMC11425260 DOI: 10.1002/advs.202400586] [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: 01/16/2024] [Revised: 06/28/2024] [Indexed: 07/11/2024]
Abstract
Electrical conductivity is a pivotal biophysical factor for neural interfaces, though optimal values remain controversial due to challenges isolating this cue. To address this issue, conductive substrates made of carbon nanotubes and graphene oxide nanoribbons, exhibiting a spectrum of conductivities from 0.02 to 3.2 S m-1, while controlling other surface properties is designed. The focus is to ascertain whether varying conductivity in isolation has any discernable impact on neural lineage specification. Remarkably, neural-tissue-like low conductivity (0.02-0.1 S m-1) prompted neural stem/progenitor cells to exhibit a greater propensity toward neuronal lineage specification (neurons and oligodendrocytes, not astrocytes) compared to high supraphysiological conductivity (3.2 S m-1). High conductivity instigated the apoptotic process, characterized by increased apoptotic fraction and decreased neurogenic morphological features, primarily due to calcium overload. Conversely, cells exposed to physiological conductivity displayed epigenetic changes, specifically increased chromatin openness with H3acetylation (H3ac) and neurogenic-transcription-factor activation, along with a more balanced intracellular calcium response. The pharmacological inhibition of H3ac further supported the idea that such epigenetic changes might play a key role in driving neuronal specification in response to neural-tissue-like, not supraphysiological, conductive cues. These findings underscore the necessity of optimal conductivity when designing neural interfaces and scaffolds to stimulate neuronal differentiation and facilitate the repair process.
Collapse
Affiliation(s)
- Yu-Meng Li
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Yunseong Ji
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Fuel Cell Laboratory, Korea Institute of Energy Research (KIER), Daejeon, 34129, Republic of Korea
| | - Yu-Xuan Meng
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Yu-Jin Kim
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Hwalim Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Amal George Kurian
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Jeong-Hui Park
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Ji-Young Yoon
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Jonathan C Knowles
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, Royal Free Hospital, Rowland Hill Street, London, NW3 2PF, UK
| | - Yunkyu Choi
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yoon-Sik Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Bo-Eun Yoon
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Rajendra K Singh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
23
|
Granero-Moya I, Venturini V, Belthier G, Groenen B, Molina-Jordán M, González-Martín M, Trepat X, van Rheenen J, Andreu I, Roca-Cusachs P. Nucleocytoplasmic transport senses mechanical forces independently of cell density in cell monolayers. J Cell Sci 2024; 137:jcs262363. [PMID: 39120491 PMCID: PMC11423809 DOI: 10.1242/jcs.262363] [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: 06/18/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Cells sense and respond to mechanical forces through mechanotransduction, which regulates processes in health and disease. In single adhesive cells, mechanotransduction involves the transmission of force from the extracellular matrix to the cell nucleus, where it affects nucleocytoplasmic transport (NCT) and the subsequent nuclear localization of transcriptional regulators, such as YAP (also known as YAP1). However, if and how NCT is mechanosensitive in multicellular systems is unclear. Here, we characterize and use a fluorescent sensor of nucleocytoplasmic transport (Sencyt) and demonstrate that NCT responds to mechanical forces but not cell density in cell monolayers. Using monolayers of both epithelial and mesenchymal phenotype, we show that NCT is altered in response both to osmotic shocks and to the inhibition of cell contractility. Furthermore, NCT correlates with the degree of nuclear deformation measured through nuclear solidity, a shape parameter related to nuclear envelope tension. In contrast, YAP is sensitive to cell density, showing that the YAP response to cell-cell contacts is not via a mere mechanical effect of NCT. Our results demonstrate the generality of the mechanical regulation of NCT.
Collapse
Affiliation(s)
- Ignasi Granero-Moya
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08014 Barcelona, Spain
- University of Barcelona, 08036 Barcelona, Spain
| | - Valeria Venturini
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08014 Barcelona, Spain
| | - Guillaume Belthier
- Oncode Institute, 1066 CX Amsterdam, The Netherlands
- Department of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Bart Groenen
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08014 Barcelona, Spain
- Eindhoven University of Technology, Department of Biomedical Engineering, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Marc Molina-Jordán
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08014 Barcelona, Spain
| | - Miguel González-Martín
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08014 Barcelona, Spain
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08014 Barcelona, Spain
- University of Barcelona, 08036 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08014 Barcelona, Spain
| | - Jacco van Rheenen
- Oncode Institute, 1066 CX Amsterdam, The Netherlands
- Department of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Ion Andreu
- Biofisika Institute (CSIC, UPV/EHU), 48940 Leioa, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08014 Barcelona, Spain
- University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
24
|
Jaganathan A, Toth J, Chen X, Basir R, Pieuchot L, Shen Y, Reinhart-King C, Shenoy VB. Mechano-metabolism of metastatic breast cancer cells in 2D and 3D microenvironments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591879. [PMID: 38746096 PMCID: PMC11092625 DOI: 10.1101/2024.04.30.591879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Cells regulate their shape and metabolic activity in response to the mechano-chemical properties of their microenvironment. To elucidate the impact of matrix stiffness and ligand density on the bioenergetics of mesenchymal cells, we developed a nonequilibrium, active chemo-mechanical model that accounts for the mechanical energy of the cell and matrix, chemical energy from ATP hydrolysis, interfacial energy, and mechano-sensitive regulation of stress fiber assembly through signaling. By integrating the kinetics and energetics of these processes, we define the cell "metabolic potential" that, when minimized, provides testable predictions of cell contractility, shape, and ATP consumption. Specifically, we show that the morphology of MDA-MB-231 breast cancer cells in 3D collagen changes from spherical to elongated to spherical with increasing matrix stiffness, which is consistent with experimental observations. On 2D hydrogels, our model predicts a hemispherical-to-spindle-to-disc shape transition with increasing gel stiffness. In both cases, we show that these shape transitions emerge from competition between the energy of ATP hydrolysis associated with increased contractility that drives cell elongation and the interfacial energy that favors a rounded shape. Furthermore, our model can predict how increased energy demand in stiffer microenvironments is met by AMPK activation, which is confirmed experimentally in both 2D and 3D microenvironments and found to correlate with the upregulation of mitochondrial potential, glucose uptake, and ATP levels, as well as provide estimates of changes in intracellular adenosine nucleotide concentrations with changing environmental stiffness. Overall, we present a framework for relating adherent cell energy levels and contractility through biochemical regulation of underlying physical processes. Statement of Significance Increasing evidence indicates that cellular metabolism is regulated by mechanical cues from the extracellular environment. Forces transmitted from the microenvironment activate mechanotransduction pathways in the cell, which trigger a cascade of biochemical events that impact cytoskeletal tension, cellular morphology and energy budget available to the cell. Using a nonequilibrium free energy-based theory, we can predict the ATP consumption, contractility, and shape of mesenchymal cancer cells, as well as how cells regulate energy levels dependent on the mechanosensitive metabolic regulator AMPK. The insights from our model can be used to understand the mechanosensitive regulation of metabolism during metastasis and tumor progression, during which cells experience dynamic changes in their microenvironment and metabolic state.
Collapse
|
25
|
Rashid F, Kabbo SA, Wang N. Mechanomemory of nucleoplasm and RNA polymerase II after chromatin stretching by a microinjected magnetic nanoparticle force. Cell Rep 2024; 43:114462. [PMID: 39002538 PMCID: PMC11289711 DOI: 10.1016/j.celrep.2024.114462] [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: 03/20/2024] [Revised: 06/09/2024] [Accepted: 06/23/2024] [Indexed: 07/15/2024] Open
Abstract
Increasing evidence suggests that the mechanics of chromatin and nucleoplasm regulate gene transcription and nuclear function. However, how the chromatin and nucleoplasm sense and respond to forces remains elusive. Here, we employed a strategy of applying forces directly to the chromatin of a cell via a microinjected 200-nm anti-H2B-antibody-coated ferromagnetic nanoparticle (FMNP) and an anti-immunoglobulin G (IgG)-antibody-coated or an uncoated FMNP. The chromatin behaved as a viscoelastic gel-like structure and the nucleoplasm was a softer viscoelastic structure at loading frequencies of 0.1-5 Hz. Protein diffusivity of the chromatin, nucleoplasm, and RNA polymerase II (RNA Pol II) and RNA Pol II activity were upregulated in a chromatin-stretching-dependent manner and stayed upregulated for tens of minutes after force cessation. Chromatin stiffness increased, but the mechanomemory duration of chromatin diffusivity decreased, with substrate stiffness. These findings may provide a mechanomemory mechanism of transcription upregulation and have implications on cell and nuclear functions.
Collapse
Affiliation(s)
- Fazlur Rashid
- The Institute for Mechanobiology, Northeastern University, Boston, MA 02115, USA; Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA 02115, USA; Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Sadia Amin Kabbo
- The Institute for Mechanobiology, Northeastern University, Boston, MA 02115, USA; Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA 02115, USA
| | - Ning Wang
- The Institute for Mechanobiology, Northeastern University, Boston, MA 02115, USA; Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Dhankhar M, Guo Z, Kant A, Basir R, Joshi R, Heo SC, Mauck RL, Lakadamyali M, Shenoy VB. Revealing the Biophysics of Lamina-Associated Domain Formation by Integrating Theoretical Modeling and High-Resolution Imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600310. [PMID: 38979207 PMCID: PMC11230226 DOI: 10.1101/2024.06.24.600310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The interactions between chromatin and the nuclear lamina orchestrate cell type-specific gene activity by forming lamina-associated domains (LADs) which preserve cellular characteristics through gene repression. However, unlike the interactions between chromatin segments, the strength of chromatin-lamina interactions and their dependence on cellular environment are not well understood. Here, we develop a theory to predict the size and shape of peripheral heterochromatin domains by considering the energetics of chromatin-chromatin interactions, the affinity between chromatin and the nuclear lamina and the kinetics of methylation and acetylation9in human mesenchymal stem cells (hMSCs). Through the analysis of super-resolution images of peripheral heterochromatin domains using this theoretical framework, we determine the nuclear lamina-wide distribution of chromatin-lamina affinities. We find that the extracted affinity is highly spatially heterogeneous and shows a bimodal distribution, indicating regions along the lamina with strong chromatin binding and those exhibiting vanishing chromatin affinity interspersed with some regions exhibiting a relatively diminished chromatin interactions, in line with the presence of structures such as nuclear pores. Exploring the role of environmental cues on peripheral chromatin, we find that LAD thickness increases when hMSCs are cultured on a softer substrate, in correlation with contractility-dependent translocation of histone deacetylase 3 (HDAC3) from the cytosol to the nucleus. In soft microenvironments, chromatin becomes sequestered at the nuclear lamina, likely due to the interactions of HDAC3 with the chromatin anchoring protein LAP2 β ,increasing chromatin-lamina affinity, as well as elevated levels of the intranuclear histone methylation. Our findings are further corroborated by pharmacological interventions that inhibit contractility, as well as by manipulating methylation levels using epigenetic drugs. Notably, in the context of tendinosis, a chronic condition characterized by collagen degeneration, we observed a similar increase in the thickness of peripheral chromatin akin to that of cells cultured on soft substrates consistent with theoretical predictions. Our findings underscore the pivotal role of the microenvironment in shaping genome organization and highlight its relevance in pathological conditions.
Collapse
|
27
|
Wyle Y, Lu N, Hepfer J, Sayal R, Martinez T, Wang A. The Role of Biophysical Factors in Organ Development: Insights from Current Organoid Models. Bioengineering (Basel) 2024; 11:619. [PMID: 38927855 PMCID: PMC11200479 DOI: 10.3390/bioengineering11060619] [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: 04/17/2024] [Revised: 05/26/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Biophysical factors play a fundamental role in human embryonic development. Traditional in vitro models of organogenesis focused on the biochemical environment and did not consider the effects of mechanical forces on developing tissue. While most human tissue has a Young's modulus in the low kilopascal range, the standard cell culture substrate, plasma-treated polystyrene, has a Young's modulus of 3 gigapascals, making it 10,000-100,000 times stiffer than native tissues. Modern in vitro approaches attempt to recapitulate the biophysical niche of native organs and have yielded more clinically relevant models of human tissues. Since Clevers' conception of intestinal organoids in 2009, the field has expanded rapidly, generating stem-cell derived structures, which are transcriptionally similar to fetal tissues, for nearly every organ system in the human body. For this reason, we conjecture that organoids will make their first clinical impact in fetal regenerative medicine as the structures generated ex vivo will better match native fetal tissues. Moreover, autologously sourced transplanted tissues would be able to grow with the developing embryo in a dynamic, fetal environment. As organoid technologies evolve, the resultant tissues will approach the structure and function of adult human organs and may help bridge the gap between preclinical drug candidates and clinically approved therapeutics. In this review, we discuss roles of tissue stiffness, viscoelasticity, and shear forces in organ formation and disease development, suggesting that these physical parameters should be further integrated into organoid models to improve their physiological relevance and therapeutic applicability. It also points to the mechanotransductive Hippo-YAP/TAZ signaling pathway as a key player in the interplay between extracellular matrix stiffness, cellular mechanics, and biochemical pathways. We conclude by highlighting how frontiers in physics can be applied to biology, for example, how quantum entanglement may be applied to better predict spontaneous DNA mutations. In the future, contemporary physical theories may be leveraged to better understand seemingly stochastic events during organogenesis.
Collapse
Affiliation(s)
- Yofiel Wyle
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
| | - Nathan Lu
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Jason Hepfer
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Rahul Sayal
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Taylor Martinez
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California-Davis, Davis, CA 95616, USA
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA 95817, USA
| |
Collapse
|
28
|
Alasaadi DN, Mayor R. Mechanically guided cell fate determination in early development. Cell Mol Life Sci 2024; 81:242. [PMID: 38811420 PMCID: PMC11136904 DOI: 10.1007/s00018-024-05272-6] [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: 02/01/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024]
Abstract
Cell fate determination, a vital process in early development and adulthood, has been the focal point of intensive investigation over the past decades. Its importance lies in its critical role in shaping various and diverse cell types during embryonic development and beyond. Exploration of cell fate determination started with molecular and genetic investigations unveiling central signaling pathways and molecular regulatory networks. The molecular studies into cell fate determination yielded an overwhelming amount of information invoking the notion of the complexity of cell fate determination. However, recent advances in the framework of biomechanics have introduced a paradigm shift in our understanding of this intricate process. The physical forces and biochemical interplay, known as mechanotransduction, have been identified as a pivotal drive influencing cell fate decisions. Certainly, the integration of biomechanics into the process of cell fate pushed our understanding of the developmental process and potentially holds promise for therapeutic applications. This integration was achieved by identifying physical forces like hydrostatic pressure, fluid dynamics, tissue stiffness, and topography, among others, and examining their interplay with biochemical signals. This review focuses on recent advances investigating the relationship between physical cues and biochemical signals that control cell fate determination during early embryonic development.
Collapse
Affiliation(s)
- Delan N Alasaadi
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
29
|
Sun Q, Pan X, Wang P, Wei Q. Synergistic Influence of Fibrous Pattern Orientation and Modulus on Cellular Mechanoresponse. NANO LETTERS 2024; 24:6376-6385. [PMID: 38743504 DOI: 10.1021/acs.nanolett.4c01352] [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: 05/16/2024]
Abstract
The fibrous extracellular matrix (ECM) is vital for tissue regeneration and impacts implanted device treatments. Previous research on fibrous biomaterials shows varying cellular reactions to surface orientation, often due to unclear interactions between surface topography and substrate elasticity. Our study addresses this gap by achieving the rapid creation of hydrogels with diverse fibrous topographies and varying substrate moduli through a surface printing strategy. Cells exhibit heightened traction force on nanopatterned soft hydrogels, particularly with randomly distributed patterns compared with regular soft hydrogels. Meanwhile, on stiff hydrogels featuring an aligned topography, optimal cellular mechanosensing is observed compared to random topography. Mechanistic investigations highlight that cellular force-sensing and adhesion are influenced by the interplay of pattern deformability and focal adhesion orientation, subsequently mediating stem cell differentiation. Our findings highlight the importance of combining substrate modulus and topography to guide cellular behavior in designing advanced tissue engineering biomaterials.
Collapse
Affiliation(s)
- Qian Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiaokai Pan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Peng Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| |
Collapse
|
30
|
Alisafaei F, Mandal K, Saldanha R, Swoger M, Yang H, Shi X, Guo M, Hehnly H, Castañeda CA, Janmey PA, Patteson AE, Shenoy VB. Vimentin is a key regulator of cell mechanosensing through opposite actions on actomyosin and microtubule networks. Commun Biol 2024; 7:658. [PMID: 38811770 PMCID: PMC11137025 DOI: 10.1038/s42003-024-06366-4] [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: 11/14/2022] [Accepted: 05/21/2024] [Indexed: 05/31/2024] Open
Abstract
The cytoskeleton is a complex network of interconnected biopolymers consisting of actin filaments, microtubules, and intermediate filaments. These biopolymers work in concert to transmit cell-generated forces to the extracellular matrix required for cell motility, wound healing, and tissue maintenance. While we know cell-generated forces are driven by actomyosin contractility and balanced by microtubule network resistance, the effect of intermediate filaments on cellular forces is unclear. Using a combination of theoretical modeling and experiments, we show that vimentin intermediate filaments tune cell stress by assisting in both actomyosin-based force transmission and reinforcement of microtubule networks under compression. We show that the competition between these two opposing effects of vimentin is regulated by the microenvironment stiffness. These results reconcile seemingly contradictory results in the literature and provide a unified description of vimentin's effects on the transmission of cell contractile forces to the extracellular matrix.
Collapse
Affiliation(s)
- Farid Alisafaei
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Kalpana Mandal
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Medicine and Engineering, University of Pennsylvania, 3340 Smith Walk, Philadelphia, PA, 19104, USA
| | - Renita Saldanha
- Physics Department, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA
| | - Maxx Swoger
- Physics Department, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA
| | - Haiqian Yang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Xuechen Shi
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Medicine and Engineering, University of Pennsylvania, 3340 Smith Walk, Philadelphia, PA, 19104, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Heidi Hehnly
- Department of Biology, Syracuse University, Syracuse, NY, 13244, USA
| | - Carlos A Castañeda
- Departments of Biology and Chemistry, Syracuse University, Syracuse, NY, 13244, USA
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA
| | - Paul A Janmey
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Medicine and Engineering, University of Pennsylvania, 3340 Smith Walk, Philadelphia, PA, 19104, USA
- Departments of Physiology, and Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alison E Patteson
- Physics Department, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA
| | - Vivek B Shenoy
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
31
|
Dupouy G, Dong Y, Herzog E, Chabouté ME, Berr A. Nuclear envelope dynamics in connection to chromatin remodeling. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2024; 118:963-981. [PMID: 37067011 DOI: 10.1111/tpj.16246] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/29/2023] [Accepted: 04/12/2023] [Indexed: 05/11/2023]
Abstract
The nucleus is a central organelle of eukaryotic cells undergoing dynamic structural changes during cellular fundamental processes such as proliferation and differentiation. These changes rely on the integration of developmental and stress signals at the nuclear envelope (NE), orchestrating responses at the nucleo-cytoplasmic interface for efficient genomic functions such as DNA transcription, replication and repair. While in animals, correlation has already been established between NE dynamics and chromatin remodeling using last-generation tools and cutting-edge technologies, this topic is just emerging in plants, especially in response to mechanical cues. This review summarizes recent data obtained in this field with more emphasis on the mechanical stress response. It also highlights similarities/differences between animal and plant cells at multiples scales, from the structural organization of the nucleo-cytoplasmic continuum to the functional impacts of NE dynamics.
Collapse
Affiliation(s)
- Gilles Dupouy
- Institut de Biologie Moléculaire des Plantes du CNRS- Université de Strasbourg, 12 rue du Général Zimmer,, F-67084, Strasbourg, France
| | - Yihan Dong
- Institut de Biologie Moléculaire des Plantes du CNRS- Université de Strasbourg, 12 rue du Général Zimmer,, F-67084, Strasbourg, France
| | - Etienne Herzog
- Institut de Biologie Moléculaire des Plantes du CNRS- Université de Strasbourg, 12 rue du Général Zimmer,, F-67084, Strasbourg, France
| | - Marie-Edith Chabouté
- Institut de Biologie Moléculaire des Plantes du CNRS- Université de Strasbourg, 12 rue du Général Zimmer,, F-67084, Strasbourg, France
| | - Alexandre Berr
- Institut de Biologie Moléculaire des Plantes du CNRS- Université de Strasbourg, 12 rue du Général Zimmer,, F-67084, Strasbourg, France
| |
Collapse
|
32
|
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
|
33
|
Tajvidi Safa B, Huang C, Kabla A, Yang R. Active viscoelastic models for cell and tissue mechanics. ROYAL SOCIETY OPEN SCIENCE 2024; 11:231074. [PMID: 38660600 PMCID: PMC11040246 DOI: 10.1098/rsos.231074] [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: 07/24/2023] [Revised: 02/01/2024] [Accepted: 02/25/2024] [Indexed: 04/26/2024]
Abstract
Living cells are out of equilibrium active materials. Cell-generated forces are transmitted across the cytoskeleton network and to the extracellular environment. These active force interactions shape cellular mechanical behaviour, trigger mechano-sensing, regulate cell adaptation to the microenvironment and can affect disease outcomes. In recent years, the mechanobiology community has witnessed the emergence of many experimental and theoretical approaches to study cells as mechanically active materials. In this review, we highlight recent advancements in incorporating active characteristics of cellular behaviour at different length scales into classic viscoelastic models by either adding an active tension-generating element or adjusting the resting length of an elastic element in the model. Summarizing the two groups of approaches, we will review the formulation and application of these models to understand cellular adaptation mechanisms in response to various types of mechanical stimuli, such as the effect of extracellular matrix properties and external loadings or deformations.
Collapse
Affiliation(s)
- Bahareh Tajvidi Safa
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE68588, USA
| | - Changjin Huang
- School of Mechanical & Aerospace Engineering, Nanyang Technological University, Singapore639798, Singapore
| | - Alexandre Kabla
- Department of Engineering, University of Cambridge, CambridgeCB2 1PZ, UK
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE68588, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI48824, USA
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI48824, USA
| |
Collapse
|
34
|
Chen N, Wan X, Wang M, Li Y, Wang X, Zeng L, Zhou J, Zhang Y, Cheng S, Shen Y. Cross-talk between Vimentin and autophagy regulates blood-testis barrier disruption induced by cadmium. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 346:123625. [PMID: 38401636 DOI: 10.1016/j.envpol.2024.123625] [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: 12/28/2023] [Revised: 02/06/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
The blood-testis barrier (BTB) plays a vital role in mammalian spermatogenesis by separating the seminiferous epithelium into an adluminal and a basal compartment. Cadmium (Cd) is a toxic heavy metal that is widely present in the environment. We observed that Cd can induce BTB disruption, leading to apoptosis of testicular cells. However, the molecular mechanisms contributing to BTB injury induced by Cd have not yet been fully clarified. Vimentin (Vim) is an important desmosome-like junction protein that mediates robust adhesion in the BTB. In this study, we investigated how Vim responds to Cd. We found that Cd treatment led to a significant decrease in Vim expression, accompanied by a marked increase in LC3-II expression and a higer number of autophagosomes. Interestingly, we also observed that Cd-induced autophagy was associated with decreased Vim activity and enhanced apoptosis of testicular cells. To further investigate the role of autophagy in Vim regulation under Cd exposure, we treated cells with an autophagy inhibitor called 3-MA. We found that 3-MA treatment enhanced Vim expression and improved the disruption of the BTB under Cd exposure. Additionally, the inhibition of Vim confirmed the role of autophagy in modulating Vim expression. These results reveal a previously unknown regulatory mechanism of Cd involving the interplay between a heavy metal and a protein.
Collapse
Affiliation(s)
- Na Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China
| | - Xiaoyan Wan
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510620, PR China
| | - Mei Wang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, PR China
| | - Yamin Li
- Department of Woman's Health Care, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430071, Hubei, PR China
| | - Xiaofei Wang
- Center for Reproductive Medicine, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443000, Hubei, PR China
| | - Ling Zeng
- Medical Genetics Center, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, PR China
| | - Jinzhao Zhou
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Yanwei Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Shun Cheng
- College of Zhixing, Hubei University, Wuhan, 430011, PR China
| | - Yi Shen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China.
| |
Collapse
|
35
|
Cao R, Tian H, Tian Y, Fu X. A Hierarchical Mechanotransduction System: From Macro to Micro. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302327. [PMID: 38145330 PMCID: PMC10953595 DOI: 10.1002/advs.202302327] [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: 04/12/2023] [Revised: 10/27/2023] [Indexed: 12/26/2023]
Abstract
Mechanotransduction is a strictly regulated process whereby mechanical stimuli, including mechanical forces and properties, are sensed and translated into biochemical signals. Increasing data demonstrate that mechanotransduction is crucial for regulating macroscopic and microscopic dynamics and functionalities. However, the actions and mechanisms of mechanotransduction across multiple hierarchies, from molecules, subcellular structures, cells, tissues/organs, to the whole-body level, have not been yet comprehensively documented. Herein, the biological roles and operational mechanisms of mechanotransduction from macro to micro are revisited, with a focus on the orchestrations across diverse hierarchies. The implications, applications, and challenges of mechanotransduction in human diseases are also summarized and discussed. Together, this knowledge from a hierarchical perspective has the potential to refresh insights into mechanotransduction regulation and disease pathogenesis and therapy, and ultimately revolutionize the prevention, diagnosis, and treatment of human diseases.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Huimin Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Yan Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Xianghui Fu
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| |
Collapse
|
36
|
Fenelon KD, Krause J, Koromila T. Opticool: Cutting-edge transgenic optical tools. PLoS Genet 2024; 20:e1011208. [PMID: 38517915 PMCID: PMC10959397 DOI: 10.1371/journal.pgen.1011208] [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] [Indexed: 03/24/2024] Open
Abstract
Only a few short decades have passed since the sequencing of GFP, yet the modern repertoire of transgenically encoded optical tools implies an exponential proliferation of ever improving constructions to interrogate the subcellular environment. A myriad of tags for labeling proteins, RNA, or DNA have arisen in the last few decades, facilitating unprecedented visualization of subcellular components and processes. Development of a broad array of modern genetically encoded sensors allows real-time, in vivo detection of molecule levels, pH, forces, enzyme activity, and other subcellular and extracellular phenomena in ever expanding contexts. Optogenetic, genetically encoded optically controlled manipulation systems have gained traction in the biological research community and facilitate single-cell, real-time modulation of protein function in vivo in ever broadening, novel applications. While this field continues to explosively expand, references are needed to assist scientists seeking to use and improve these transgenic devices in new and exciting ways to interrogate development and disease. In this review, we endeavor to highlight the state and trajectory of the field of in vivo transgenic optical tools.
Collapse
Affiliation(s)
- Kelli D. Fenelon
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| | - Julia Krause
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| | - Theodora Koromila
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
37
|
Ma T, Liu X, Su H, Shi Q, He Y, Wu F, Gao C, Li K, Liang Z, Zhang D, Zhang X, Hu K, Li S, Wang L, Wang M, Yue S, Hong W, Chen X, Zhang J, Zheng L, Deng X, Wang P, Fan Y. Coupling of Perinuclear Actin Cap and Nuclear Mechanics in Regulating Flow-Induced Yap Spatiotemporal Nucleocytoplasmic Transport. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305867. [PMID: 38161226 PMCID: PMC10953556 DOI: 10.1002/advs.202305867] [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: 08/19/2023] [Revised: 11/10/2023] [Indexed: 01/03/2024]
Abstract
Mechanical forces, including flow shear stress, govern fundamental cellular processes by modulating nucleocytoplasmic transport of transcription factors like Yes-associated Protein (YAP). However, the underlying mechanical mechanism remains elusive. In this study, it is reported that unidirectional flow induces biphasic YAP transport with initial nuclear import, followed by nuclear export as actin cap formation and nuclear stiffening. Conversely, pathological oscillatory flow induces slight actin cap formation, nuclear softening, and sustained YAP nuclear localization. To elucidate the disparately YAP spatiotemporal distribution, a 3D mechanochemical model is developed, which integrates flow sensing, cytoskeleton organization, nucleus mechanotransduction, and YAP transport. The results unveiled that despite the significant localized nuclear stress imposed by the actin cap, its inherent stiffness counteracts the dispersed contractile stress exerted by conventional fibers on the nuclear membrane. Moreover, alterations in nuclear stiffness synergistically regulate nuclear deformation, thereby governing YAP transport. Furthermore, by expanding the single-cell model to a collective vertex framework, it is revealed that the irregularities in actin cap formation within individual cells have the potential to induce topological defects and spatially heterogeneous YAP distribution in the cellular monolayer. This work unveils a unified mechanism of flow-induced nucleocytoplasmic transport, providing a linkage between transcription factor localization and mechanical stimulation.
Collapse
Affiliation(s)
- Tianxiang Ma
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Xiao Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Haoran Su
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Qiusheng Shi
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Yuan He
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Fan Wu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Chenxing Gao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Kexin Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Zhuqing Liang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Dongrui Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Xing Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Ke Hu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Shangyu Li
- Biomedical Pioneering Innovation Center (BIOPIC)Peking UniversityBeijing100871China
- Academy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
| | - Li Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Min Wang
- Department of Gynecology and ObstetricsStrategic Support Force Medical CenterBeijing100101China
| | - Shuhua Yue
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Weili Hong
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Xun Chen
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Jing Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Lisha Zheng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Xiaoyan Deng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Pu Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
- School of Engineering MedicineBeihang UniversityBeijing100083China
| |
Collapse
|
38
|
Dai Y, Xie Q, Zhang Y, Sun Y, Zhu S, Wang C, Tan Y, Gou X. Neoteric Semiembedded β-Tricalcium Phosphate Promotes Osteogenic Differentiation of Mesenchymal Stem Cells under Cyclic Stretch. ACS APPLIED MATERIALS & INTERFACES 2024; 16:8289-8300. [PMID: 38329794 DOI: 10.1021/acsami.3c15090] [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: 02/09/2024]
Abstract
β-Tricalcium phosphate (β-TCP) is a bioactive material for bone regeneration, but its brittleness limits its use as a standalone scaffold. Therefore, continuous efforts are necessary to effectively integrate β-TCP into polymers, facilitating a sturdy ion exchange for cell regulation. Herein, a novel semiembedded technique was utilized to anchor β-TCP nanoparticles onto the surface of the elastic polymer, followed by hydrophilic modification with the polymerization of dopamine. Cell adhesion and osteogenic differentiation of mesenchymal stem cells (MSCs) under static and dynamic uniaxial cyclic stretching conditions were investigated. The results showed that the new strategy was effective in promoting cell adhesion, proliferation, and osteogenic induction by the sustained release of Ca2+ in the vicinity and creating a reasonable roughness. Specifically, released Ca2+ from β-TCP could activate the calcium signaling pathway, which further upregulated calmodulin and calcium/calmodulin-dependent protein kinase II genes in MSCs. Meanwhile, the roughness of the membrane and the uniaxial cyclic stretching activated the PIEZO1 signaling pathway. Chemical and mechanical stimulation promotes osteogenic differentiation and increases the expression of related genes 2-8-fold. These findings demonstrated that the neoteric semiembedded structure was a promising strategy in controlling both chemical and mechanical factors of biomaterials for cell regulation.
Collapse
Affiliation(s)
- Yujie Dai
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Qingyun Xie
- Department of Orthopedics, General Hospital of Western Theater Command, Chengdu 610031, China
| | - Yimeng Zhang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Yiwan Sun
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Shaomei Zhu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Chongyu Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. 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
| | - Xue Gou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| |
Collapse
|
39
|
Song Y, Soto J, Wong SY, Wu Y, Hoffman T, Akhtar N, Norris S, Chu J, Park H, Kelkhoff DO, Ang CE, Wernig M, Kasko A, Downing TL, Poo MM, Li S. Biphasic regulation of epigenetic state by matrix stiffness during cell reprogramming. SCIENCE ADVANCES 2024; 10:eadk0639. [PMID: 38354231 PMCID: PMC10866547 DOI: 10.1126/sciadv.adk0639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
We investigate how matrix stiffness regulates chromatin reorganization and cell reprogramming and find that matrix stiffness acts as a biphasic regulator of epigenetic state and fibroblast-to-neuron conversion efficiency, maximized at an intermediate stiffness of 20 kPa. ATAC sequencing analysis shows the same trend of chromatin accessibility to neuronal genes at these stiffness levels. Concurrently, we observe peak levels of histone acetylation and histone acetyltransferase (HAT) activity in the nucleus on 20 kPa matrices, and inhibiting HAT activity abolishes matrix stiffness effects. G-actin and cofilin, the cotransporters shuttling HAT into the nucleus, rises with decreasing matrix stiffness; however, reduced importin-9 on soft matrices limits nuclear transport. These two factors result in a biphasic regulation of HAT transport into nucleus, which is directly demonstrated on matrices with dynamically tunable stiffness. Our findings unravel a mechanism of the mechano-epigenetic regulation that is valuable for cell engineering in disease modeling and regenerative medicine applications.
Collapse
Affiliation(s)
- Yang Song
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jennifer Soto
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Sze Yue Wong
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yifan Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Navied Akhtar
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Sam Norris
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Julia Chu
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hyungju Park
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Structure and Function of Neural Network, Korea Brain Research Institute (KBRI), Daegu 41068, South Korea
| | - Douglas O. Kelkhoff
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Cheen Euong Ang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Pathology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Marius Wernig
- Department of Pathology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Andrea Kasko
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Timothy L. Downing
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Mu-ming Poo
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
40
|
Subramanian D, Tjahjono N, Hernandez PA, Varner VD, Petroll WM, Schmidtke DW. Fabrication of Micropatterns of Aligned Collagen Fibrils. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:2551-2561. [PMID: 38277615 PMCID: PMC11001481 DOI: 10.1021/acs.langmuir.3c02676] [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] [Indexed: 01/28/2024]
Abstract
Many tissues in vivo contain aligned structures such as filaments, fibrils, and fibers, which expose cells to anisotropic structural and topographical cues that range from the nanometer to micrometer scales. Understanding how cell behavior is regulated by these cues during physiological and pathological processes (e.g., wound healing, cancer invasion) requires substrates that can expose cells to anisotropic cues over several length scales. In this study, we developed a novel method of fabricating micropatterns of aligned collagen fibrils of different geometry onto PDMS-coated glass coverslips that allowed us to investigate the roles of topography and confinement on corneal cell behavior. When corneal cells were cultured on micropatterns of aligned collagen fibrils in the absence of confinement, the degree of cell alignment increased from 40 ± 14 to 82 ± 5% as the size of the micropattern width decreased from 750 to 50 μm. Although the cell area (∼2500 μm2), cell length (∼160 μm), and projected nuclear area (∼175 μm2) were relatively constant on the different micropattern widths, cells displayed an increased aspect ratio as the width of the aligned collagen fibril micropatterns decreased. We also observed that the morphology of cells adhering to the surrounding uncoated PDMS was dependent upon both the size of the aligned collagen fibril micropattern and the distance from the micropatterns. When corneal cells were confined to the micropatterns of aligned collagen fibrils by a Pluronic coating to passivate the surrounding area, a similar trend in increasing cell alignment was observed (35 ± 10 to 89 ± 2%). However, the projected nuclear area decreased significantly (∼210 to 130 μm2) as the micropattern width decreased from 750 to 50 μm. The development of this method allows for the deposition of aligned collagen fibril micropatterns of different geometries on a transparent and elastic substrate and provides an excellent model system to investigate the role of anisotropic cues in cell behavior.
Collapse
Affiliation(s)
- Divya Subramanian
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
| | - Nathaniel Tjahjono
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
| | - Paula A. Hernandez
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center at Dallas, TX, 75390
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center at Dallas, TX, 75390
| | - Victor D. Varner
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
- Department of Surgery, University of Texas Southwestern Medical Center at Dallas, TX, 75390
| | - W. Matthew Petroll
- Department of Ophthalmology, University of Texas Southwestern Medical Center at Dallas, TX, 75390
| | - David W. Schmidtke
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
- Department of Surgery, University of Texas Southwestern Medical Center at Dallas, TX, 75390
| |
Collapse
|
41
|
Bermudez A, Muñoz SN, Blaik R, Rowat AC, Hu J, Lin NYC. Using Histologic Image Analysis to Understand Biophysical Regulations of Epithelial Cell Morphology. BIOPHYSICIST (ROCKVILLE, MD.) 2023; 5:1-14. [PMID: 39165674 PMCID: PMC11335341 DOI: 10.35459/tbp.2023.000253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Epithelial mechanics and mechanobiology have become 2 important research fields in life sciences and bioengineering. These fields investigate how physical factors induced by cell adhesion and collective behaviors can directly regulate biologic processes, such as organ development and disease progression. Cell mechanics and mechanobiology thus make exciting biophysics education topics to illustrate how fundamental physics principles play a role in regulating cell biology. However, the field currently lacks hands-on activities that engage students in learning science and outreach programs in these topics. One such area is the development of robust hands-on modules that allow students to observe features of cell shape and mechanics and connect them to fundamental physics principles. Here, we demonstrate a workflow that engages students in studying epithelial cell mechanics by using commercial histology slides of frog skin. We show that by using recently developed artificial intelligence-based image-segmentation tools, students can easily quantify different cell morphologic features in a high-throughput manner. Using our workflow, students can reproduce 2 essential findings in cell mechanics: the common gamma distribution of normalized cell aspect ratio in jammed epithelia and the constant ratio between the nuclear and cellular area. Importantly, because the only required instrument for this active learning module is a readily available light microscope and a computer, our module is relatively low cost, as well as portable. These features make the module scalable for students at various education levels and outreach programs. This highly accessible education module provides a fun and engaging way to introduce students to the world of epithelial tissue mechanics.
Collapse
Affiliation(s)
- Alexandra Bermudez
- Bioengineering Department, University of California, Los Angeles, Los Angeles, CA, USA
| | - Samanta Negrete Muñoz
- Bioengineering Department, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rita Blaik
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Amy C Rowat
- Bioengineering Department, University of California, Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jimmy Hu
- Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Neil Y C Lin
- Bioengineering Department, University of California, Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA, USA
- Mechanical and Aerospace Engineering Department, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
42
|
Armour EM, Thomas CM, Greco G, Bhatnagar A, Elefant F. Experience-dependent Tip60 nucleocytoplasmic transport is regulated by its NLS/NES sequences for neuroplasticity gene control. Mol Cell Neurosci 2023; 127:103888. [PMID: 37598897 PMCID: PMC11337217 DOI: 10.1016/j.mcn.2023.103888] [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: 06/13/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/22/2023] Open
Abstract
Nucleocytoplasmic transport (NCT) in neurons is critical for enabling proteins to enter the nucleus and regulate plasticity genes in response to environmental cues. Such experience-dependent (ED) neural plasticity is central for establishing memory formation and cognitive function and can influence the severity of neurodegenerative disorders like Alzheimer's disease (AD). ED neural plasticity is driven by histone acetylation (HA) mediated epigenetic mechanisms that regulate dynamic activity-dependent gene transcription profiles in response to neuronal stimulation. Yet, how histone acetyltransferases (HATs) respond to extracellular cues in the in vivo brain to drive HA-mediated activity-dependent gene control remains unclear. We previously demonstrated that extracellular stimulation of rat hippocampal neurons in vitro triggers Tip60 HAT nuclear import with concomitant synaptic gene induction. Here, we focus on investigating Tip60 HAT subcellular localization and NCT specifically in neuronal activity-dependent gene control by using the learning and memory mushroom body (MB) region of the Drosophila brain as a powerful in vivo cognitive model system. We used immunohistochemistry (IHC) to compare the subcellular localization of Tip60 HAT in the Drosophila brain under normal conditions and in response to stimulation of fly brain neurons in vivo either by genetically inducing potassium channels activation or by exposure to natural positive ED conditions. Furthermore, we found that both inducible and ED condition-mediated neural induction triggered Tip60 nuclear import with concomitant induction of previously identified Tip60 target genes and that Tip60 levels in both the nucleus and cytoplasm were significantly decreased in our well-characterized Drosophila AD model. Mutagenesis of a putative nuclear localization signal (NLS) sequence and nuclear export signal (NES) sequence that we identified in the Drosophila Tip60 protein revealed that both are functionally required for appropriate Tip60 subcellular localization. Our results support a model by which neuronal stimulation triggers Tip60 NCT via its NLS and NES sequences to promote induction of activity-dependent neuroplasticity gene transcription and that this process may be disrupted in AD.
Collapse
Affiliation(s)
- Ellen M Armour
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Christina M Thomas
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Gabrielle Greco
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Akanksha Bhatnagar
- Department of Biology, Drexel University, Philadelphia, PA, United States of America
| | - Felice Elefant
- Department of Biology, Drexel University, Philadelphia, PA, United States of America.
| |
Collapse
|
43
|
Zhang Z, Zhu H, Zhao G, Miao Y, Zhao L, Feng J, Zhang H, Miao R, Sun L, Gao B, Zhang W, Wang Z, Zhang J, Zhang Y, Guo H, Xu F, Lu TJ, Genin GM, Lin M. Programmable and Reversible Integrin-Mediated Cell Adhesion Reveals Hysteresis in Actin Kinetics that Alters Subsequent Mechanotransduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302421. [PMID: 37849221 PMCID: PMC10724447 DOI: 10.1002/advs.202302421] [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: 04/17/2023] [Revised: 08/16/2023] [Indexed: 10/19/2023]
Abstract
Dynamically evolving adhesions between cells and extracellular matrix (ECM) transmit time-varying signals that control cytoskeletal dynamics and cell fate. Dynamic cell adhesion and ECM stiffness regulate cellular mechanosensing cooperatively, but it has not previously been possible to characterize their individual effects because of challenges with controlling these factors independently. Therefore, a DNA-driven molecular system is developed wherein the integrin-binding ligand RGD can be reversibly presented and removed to achieve cyclic cell attachment/detachment on substrates of defined stiffness. Using this culture system, it is discovered that cyclic adhesion accelerates F-actin kinetics and nuclear mechanosensing in human mesenchymal stem cells (hMSCs), with the result that hysteresis can completely change how hMSCs transduce ECM stiffness. Results are dramatically different from well-known results for mechanotransduction on static substrates, but are consistent with a mathematical model of F-actin fragments retaining structure following loss of integrin ligation and participating in subsequent repolymerization. These findings suggest that cyclic integrin-mediated adhesion alters the mechanosensing of ECM stiffness by hMSCs through transient, hysteretic memory that is stored in F-actin.
Collapse
Affiliation(s)
- Zheng Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Guoqing Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Yunyi Miao
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Lingzhu Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Jinteng Feng
- Department of Medical OncologyFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710061P. R. China
| | - Huan Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Run Miao
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Lin Sun
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Bin Gao
- Department of EndocrinologySecond Affiliated Hospital of Air Force Military Medical UniversityXi'an710038P. R. China
| | - Wencheng Zhang
- Department of EndocrinologySecond Affiliated Hospital of Air Force Military Medical UniversityXi'an710038P. R. China
| | - Zheng Wang
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710061P. R. China
| | - Jianfang Zhang
- Department of Gynaecology and Obstetrics of Xijing Hospital, Fourth Military Medical University710054Xi'anP. R. China
| | - Ying Zhang
- Xijing 986 Hospital DepartmentFourth Military Medical UniversityXi'an710054P. R. China
| | - Hui Guo
- Department of Medical OncologyFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710061P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical StructuresNanjing University of Aeronautics and AstronauticsNanjing210016P. R. China
| | - Guy M. Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
- Department of Mechanical Engineering & Materials ScienceWashington University in St. LouisSt. LouisMO63130USA
- NSF Science and Technology Center for Engineering MechanobiologyWashington University in St. LouisSt. LouisMO63130USA
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| |
Collapse
|
44
|
Peters MM, Brister JK, Tang EM, Zhang FW, Lucian VM, Trackey PD, Bone Z, Zimmerman JF, Jin Q, Burpo FJ, Parker KK. Self-organizing behaviors of cardiovascular cells on synthetic nanofiber scaffolds. APL Bioeng 2023; 7:046114. [PMID: 38046543 PMCID: PMC10693444 DOI: 10.1063/5.0172423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/16/2023] [Indexed: 12/05/2023] Open
Abstract
In tissues and organs, the extracellular matrix (ECM) helps maintain inter- and intracellular architectures that sustain the structure-function relationships defining physiological homeostasis. Combining fiber scaffolds and cells to form engineered tissues is a means of replicating these relationships. Engineered tissues' fiber scaffolds are designed to mimic the topology and chemical composition of the ECM network. Here, we asked how cells found in the heart compare in their propensity to align their cytoskeleton and self-organize in response to topological cues in fibrous scaffolds. We studied cardiomyocytes, valvular interstitial cells, and vascular endothelial cells as they adapted their inter- and intracellular architectures to the extracellular space. We used focused rotary jet spinning to manufacture aligned fibrous scaffolds to mimic the length scale and three-dimensional (3D) nature of the native ECM in the muscular, valvular, and vascular tissues of the heart. The representative cardiovascular cell types were seeded onto fiber scaffolds and infiltrated the fibrous network. We measured different cell types' propensity for cytoskeletal alignment in response to fiber scaffolds with differing levels of anisotropy. The results indicated that valvular interstitial cells on moderately anisotropic substrates have a higher propensity for cytoskeletal alignment than cardiomyocytes and vascular endothelial cells. However, all cell types displayed similar levels of alignment on more extreme (isotropic and highly anisotropic) fiber scaffold organizations. These data suggest that in the hierarchy of signals that dictate the spatiotemporal organization of a tissue, geometric cues within the ECM and cellular networks may homogenize behaviors across cell populations and demographics.
Collapse
Affiliation(s)
- Michael M. Peters
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - Jackson K. Brister
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - Edward M. Tang
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - Felita W. Zhang
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - Veronica M. Lucian
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - Paul D. Trackey
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - Zachary Bone
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - John F. Zimmerman
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - Qianru Jin
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - F. John Burpo
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | | |
Collapse
|
45
|
Dickinson RB, Lele TP. A new function for nuclear lamins: providing surface tension to the nuclear drop. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 28:100483. [PMID: 38283102 PMCID: PMC10812902 DOI: 10.1016/j.cobme.2023.100483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
The nuclear lamina, a conserved structure in metazoans, provides mechanical rigidity to the nuclear envelope. A decrease in lamin levels and/or lamin mutations are associated with a host of human diseases. Despite being only about 15 nm thick, perturbation of components of the nuclear lamina dramatically impacts the deformation response of the entire nucleus through mechanisms that are not well understood. Here we discuss evidence for the recently proposed 'nuclear drop' model that explains the role of A-type lamins in nuclear deformation in migrating cells. In this model, the nuclear lamina acts as an inextensible surface, supporting a surface tension when fully unfolded, that balances nuclear interior pressure. Much like a liquid drop surface where the molecularly thin interface governs surface tension and drop shape under external forces, the thin nuclear lamina imparts a surface tension on the nuclear drop to resist nuclear deformation as well as to establish nuclear shape. We discuss implications of the nuclear drop model for the function of this crucially important eukaryotic organelle.
Collapse
Affiliation(s)
- Richard B. Dickinson
- Department of Chemical Engineering, University of Florida, 1030 Center Drive, Gainesville, FL, 32611, USA
| | - Tanmay P. Lele
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St., College Station, TX, 77843, USA
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, 3122 TAMU, College Station, TX, 77843, USA
- Department of Translational Medical Sciences, Texas A&M University, 2121 W Holcombe St., Houston, TX, 77030, USA
| |
Collapse
|
46
|
Mei Y, Feng X, Jin Y, Kang R, Wang X, Zhao D, Ghosh S, Neu CP, Avril S. Cell nucleus elastography with the adjoint-based inverse solver. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 242:107827. [PMID: 37801883 DOI: 10.1016/j.cmpb.2023.107827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/09/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND AND OBJECTIVES The mechanics of the nucleus depends on cellular structures and architecture, and impact a number of diseases. Nuclear mechanics is yet rather complex due to heterogeneous distribution of dense heterochromatin and loose euchromatin domains, giving rise to spatially variable stiffness properties. METHODS In this study, we propose to use the adjoint-based inverse solver to identify for the first time the nonhomogeneous elastic property distribution of the nucleus. Inputs of the inverse solver are deformation fields measured with microscopic imaging in contracting cardiomyocytes. RESULTS The feasibility of the proposed method is first demonstrated using simulated data. Results indicate accurate identification of the assumed heterochromatin region, with a maximum relative error of less than 5%. We also investigate the influence of unknown Poisson's ratio on the reconstruction and find that variations of the Poisson's ratio in the range [0.3-0.5] result in uncertainties of less than 15% in the identified stiffness. Finally, we apply the inverse solver on actual deformation fields acquired within the nuclei of two cardiomyocytes. The obtained results are in good agreement with the density maps obtained from microscopy images. CONCLUSIONS Overall, the proposed approach shows great potential for nuclear elastography, with promising value for emerging fields of mechanobiology and mechanogenetics.
Collapse
Affiliation(s)
- Yue Mei
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China; Ningbo Institute of Dalian University of Technology, No. 26 Yucai Road, Jiangbei District, Ningbo 315016, China
| | - Xuan Feng
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - Yun Jin
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - Rongyao Kang
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - XinYu Wang
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - Dongmei Zhao
- State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian 116023, China; International Research Center for Computational Mechanics, Dalian University of Technology, Dalian 116023, China
| | - Soham Ghosh
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States of America
| | - Corey P Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States of America; Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, United States of America; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, United States of America
| | - Stephane Avril
- Mines Saint-Étienne, Univ Jean Monnet, INSERM, U 1059 Sainbiose, F - 42023, Saint-Étienne, France.
| |
Collapse
|
47
|
Beedle AE, Jaganathan A, Albajar-Sigalés A, Yavitt FM, Bera K, Andreu I, Granero-Moya I, Zalvidea D, Kechagia Z, Wiche G, Trepat X, Ivaska J, Anseth KS, Shenoy VB, Roca-Cusachs P. Fibrillar adhesion dynamics govern the timescales of nuclear mechano-response via the vimentin cytoskeleton. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566191. [PMID: 37986921 PMCID: PMC10659263 DOI: 10.1101/2023.11.08.566191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The cell nucleus is continuously exposed to external signals, of both chemical and mechanical nature. To ensure proper cellular response, cells need to regulate not only the transmission of these signals, but also their timing and duration. Such timescale regulation is well described for fluctuating chemical signals, but if and how it applies to mechanical signals reaching the nucleus is still unknown. Here we demonstrate that the formation of fibrillar adhesions locks the nucleus in a mechanically deformed conformation, setting the mechanical response timescale to that of fibrillar adhesion remodelling (~1 hour). This process encompasses both mechanical deformation and associated mechanotransduction (such as via YAP), in response to both increased and decreased mechanical stimulation. The underlying mechanism is the anchoring of the vimentin cytoskeleton to fibrillar adhesions and the extracellular matrix through plectin 1f, which maintains nuclear deformation. Our results reveal a mechanism to regulate the timescale of mechanical adaptation, effectively setting a low pass filter to mechanotransduction.
Collapse
Affiliation(s)
- Amy E.M. Beedle
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Department of Physics, King’s College London, London WC2R 2LS, UK
| | - Anuja Jaganathan
- Center for Engineering Mechanobiology and Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Aina Albajar-Sigalés
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - F. Max Yavitt
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303 USA
| | - Kaustav Bera
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303 USA
| | - Ion Andreu
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, E-48940, Leioa, Spain
- Ikerbasque, Basque Foundation for Science, 48013, Bilbao, Spain
| | - Ignasi Granero-Moya
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Dobryna Zalvidea
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Zanetta Kechagia
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Gerhard Wiche
- Max Perutz Laboratories, Department of Biochemistry and Cell Biology, University of Vienna, 1030 Vienna, Austria
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- University of Barcelona, 08028 Barcelona, Spain
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Life Technologies, University of Turku, FI-20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Foundation for the Finnish Cancer Institute, Tukholmankatu 8, FI-00014 Helsinki, Finland
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303 USA
| | - Vivek B. Shenoy
- Center for Engineering Mechanobiology and Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
48
|
Peng X, Huang Y, Genin GM. The fibrous character of pericellular matrix mediates cell mechanotransduction. JOURNAL OF THE MECHANICS AND PHYSICS OF SOLIDS 2023; 180:105423. [PMID: 38559448 PMCID: PMC10978028 DOI: 10.1016/j.jmps.2023.105423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cells in solid tissues sense and respond to mechanical signals that are transmitted through extracellular matrix (ECM) over distances that are many times their size. This long-range force transmission is known to arise from strain-stiffening and buckling in the collagen fiber ECM network, but must also pass through the denser pericellular matrix (PCM) that cells form by secreting and compacting nearby collagen. However, the role of the PCM in the transmission of mechanical signals is still unclear. We therefore studied an idealized computational model of cells embedded within fibrous collagen ECM and PCM. Our results suggest that the smaller network pore sizes associated with PCM attenuates tension-driven collagen-fiber alignment, undermining long-range force transmission and shielding cells from mechanical stress. However, elongation of the cell body or anisotropic cell contraction can compensate for these effects to enable long distance force transmission. Results are consistent with recent experiments that highlight an effect of PCM on shielding cells from high stresses. Results have implications for the transmission of mechanical signaling in development, wound healing, and fibrosis.
Collapse
Affiliation(s)
- Xiangjun Peng
- U.S. National Science Foundation Science and Technology Center for Engineering Mechanobiology, and Department of Biomedical Engineering, Washington University, St. Louis, MO 63130 United States
| | - Yuxuan Huang
- U.S. National Science Foundation Science and Technology Center for Engineering Mechanobiology, and Department of Biomedical Engineering, Washington University, St. Louis, MO 63130 United States
| | - Guy M. Genin
- U.S. National Science Foundation Science and Technology Center for Engineering Mechanobiology, and Department of Biomedical Engineering, Washington University, St. Louis, MO 63130 United States
| |
Collapse
|
49
|
Wang X, Agrawal V, Dunton CL, Liu Y, Virk RKA, Patel PA, Carter L, Pujadas EM, Li Y, Jain S, Wang H, Ni N, Tsai HM, Rivera-Bolanos N, Frederick J, Roth E, Bleher R, Duan C, Ntziachristos P, He TC, Reid RR, Jiang B, Subramanian H, Backman V, Ameer GA. Chromatin reprogramming and bone regeneration in vitro and in vivo via the microtopography-induced constriction of cell nuclei. Nat Biomed Eng 2023; 7:1514-1529. [PMID: 37308586 PMCID: PMC10804399 DOI: 10.1038/s41551-023-01053-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/10/2023] [Indexed: 06/14/2023]
Abstract
Topographical cues on cells can, through contact guidance, alter cellular plasticity and accelerate the regeneration of cultured tissue. Here we show how changes in the nuclear and cellular morphologies of human mesenchymal stromal cells induced by micropillar patterns via contact guidance influence the conformation of the cells' chromatin and their osteogenic differentiation in vitro and in vivo. The micropillars impacted nuclear architecture, lamin A/C multimerization and 3D chromatin conformation, and the ensuing transcriptional reprogramming enhanced the cells' responsiveness to osteogenic differentiation factors and decreased their plasticity and off-target differentiation. In mice with critical-size cranial defects, implants with micropillar patterns inducing nuclear constriction altered the cells' chromatin conformation and enhanced bone regeneration without the need for exogenous signalling molecules. Our findings suggest that medical device topographies could be designed to facilitate bone regeneration via chromatin reprogramming.
Collapse
Affiliation(s)
- Xinlong Wang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Vasundhara Agrawal
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Cody L Dunton
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Yugang Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Ranya K A Virk
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Priyam A Patel
- Quantitative Data Science Core, Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lucas Carter
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Emily M Pujadas
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Yue Li
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Surbhi Jain
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Hsiu-Ming Tsai
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | - Nancy Rivera-Bolanos
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Jane Frederick
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Eric Roth
- Department of Materials Sciences and Engineering, Northwestern University, Evanston, IL, USA
| | - Reiner Bleher
- Department of Materials Sciences and Engineering, Northwestern University, Evanston, IL, USA
| | - Chongwen Duan
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Panagiotis Ntziachristos
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
- Simpson Querrey Center for Epigenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Tong Chuan He
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Russell R Reid
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL, USA
| | - Bin Jiang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hariharan Subramanian
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA
| | - Vadim Backman
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| | - Guillermo A Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Center for Physical Genomics and Engineering, Northwestern University, Evanston, IL, USA.
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Chemistry of Life Process Institute, Northwestern University, Chicago, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
50
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|