1
|
Metkari AS, Witt RL, Cognetti DM, Dhong C, Jia X. Promoting Polarization and Differentiation of Primary Human Salivary Gland Stem/Progenitor Cells in Protease-Degradable Hydrogels via ROCK Inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.24.625065. [PMID: 39651209 PMCID: PMC11623551 DOI: 10.1101/2024.11.24.625065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Towards the goal of in vitro engineering of functional salivary gland tissues, we cultured primary human salivary stem/progenitor cells (hS/PCs) in hyaluronic acid-based matrices with varying percentages of proteolytically degradable crosslinks in the presence of Rho kinase (ROCK) inhibitor. Single cells encapsulated in the hydrogel grew into organized multicellular structures by day 15, and over 60% of the structures developed in the non-degradable and 50% degradable hydrogels contained a central lumen. Importantly, ROCK inhibition led to the establishment of multicellular structures that were correctly polarized, as evidenced by apical localization of a Golgi marker GM130, apical/lateral localization of tight junction protein zonula occludens-1 (ZO-1), and basal localization of integrin β1 and basement membrane proteins laminin α1 and collagen IV. Cultures maintained in 50% degradable gels with ROCK inhibition exhibited an increased expression of acinar markers AQP5 and SLC12A2 (at the transcript level) and AQP5 and NKCC1 (at the protein level) as compared to those without ROCK inhibition. Upon stimulation with isoproterenol, α-amylase secretion into the lumen was observed. Particle-tracking microrheology was employed to analyze the stiffness of cells using mitochondria as the passive tracer particles. Our results indicated that cells grown in 100% degradable gels were stiffer than those maintained in non-degradable gels, and cells cultured with the ROCK inhibitor were softer than those maintained without the inhibitor. We conclude that reducing cellular contractility via ROCK inhibition while retaining some degree of matrix confinement promotes the establishment of multicellular structures containing pro-acinar cells with correct apicobasal polarization.
Collapse
|
2
|
Lee SH, Li Z, Zhang EY, Kim DH, Huang Z, Lee SJ, Kang HW, Burdick JA, Mauck RL, Heo SC. Precision Repair of Zone-Specific Meniscal Injuries Using a Tunable Extracellular Matrix-Based Hydrogel System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612723. [PMID: 39345590 PMCID: PMC11429709 DOI: 10.1101/2024.09.12.612723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Meniscus injuries present significant therapeutic challenges due to their limited self-healing capacity and diverse biological and mechanical properties across meniscal tissue. Conventional repair strategies neglect to replicate the complex zonal characteristics within the meniscus, resulting in suboptimal outcomes. In this study, we introduce an innovative, age- and stiffness-tunable meniscus decellularized extracellular matrix (DEM)-based hydrogel system designed for precision repair of heterogeneous, zonal-dependent meniscus injuries. By synthesizing age-dependent DEM hydrogels, we identified distinct cellular responses: fetal bovine meniscus-derived DEM promoted chondrogenic differentiation, while adult meniscus-derived DEM supported fibrochondrogenic phenotypes. The incorporation of methacrylate hyaluronic acid (MeHA) further refined the mechanical properties and injectability of the DEM-based hydrogels. The combination of age-dependent DEM with MeHA allowed for precise stiffness tuning, influencing cell differentiation and closely mimicking native tissue environments. In vivo tests confirmed the biocompatibility of hydrogels and their integration with native meniscus tissues. Furthermore, advanced 3D bioprinting techniques enabled the fabrication of hybrid hydrogels with biomaterial and mechanical gradients, effectively emulating the zonal properties of meniscus tissue and enhancing cell integration. This study represents a significant advancement in meniscus tissue engineering, providing a promising platform for customized regenerative therapies across a range of heterogeneous fibrous connective tissues.
Collapse
Affiliation(s)
- Se-Hwan Lee
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Zizhao Li
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Ellen Y. Zhang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Dong Hwa Kim
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Ziqi Huang
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| | - Sang Jin Lee
- Biofunctional Materials, Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, PR China
| | - Hyun-Wook Kang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Jason A. Burdick
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, United States
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, United States
- Translational Musculoskeletal Research Center, Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA 19104, United States
| | - Su Chin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, United States
- Translational Musculoskeletal Research Center, Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA 19104, United States
| |
Collapse
|
3
|
Yang Y, Han K, Huang S, Wang K, Wang Y, Ding S, Zhang L, Zhang M, Xu B, Ma S, Wang Y, Wu S, Wang X. Revelation of adhesive proteins affecting cellular contractility through reference-free traction force microscopy. J Mater Chem B 2024; 12:3249-3261. [PMID: 38466580 DOI: 10.1039/d4tb00065j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Over the past few decades, the critical role played by cellular contractility associated mechanotransduction in the regulation of cell functions has been revealed. In this case, numerous biomaterials have been chemically or structurally designed to manipulate cell behaviors through the regulation of cellular contractility. In particular, adhesive proteins including fibronectin, poly-L-lysine and collagen type I have been widely applied in various biomaterials to improve cell adhesion. Therefore, clarifying the effects of adhesive proteins on cellular contractility has been valuable for the development of biomaterial design. In this study, reference-free traction force microscopy with a well-organized microdot array was designed and prepared to investigate the relationship between adhesive proteins, cellular contractility, and mechanotransduction. The results showed that fibronectin and collagen type I were able to promote the assembly of focal adhesions and further enhance cellular contraction and YAP activity. In contrast, although poly-L-lysine supported cell spreading and elongation, it was inefficient at inducing cell contractility and activating YAP. Additionally, compared with cellular morphogenesis, cellular contraction was essential for YAP activation.
Collapse
Affiliation(s)
- Yingjun Yang
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, P. R. China
| | - Kuankuan Han
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
| | - Siyuan Huang
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, China
| | - Kai Wang
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
| | - Yuchen Wang
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
| | - Shukai Ding
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
| | - Le Zhang
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
| | - Miao Zhang
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
| | - Bingshe Xu
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, P. R. China
- Laboratory of Interface Science and Engineering in Advanced Materials, Taiyuan University of Technology, Taiyuan, China
| | - Shufang Ma
- Materials Institute of Atomic and Molecular Science, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, P. R. China
| | - Yongtao Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China.
| | - Shengli Wu
- Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Xinlong Wang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
4
|
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
|
5
|
Xiong X, Huo Q, Li K, Cui C, Chang C, Park C, Ku B, Hong CS, Lim H, Pandya PH, Saadatzadeh MR, Bijangi-Vishehsaraei K, Lin CC, Kacena MA, Pollok KE, Chen A, Liu J, Thompson WR, Li XL, Li BY, Yokota H. Enhancing anti-tumor potential: low-intensity vibration suppresses osteosarcoma progression and augments MSCs' tumor-suppressive abilities. Theranostics 2024; 14:1430-1449. [PMID: 38389836 PMCID: PMC10879868 DOI: 10.7150/thno.90945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: Osteosarcoma (OS), a common malignant bone tumor, calls for the investigation of novel treatment strategies. Low-intensity vibration (LIV) presents itself as a promising option, given its potential to enhance bone health and decrease cancer susceptibility. This research delves into the effects of LIV on OS cells and mesenchymal stem cells (MSCs), with a primary focus on generating induced tumor-suppressing cells (iTSCs) and tumor-suppressive conditioned medium (CM). Methods: To ascertain the influence of vibration frequency, we employed numerical simulations and conducted experiments to determine the most effective LIV conditions. Subsequently, we generated iTSCs and CM through LIV exposure and assessed the impact of CM on OS cells. We also explored the underlying mechanisms of the tumor-suppressive effects of LIV-treated MSC CM, with a specific focus on vinculin (VCL). We employed cytokine array, RNA sequencing, and Western blot techniques to investigate alterations in cytokine profiles, transcriptomes, and tumor suppressor proteins. Results: Numerical simulations validated LIV frequencies within the 10-100 Hz range. LIV induced notable morphological changes in OS cells and MSCs, confirming its dual role in inhibiting OS cell progression and promoting MSC conversion into iTSCs. Upregulated VCL expression enhanced MSC responsiveness to LIV, significantly bolstering CM's efficacy. Notably, we identified tumor suppressor proteins in LIV-treated CM, including procollagen C endopeptidase enhancer (PCOLCE), histone H4 (H4), peptidylprolyl isomerase B (PPIB), and aldolase A (ALDOA). Consistently, cytokine levels decreased significantly in LIV-treated mouse femurs, and oncogenic transcript levels were downregulated in LIV-treated OS cells. Moreover, our study demonstrated that combining LIV-treated MSC CM with chemotherapy drugs yielded additive anti-tumor effects. Conclusions: LIV effectively impeded the progression of OS cells and facilitated the transformation of MSCs into iTSCs. Notably, iTSC-derived CM demonstrated robust anti-tumor properties and the augmentation of MSC responsiveness to LIV via VCL. Furthermore, the enrichment of tumor suppressor proteins within LIV-treated MSC CM and the reduction of cytokines within LIV-treated isolated bone underscore the pivotal tumor-suppressive role of LIV within the bone tumor microenvironment.
Collapse
Affiliation(s)
- Xue Xiong
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Qingji Huo
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Kexin Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Changpeng Cui
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Chunyi Chang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Charles Park
- Department of Physics, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - BonHeon Ku
- Department of Mechanical Engineering, Pusan National University, Busan 46241, Korea
| | - Chin-Suk Hong
- Department of Mechanical Engineering, Ulsan College, Ulsan 44022, Korea
| | - HeeChang Lim
- Department of Mechanical Engineering, Pusan National University, Busan 46241, Korea
| | - Pankita H. Pandya
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine; Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine; Indianapolis, IN 46202, USA
| | - M. Reza Saadatzadeh
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine; Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine; Indianapolis, IN 46202, USA
| | | | - Chien-Chi Lin
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine; Indianapolis, IN 46202, USA
| | - Melissa A. Kacena
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine; Indianapolis, IN 46202, USA
- Department of Orthopaedic Surgery, Indiana University School of Medicine; Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine; Indianapolis, IN 46202, USA
| | - Karen E. Pollok
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine; Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine; Indianapolis, IN 46202, USA
| | - Andy Chen
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Jing Liu
- Department of Physics, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine; Indianapolis, IN 46202, USA
| | - William R. Thompson
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine; Indianapolis, IN 46202, USA
- Department of Physical Therapy, Indiana University, Indianapolis, IN 46202, USA
| | - Xue-Lian Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine; Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine; Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Wernlé K, Thiel CS, Ullrich O. Increased H3K9me3 and F-Actin Reorganization in the Rapid Adaptive Response to Hypergravity in Human T Lymphocytes. Int J Mol Sci 2023; 24:17232. [PMID: 38139061 PMCID: PMC10743231 DOI: 10.3390/ijms242417232] [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: 10/31/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Our study explored the impact of hypergravity on human T cells, which experience additional acceleration forces beyond Earth's gravity due to various factors, such as pulsatile blood flow, and technology, such as high-performance aircraft flights or spaceflights. We investigated the histone modifications Histone 3 lysine 4 and 9 trimethylation (H3K4me3 and H3K9me3, respectively), as well as the structural and cytoskeletal organization of Jurkat T cells in response to hypergravity. Histone modifications play a crucial role in gene regulation, chromatin organization and DNA repair. In response to hypergravity, we found only minimal changes of H3K4me3 and a rapid increase in H3K9me3, which was sustained for up to 15 min and then returned to control levels after 1 h. Furthermore, rapid changes in F-actin fluorescence were observed within seconds of hypergravity exposure, indicating filament depolymerization and cytoskeletal restructuring, which subsequently recovered after 1 h of hypergravity. Our study demonstrated the rapid, dynamic and adaptive cellular response to hypergravity, particularly in terms of histone modifications and cytoskeletal changes. These responses are likely necessary for maintaining genome stability and structural integrity under hypergravity conditions as they are constantly occurring in the human body during blood cell circulation.
Collapse
Affiliation(s)
- Kendra Wernlé
- Institute of Anatomy, Faculty of Medicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland;
- Faculty of Medical Sciences, Private University of the Principality of Liechtenstein (UFL), Dorfstrasse 24, 9495 Triesen, Liechtenstein
| | - Cora S. Thiel
- Institute of Anatomy, Faculty of Medicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland;
- Institute of Machine Design, Otto-von-Guericke-University Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
- Space Life Sciences Laboratory (SLSL), Kennedy Space Center, 505 Odyssey Way, Exploration Park, Merritt Island, FL 32953, USA
- UZH Space Hub, Air Force Center, Air Base Dübendorf, Überlandstrasse 270, 8600 Dubendorf, Switzerland
| | - Oliver Ullrich
- Institute of Anatomy, Faculty of Medicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland;
- Institute of Machine Design, Otto-von-Guericke-University Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
- Space Life Sciences Laboratory (SLSL), Kennedy Space Center, 505 Odyssey Way, Exploration Park, Merritt Island, FL 32953, USA
- UZH Space Hub, Air Force Center, Air Base Dübendorf, Überlandstrasse 270, 8600 Dubendorf, Switzerland
- Department of Industrial Engineering, Ernst-Abbe-Hochschule (EAH) Jena, Carl-Zeiss-Promenade 2, 07745 Jena, Germany
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
7
|
de Lope-Planelles A, González-Novo R, Madrazo E, Peralta-Carrero G, Cruz Rodríguez MP, Zamora-Carreras H, Torrano V, López-Menéndez H, Roda-Navarro P, Monroy F, Redondo-Muñoz J. Mechanical stress confers nuclear and functional changes in derived leukemia cells from persistent confined migration. Cell Mol Life Sci 2023; 80:316. [PMID: 37801090 PMCID: PMC10558412 DOI: 10.1007/s00018-023-04968-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/07/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023]
Abstract
Nuclear deformability plays a critical role in cell migration. During this process, the remodeling of internal components of the nucleus has a direct impact on DNA damage and cell behavior; however, how persistent migration promotes nuclear changes leading to phenotypical and functional consequences remains poorly understood. Here, we described that the persistent migration through physical barriers was sufficient to promote permanent modifications in migratory-altered cells. We found that derived cells from confined migration showed changes in lamin B1 localization, cell morphology and transcription. Further analysis confirmed that migratory-altered cells showed functional differences in DNA repair, cell response to chemotherapy and cell migration in vivo homing experiments. Experimental modulation of actin polymerization affected the redistribution of lamin B1, and the basal levels of DNA damage in migratory-altered cells. Finally, since major nuclear changes were present in migratory-altered cells, we applied a multidisciplinary biochemical and biophysical approach to identify that confined conditions promoted a different biomechanical response of the nucleus in migratory-altered cells. Our observations suggest that mechanical compression during persistent cell migration has a role in stable nuclear and genomic alterations that might handle the genetic instability and cellular heterogeneity in aging diseases and cancer.
Collapse
Affiliation(s)
- Ana de Lope-Planelles
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Raquel González-Novo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Elena Madrazo
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Gracia Peralta-Carrero
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - María Pilar Cruz Rodríguez
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Héctor Zamora-Carreras
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain
| | - Verónica Torrano
- Department of Biochemistry and Molecular Biology, University of the Basque Country, Leioa, Spain
| | - Horacio López-Menéndez
- Department of Physical Chemistry, Complutense University, Madrid, Spain
- Translational Biophysics, Hospital Doce de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Pedro Roda-Navarro
- Department of Immunology, School of Medicine, University Complutense de Madrid and 12 de Octubre Health Research Institute (Imas12) Madrid, Madrid, Spain
| | - Francisco Monroy
- Department of Physical Chemistry, Complutense University, Madrid, Spain
- Translational Biophysics, Hospital Doce de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Javier Redondo-Muñoz
- Department of Molecular Medicine, Centro de Investigaciones Biológicas Margarita Salas (CIB Margarita Salas-CSIC), Madrid, Spain.
| |
Collapse
|
8
|
Scott AK, Rafuse M, Neu CP. Mechanically induced alterations in chromatin architecture guide the balance between cell plasticity and mechanical memory. Front Cell Dev Biol 2023; 11:1084759. [PMID: 37143893 PMCID: PMC10151697 DOI: 10.3389/fcell.2023.1084759] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 04/07/2023] [Indexed: 05/06/2023] Open
Abstract
Phenotypic plasticity, or adaptability, of a cell determines its ability to survive and function within changing cellular environments. Changes in the mechanical environment, ranging from stiffness of the extracellular matrix (ECM) to physical stress such as tension, compression, and shear, are critical environmental cues that influence phenotypic plasticity and stability. Furthermore, an exposure to a prior mechanical signal has been demonstrated to play a fundamental role in modulating phenotypic changes that persist even after the mechanical stimulus is removed, creating stable mechanical memories. In this mini review, our objective is to highlight how the mechanical environment alters both phenotypic plasticity and stable memories through changes in chromatin architecture, mainly focusing on examples in cardiac tissue. We first explore how cell phenotypic plasticity is modulated in response to changes in the mechanical environment, and then connect the changes in phenotypic plasticity to changes in chromatin architecture that reflect short-term and long-term memories. Finally, we discuss how elucidating the mechanisms behind mechanically induced chromatin architecture that lead to cell adaptations and retention of stable mechanical memories could uncover treatment methods to prevent mal-adaptive permanent disease states.
Collapse
Affiliation(s)
- Adrienne K. Scott
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
| | - Michael Rafuse
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
| | - Corey P. Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, United States
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, United States
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
9
|
Amiad Pavlov D, Unnikannan CP, Lorber D, Bajpai G, Olender T, Stoops E, Reuveny A, Safran S, Volk T. The LINC Complex Inhibits Excessive Chromatin Repression. Cells 2023; 12:932. [PMID: 36980273 PMCID: PMC10047284 DOI: 10.3390/cells12060932] [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: 01/16/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
The Linker of Nucleoskeleton and Cytoskeleton (LINC) complex transduces nuclear mechanical inputs suggested to control chromatin organization and gene expression; however, the underlying mechanism is currently unclear. We show here that the LINC complex is needed to minimize chromatin repression in muscle tissue, where the nuclei are exposed to significant mechanical inputs during muscle contraction. To this end, the genomic binding profiles of Polycomb, Heterochromatin Protein1 (HP1a) repressors, and of RNA-Pol II were studied in Drosophila larval muscles lacking functional LINC complex. A significant increase in the binding of Polycomb and parallel reduction of RNA-Pol-II binding to a set of muscle genes was observed. Consistently, enhanced tri-methylated H3K9 and H3K27 repressive modifications and reduced chromatin activation by H3K9 acetylation were found. Furthermore, larger tri-methylated H3K27me3 repressive clusters, and chromatin redistribution from the nuclear periphery towards nuclear center, were detected in live LINC mutant larval muscles. Computer simulation indicated that the observed dissociation of the chromatin from the nuclear envelope promotes growth of tri-methylated H3K27 repressive clusters. Thus, we suggest that by promoting chromatin-nuclear envelope binding, the LINC complex restricts the size of repressive H3K27 tri-methylated clusters, thereby limiting the binding of Polycomb transcription repressor, directing robust transcription in muscle fibers.
Collapse
Affiliation(s)
- Daria Amiad Pavlov
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | | | - Dana Lorber
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Gaurav Bajpai
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elizabeth Stoops
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Adriana Reuveny
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Samuel Safran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Talila Volk
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
10
|
Deng R, Kang R, Jin X, Wang Z, Liu X, Wang Q, Xie L. Mechanical stimulation promotes MSCs healing the lesion of intervertebral disc annulus fibrosus. Front Bioeng Biotechnol 2023; 11:1137199. [PMID: 36845186 PMCID: PMC9950411 DOI: 10.3389/fbioe.2023.1137199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/26/2023] [Indexed: 02/12/2023] Open
Abstract
Mesenchymal stem cells (MSCs) and scaffolds offer promising perspectives for annulus fibrosus (AF) repair. The repair effect was linked to features of the local mechanical environment related to the differentiation of MSCs. In this study, we established a Fibrinogen-Thrombin-Genipin (Fib-T-G) gel which is sticky and could transfer strain force from AF tissue to the human mesenchymal stem cells (hMSCs) embedded in the gel. After the Fib-T-G biological gel was injected into the AF fissures, the histology scores of intervertebral disc (IVD) and AF tissue showed that Fib-T-G gel could better repair the AF fissure in caudal IVD of rats, and increase the expression of AF-related proteins including Collagen 1 (COL1), Collagen 2 (COL2) as well as mechanotransduction-related proteins including RhoA and ROCK1. To clarify the mechanism that sticky Fib-T-G gel induces the healing of AF fissures and the differentiation of hMSCs, we further investigated the differentiation of hMSCs under mechanical strain in vitro. It was demonstrated that both AF-specific genes, including Mohawk and SOX-9, and ECM markers (COL1, COL2, aggrecan) of hMSCs were up-regulated in the environment of strain force. Moreover, RhoA/ROCK1 proteins were also found to be significantly up-regulated. In addition, we further -demonstrated that the fibrochondroinductive effect of the mechanical microenvironment process could be significantly blocked or up-regulated by inhibiting the RhoA/ROCK1 pathway or overexpressing RhoA in MSCs, respectively. Summarily, this study will provide a therapeutic alternative to repair AF tears and provide evidence that RhoA/ROCK1 is vital for hMSCs response to mechanical strain and AF-like differentiation.
Collapse
Affiliation(s)
- Rongrong Deng
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ran Kang
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China,Department of Orthopedics, Nanjing Lishui Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China,*Correspondence: Ran Kang, ; Xin Liu, ; Lin Xie,
| | - Xiaoyu Jin
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China,Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Zihan Wang
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xin Liu
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China,*Correspondence: Ran Kang, ; Xin Liu, ; Lin Xie,
| | - Qing Wang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lin Xie
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China,*Correspondence: Ran Kang, ; Xin Liu, ; Lin Xie,
| |
Collapse
|
11
|
Heo SJ, Thakur S, Chen X, Loebel C, Xia B, McBeath R, Burdick JA, Shenoy VB, Mauck RL, Lakadamyali M. Aberrant chromatin reorganization in cells from diseased fibrous connective tissue in response to altered chemomechanical cues. Nat Biomed Eng 2023; 7:177-191. [PMID: 35996026 PMCID: PMC10053755 DOI: 10.1038/s41551-022-00910-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 06/14/2022] [Indexed: 11/09/2022]
Abstract
Changes in the micro-environment of fibrous connective tissue can lead to alterations in the phenotypes of tissue-resident cells, yet the underlying mechanisms are poorly understood. Here, by visualizing the dynamics of histone spatial reorganization in tenocytes and mesenchymal stromal cells from fibrous tissue of human donors via super-resolution microscopy, we show that physiological and pathological chemomechanical cues can directly regulate the spatial nanoscale organization and density of chromatin in these tissue-resident cell populations. Specifically, changes in substrate stiffness, altered oxygen tension and the presence of inflammatory signals drive chromatin relocalization and compaction into the nuclear boundary, mediated by the activity of the histone methyltransferase EZH2 and an intact cytoskeleton. In healthy cells, chemomechanically triggered changes in the spatial organization and density of chromatin are reversible and can be attenuated by dynamically stiffening the substrate. In diseased human cells, however, the link between mechanical or chemical inputs and chromatin remodelling is abrogated. Our findings suggest that aberrant chromatin organization in fibrous connective tissue may be a hallmark of disease progression that could be leveraged for therapeutic intervention.
Collapse
Affiliation(s)
- Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Shreyasi Thakur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xingyu Chen
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Claudia Loebel
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Boao Xia
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Rowena McBeath
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jason A Burdick
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Vivek B Shenoy
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA.
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 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.
- Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Markov P, Zhu H, Boote C, Blain EJ. Delayed reorganisation of F-actin cytoskeleton and reversible chromatin condensation in scleral fibroblasts under simulated pathological strain. Biochem Biophys Rep 2022; 32:101338. [PMID: 36123992 PMCID: PMC9482111 DOI: 10.1016/j.bbrep.2022.101338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/06/2022] Open
Abstract
Mechanical loading regulates the functional capabilities of the ocular system, particularly in the sclera (‘white of the eye’) – the principal load-bearing tissue of the ocular globe. Resident fibroblasts of the scleral eye wall are continuously subjected to fluctuating mechanical strains arising from eye movements, cerebrospinal fluid pressure and, most influentially, intra-ocular pressure (IOP). Whilst fibroblasts are hypothesised to actively participate in scleral biomechanics, to date limited information has been reported on how the macroscopic stresses and strains are transmitted via their cytoskeletal networks. In this study, the effect of applying either a ‘physiological load’ (simulating healthy IOP) or a ‘pathological load’ (simulating an elevated glaucomatous IOP) to bovine scleral fibroblasts, as a model of human glaucoma, was conducted to characterise cytoskeletal organisation, chromatin condensation and cell dimensions using immunofluorescence confocal microscopy. Quantification of cell parameters and cytoskeletal element anisotropy were subsequently performed using FibrilTool, and chromatin condensation parameter assessment through a bespoke MATLAB script. The novel findings suggest that physiological load-induced F-actin rearrangement is transient, whereas pathological load, recapitulating in vivo glaucomatous IOP levels, had a reversible and inhibitory influence on remodelling of the cytoskeletal architecture and, further, induction of chromatin condensation. Ultimately, this could compromise cell behaviour. These findings could provide valuable insight into the mechanism(s) used by scleral fibroblasts to mechanically adapt to support biomechanical tissue integrity, and how it could be potentially modified for therapeutic avenues targeting mechanically mediated ocular pathologies such as glaucoma. Physiological strain induced a transient F-actin rearrangement in scleral fibroblasts. In contrast, pathological strain reversibly delayed F-actin rearrangement. Vimentin and β-tubulin networks were largely unaffected by strain regimens. Pathological strain reversibly increased chromatin condensation parameter. Pathological strain may induce ‘inhibition delay’ to confer cytoprotection.
Collapse
|
13
|
Kalukula Y, Stephens AD, Lammerding J, Gabriele S. Mechanics and functional consequences of nuclear deformations. Nat Rev Mol Cell Biol 2022; 23:583-602. [PMID: 35513718 PMCID: PMC9902167 DOI: 10.1038/s41580-022-00480-z] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 02/08/2023]
Abstract
As the home of cellular genetic information, the nucleus has a critical role in determining cell fate and function in response to various signals and stimuli. In addition to biochemical inputs, the nucleus is constantly exposed to intrinsic and extrinsic mechanical forces that trigger dynamic changes in nuclear structure and morphology. Emerging data suggest that the physical deformation of the nucleus modulates many cellular and nuclear functions. These functions have long been considered to be downstream of cytoplasmic signalling pathways and dictated by gene expression. In this Review, we discuss an emerging perspective on the mechanoregulation of the nucleus that considers the physical connections from chromatin to nuclear lamina and cytoskeletal filaments as a single mechanical unit. We describe key mechanisms of nuclear deformations in time and space and provide a critical review of the structural and functional adaptive responses of the nucleus to deformations. We then consider the contribution of nuclear deformations to the regulation of important cellular functions, including muscle contraction, cell migration and human disease pathogenesis. Collectively, these emerging insights shed new light on the dynamics of nuclear deformations and their roles in cellular mechanobiology.
Collapse
Affiliation(s)
- Yohalie Kalukula
- University of Mons, Soft Matter and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, CIRMAP, Place du Parc, 20 B-7000 Mons, Belgium
| | - Andrew D. Stephens
- Biology Department, University of Massachusetts Amherst, Amherst, MA, USA
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Sylvain Gabriele
- University of Mons, Soft Matter and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, CIRMAP, Place du Parc, 20 B-7000 Mons, Belgium
| |
Collapse
|
14
|
Dupont S, Wickström SA. Mechanical regulation of chromatin and transcription. Nat Rev Genet 2022; 23:624-643. [DOI: 10.1038/s41576-022-00493-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 01/14/2023]
|
15
|
Ghosh S, Scott AK, Seelbinder B, Barthold JE, Martin BMS, Kaonis S, Schneider SE, Henderson JT, Neu CP. Dedifferentiation alters chondrocyte nuclear mechanics during in vitro culture and expansion. Biophys J 2022; 121:131-141. [PMID: 34800469 PMCID: PMC8758405 DOI: 10.1016/j.bpj.2021.11.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 08/23/2021] [Accepted: 11/10/2021] [Indexed: 01/07/2023] Open
Abstract
The biophysical features of a cell can provide global insights into diverse molecular changes, especially in processes like the dedifferentiation of chondrocytes. Key biophysical markers of chondrocyte dedifferentiation include flattened cellular morphology and increased stress-fiber formation. During cartilage regeneration procedures, dedifferentiation of chondrocytes during in vitro expansion presents a critical limitation to the successful repair of cartilage tissue. Our study investigates how biophysical changes of chondrocytes during dedifferentiation influence the nuclear mechanics and gene expression of structural proteins located at the nuclear envelope. Through an experimental model of cell stretching and a detailed spatial intranuclear strain quantification, we identified that strain is amplified and the distribution of strain within the chromatin is altered under tensile loading in the dedifferentiated state. Further, using a confocal microscopy image-based finite element model and simulation of cell stretching, we found that the cell shape is the primary determinant of the strain amplification inside the chondrocyte nucleus in the dedifferentiated state. Additionally, we found that nuclear envelope proteins have lower gene expression in the dedifferentiated state. This study highlights the role of cell shape in nuclear mechanics and lays the groundwork to design biophysical strategies for the maintenance and enhancement of the chondrocyte phenotype during cell expansion with a goal of successful cartilage tissue engineering.
Collapse
Affiliation(s)
- Soham Ghosh
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO; School of Biomedical Engineering, Colorado State University, Fort Collins, CO; Translational Medicine Institute, Colorado State University, Fort Collins, CO.
| | - Adrienne K Scott
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Benjamin Seelbinder
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Jeanne E Barthold
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Brittany M St Martin
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Samantha Kaonis
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO; Translational Medicine Institute, Colorado State University, Fort Collins, CO
| | - Stephanie E Schneider
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | | | - Corey P Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO; Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO; BioFrontiers Institute, University of Colorado Boulder, Boulder, CO
| |
Collapse
|
16
|
Price CC, Mathur J, Boerckel JD, Pathak A, Shenoy VB. Dynamic self-reinforcement of gene expression determines acquisition of cellular mechanical memory. Biophys J 2021; 120:5074-5089. [PMID: 34627766 PMCID: PMC8633715 DOI: 10.1016/j.bpj.2021.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/17/2021] [Accepted: 10/05/2021] [Indexed: 01/26/2023] Open
Abstract
Mechanotransduction describes activation of gene expression by changes in the cell's physical microenvironment. Recent experiments show that mechanotransduction can lead to long-term "mechanical memory," in which cells cultured on stiff substrates for sufficient time (priming phase) maintain altered phenotype after switching to soft substrates (dissipation phase) as compared to unprimed controls. The timescale of memory acquisition and retention is orders of magnitude larger than the timescale of mechanosensitive cellular signaling, and memory retention time changes continuously with priming time. We develop a model that captures these features by accounting for positive reinforcement in mechanical signaling. The sensitivity of reinforcement represents the dynamic transcriptional state of the cell composed of protein lifetimes and three-dimensional chromatin organization. Our model provides a single framework connecting microenvironment mechanical history to cellular outcomes ranging from no memory to terminal differentiation. Predicting cellular memory of environmental changes can help engineer cellular dynamics through changes in culture environments.
Collapse
Affiliation(s)
- Christopher C Price
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jairaj Mathur
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri
| | - Joel D Boerckel
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amit Pathak
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri.
| | - Vivek B Shenoy
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
17
|
McDonough RC, Price C. Targeted Activation of GPCR-Mediated Ca 2+ Signaling Drives Enhanced Cartilage-Like Matrix Formation. Tissue Eng Part A 2021; 28:405-419. [PMID: 34693731 PMCID: PMC9271335 DOI: 10.1089/ten.tea.2021.0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Intracellular calcium ([Ca2+]i) signaling is a critical regulator of chondrogenesis, chondrocyte differentiation, and cartilage development. Calcium (Ca2+) signaling is known to direct processes that govern chondrocyte gene expression, protein synthesis, cytoskeletal remodeling, and cell fate. Control of chondrocyte/chondroprogenitor Ca2+ signaling has been attempted through mechanical and/or pharmacological activation of endogenous Ca2+ signaling transducers; however, such approaches can lack specificity and/or precision regarding Ca2+ activation mechanisms. Synthetic signaling platforms permitting precise and selective Ca2+ signal transduction can improve dissection of the roles that [Ca2+]i signaling play in chondrocyte behavior. One such platform is the chemogenetic hM3Dq DREADD (designer receptor exclusively activated by designer drugs) that activates [Ca2+]i signaling via the Gαq-PLCβ-IP3-ER pathway upon clozapine N-oxide (CNO) administration. We previously demonstrated hM3Dq's ability to precisely and synthetically initiate robust [Ca2+]i transients and oscillatory [Ca2+]i signaling in chondrocyte-like ATDC5 cells. Here, we investigate the effects that long-term CNO stimulatory culture have on hM3Dq [Ca2+]i signaling dynamics, proliferation, and protein deposition in 2D ATDC5 cultures. Long-term culturing under repeated CNO stimulation modified the temporal dynamics of hM3Dq [Ca2+]i signaling, increased cell proliferation, and enhanced matrix production in a CNO dose- and frequency-dependent manner, and triggered the formation of cell condensations that developed aligned, anisotropic neotissue structures rich in cartilaginous proteoglycans and collagens, all in the absence of differentiation inducers. This study demonstrated Gαq-GPCR-mediated [Ca2+]i signaling involvement in chondroprogenitor proliferation and cartilage-like matrix production, and established hM3Dq as a powerful tool for elucidating the role of GPCR-mediated Ca2+ signaling in chondrogenesis and chondrocyte differentiation.
Collapse
Affiliation(s)
- Ryan C McDonough
- University of Delaware, 5972, Biomedical Engineering, 161 Colburn Lab, Newark, Delaware, United States, 19716-5600;
| | - Christopher Price
- University of Delaware, 5972, Biomedical Engineering, Newark, Delaware, United States;
| |
Collapse
|
18
|
Fibroblast Memory in Development, Homeostasis and Disease. Cells 2021; 10:cells10112840. [PMID: 34831065 PMCID: PMC8616330 DOI: 10.3390/cells10112840] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023] Open
Abstract
Fibroblasts are the major cell population in the connective tissue of most organs, where they are essential for their structural integrity. They are best known for their role in remodelling the extracellular matrix, however more recently they have been recognised as a functionally highly diverse cell population that constantly responds and adapts to their environment. Biological memory is the process of a sustained altered cellular state and functions in response to a transient or persistent environmental stimulus. While it is well established that fibroblasts retain a memory of their anatomical location, how other environmental stimuli influence fibroblast behaviour and function is less clear. The ability of fibroblasts to respond and memorise different environmental stimuli is essential for tissue development and homeostasis and may become dysregulated in chronic disease conditions such as fibrosis and cancer. Here we summarise the four emerging key areas of fibroblast adaptation: positional, mechanical, inflammatory, and metabolic memory and highlight the underlying mechanisms and their implications in tissue homeostasis and disease.
Collapse
|
19
|
Perspective: The Mechanobiology of Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13174275. [PMID: 34503085 PMCID: PMC8428343 DOI: 10.3390/cancers13174275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second most deadly primary cancer in the world and is thus a major global health challenge. HCC primarily develops in patients with an underlying chronic liver disease, the vast majority with advanced cirrhosis, characterized by increased matrix deposition and liver stiffness. Liver stiffness is highly associated with cancer development and poor patient outcome and is measured clinically to assess cancer risk; cirrhotic livers greatly exceed the threshold stiffness shown to alter hepatocyte cell behavior and to increase the malignancy of cancer cells. Recent studies have shown that cirrhotic liver cells have highly irregular nuclear morphologies and that nuclear deformation mediates mechanosensitive signaling. Separate research has shown that nuclear deformation can increase genetic instability and the accumulation of DNA damage in migrating cancer cells. We hypothesize that the mechanical changes associated with chronic liver disease are drivers of oncogenesis, activating mechanosensitive signaling pathways, increasing rates of DNA damage, and ultimately inducing malignant transformation.
Collapse
|
20
|
Zhang D, Zhang R, Song X, Yan KC, Liang H. Uniaxial Cyclic Stretching Promotes Chromatin Accessibility of Gene Loci Associated With Mesenchymal Stem Cells Morphogenesis and Osteogenesis. Front Cell Dev Biol 2021; 9:664545. [PMID: 34307349 PMCID: PMC8294092 DOI: 10.3389/fcell.2021.664545] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/28/2021] [Indexed: 01/08/2023] Open
Abstract
It has been previously demonstrated that uniaxial cyclic stretching (UCS) induces differentiation of mesenchymal stem cells (MSCs) into osteoblasts in vitro. It is also known that interactions between cells and external forces occur at various aspects including cell–matrix, cytoskeleton, nucleus membrane, and chromatin. However, changes in chromatin landscape during this process are still not clear. The present study was aimed to determine changes of chromatin accessibility under cyclic stretch. The influence of cyclic stretching on the morphology, proliferation, and differentiation of hMSCs was characterized. Changes of open chromatin sites were determined by assay for transposase accessible chromatin with high-throughput sequencing (ATAC-seq). Our results showed that UCS induced cell reorientation and actin stress fibers realignment, and in turn caused nuclear reorientation and deformation. Compared with unstrained group, the expression of osteogenic and chondrogenic marker genes were the highest in group of 1 Hz + 8% strain; this condition also led to lower cell proliferation rate. Furthermore, there were 2022 gene loci with upregulated chromatin accessibility in 1 Hz + 8% groups based on the analysis of chromatin accessibility. These genes are associated with regulation of cell morphogenesis, cell–substrate adhesion, and ossification. Signaling pathways involved in osteogenic differentiation were found in up-regulated GO biological processes. These findings demonstrated that UCS increased the openness of gene loci associated with regulation of cell morphogenesis and osteogenesis as well as the corresponding transcription activities. Moreover, the findings also connect the changes in chromatin accessibility with cell reorientation, nuclear reorientation, and deformation. Our study may provide reference for directed differentiation of stem cells induced by mechanical microenvironments.
Collapse
Affiliation(s)
- Duo Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Ran Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Xiaoyuan Song
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Division of Life Sciences and Medicine, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Karen Chang Yan
- Mechanical Engineering and Biomedical Engineering, The College of New Jersey, Ewing Township, NJ, United States
| | - Haiyi Liang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| |
Collapse
|
21
|
Stowers RS. Advances in Extracellular Matrix-Mimetic Hydrogels to Guide Stem Cell Fate. Cells Tissues Organs 2021; 211:703-720. [PMID: 34082418 DOI: 10.1159/000514851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/11/2021] [Indexed: 01/25/2023] Open
Abstract
In the fields of regenerative medicine and tissue engineering, stem cells offer vast potential for treating or replacing diseased and damaged tissue. Much progress has been made in understanding stem cell biology, yielding protocols for directing stem cell differentiation toward the cell type of interest for a specific application. One particularly interesting and powerful signaling cue is the extracellular matrix (ECM) surrounding stem cells, a network of biopolymers that, along with cells, makes up what we define as a tissue. The composition, structure, biochemical features, and mechanical properties of the ECM are varied in different tissues and developmental stages, and serve to instruct stem cells toward a specific lineage. By understanding and recapitulating some of these ECM signaling cues through engineered ECM-mimicking hydrogels, stem cell fate can be directed in vitro. In this review, we will summarize recent advances in material systems to guide stem cell fate, highlighting innovative methods to capture ECM functionalities and how these material systems can be used to provide basic insight into stem cell biology or make progress toward therapeutic objectives.
Collapse
Affiliation(s)
- Ryan S Stowers
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California, USA
| |
Collapse
|
22
|
Liu H, Usprech JF, Parameshwar PK, Sun Y, Simmons CA. Combinatorial screen of dynamic mechanical stimuli for predictive control of MSC mechano-responsiveness. SCIENCE ADVANCES 2021; 7:7/19/eabe7204. [PMID: 33962940 PMCID: PMC8104874 DOI: 10.1126/sciadv.abe7204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/19/2021] [Indexed: 05/05/2023]
Abstract
Mechanobiological-based control of mesenchymal stromal cells (MSCs) to facilitate engineering and regeneration of load-bearing tissues requires systematic investigations of specific dynamic mechanical stimulation protocols. Using deformable membrane microdevice arrays paired with combinatorial experimental design and modeling, we probed the individual and integrative effects of mechanical stimulation parameters (strain magnitude, rate at which strain is changed, and duty period) on myofibrogenesis and matrix production of MSCs in three-dimensional hydrogels. These functions were found to be dominantly influenced by a previously unidentified, higher-order interactive effect between strain magnitude and duty period. Empirical models based on our combinatorial cue-response data predicted an optimal loading regime in which strain magnitude and duty period were increased synchronously over time, which was validated to most effectively promote MSC matrix production. These findings inform the design of loading regimes for MSC-based engineered tissues and validate a broadly applicable approach to probe multifactorial regulating effects of mechanobiological cues.
Collapse
Affiliation(s)
- Haijiao Liu
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Jenna F Usprech
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Prabu Karthick Parameshwar
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
23
|
Ghosh S, Cuevas VC, Seelbinder B, Neu CP. Image-Based Elastography of Heterochromatin and Euchromatin Domains in the Deforming Cell Nucleus. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006109. [PMID: 33448065 PMCID: PMC7869959 DOI: 10.1002/smll.202006109] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/29/2020] [Indexed: 05/21/2023]
Abstract
Chromatin of the eukaryotic cell nucleus comprises microscopically dense heterochromatin and loose euchromatin domains, each with distinct transcriptional ability and roles in cellular mechanotransduction. While recent methods are developed to characterize the mechanics of nucleus, measurement of intranuclear mechanics remains largely unknown. Here, the development of "nuclear elastography," which combines microscopic imaging and computational modeling to quantify the relative elasticity of the heterochromatin and euchromatin domains, is described. Using contracting murine embryonic cardiomyocytes, nuclear elastography reveals that the heterochromatin is almost four times stiffer than the euchromatin at peak deformation. The relative elasticity between the two domains changes rapidly during the active deformation of the cardiomyocyte in the normal physiological condition but progresses more slowly in cells cultured in a mechanically stiff environment, although the relative stiffness at peak deformation does not change. Further, it is found that the disruption of the Klarsicht, ANC-1, Syne Homology domain of the Linker of Nucleoskeleton and Cytoskeleton complex compromises the intranuclear elasticity distribution resulting in elastically similar heterochromatin and euchromatin. These results provide insight into the elastography dynamics of heterochromatin and euchromatin domains and provide a noninvasive framework to further investigate the mechanobiological function of subcellular and subnuclear domains limited only by the spatiotemporal resolution of the acquired images.
Collapse
Affiliation(s)
- Soham Ghosh
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Victor Crespo Cuevas
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Benjamin Seelbinder
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| | - Corey P. Neu
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO
| |
Collapse
|
24
|
Focus on time: dynamic imaging reveals stretch-dependent cell relaxation and nuclear deformation. Biophys J 2021; 120:764-772. [PMID: 33524370 DOI: 10.1016/j.bpj.2021.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 01/08/2021] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Abstract
Among the stimuli to which cells are exposed in vivo, it has been shown that tensile deformations induce specific cellular responses in musculoskeletal, cardiovascular, and stromal tissues. However, the early response of cells to sustained substrate-based stretch has remained elusive because of the short timescale at which it occurs. To measure the tensile mechanical properties of adherent cells immediately after the application of substrate deformations, we have developed a dynamic traction force microscopy method that enables subsecond temporal resolution imaging of transient subcellular events. The system employs a novel, to our knowledge, tracking approach with minimal computational overhead to compensate substrate-based, stretch-induced motion/drift of stretched single cells in real time, allowing capture of biophysical phenomena on multiple channels by fluorescent multichannel imaging on a single camera, thus avoiding the need for beam splitting with the associated loss of light. Using this tool, we have characterized the transient subcellular forces and nuclear deformations of single cells immediately after the application of equibiaxial strain. Our experiments reveal significant differences in the cell relaxation dynamics and in the intracellular propagation of force to the nuclear compartment in cells stretched at different strain rates and exposes the need for time control for the correct interpretation of dynamic cell mechanics experiments.
Collapse
|
25
|
Perea Paizal J, Au SH, Bakal C. Squeezing through the microcirculation: survival adaptations of circulating tumour cells to seed metastasis. Br J Cancer 2021; 124:58-65. [PMID: 33257836 PMCID: PMC7782506 DOI: 10.1038/s41416-020-01176-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
During metastasis, tumour cells navigating the vascular circulatory system-circulating tumour cells (CTCs)-encounter capillary beds, where they start the process of extravasation. Biomechanical constriction forces exerted by the microcirculation compromise the survival of tumour cells within capillaries, but a proportion of CTCs manage to successfully extravasate and colonise distant sites. Despite the profound importance of this step in the progression of metastatic cancers, the factors about this deadly minority of cells remain elusive. Growing evidence suggests that mechanical forces exerted by the capillaries might induce adaptive mechanisms in CTCs, enhancing their survival and metastatic potency. Advances in microfluidics have enabled a better understanding of the cell-survival capabilities adopted in capillary-mimicking constrictions. In this review, we will highlight adaptations developed by CTCs to endure mechanical constraints in the microvasculature and outline how these mechanical forces might trigger dynamic changes towards a more invasive phenotype. A better understanding of the dynamic mechanisms adopted by CTCs within the microcirculation that ultimately lead to metastasis could open up novel therapeutic avenues.
Collapse
Affiliation(s)
- Julia Perea Paizal
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK.
- Division of Cancer Biology, Chester Beatty Laboratories, Institute of Cancer Research, 237 Fulham Road, London, SW6 6JB, UK.
- Cancer Research UK Convergence Science Centre, Roderic Hill Building, Imperial College London, London, SW7 2BB, UK.
| | - Sam H Au
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
- Cancer Research UK Convergence Science Centre, Roderic Hill Building, Imperial College London, London, SW7 2BB, UK
| | - Chris Bakal
- Division of Cancer Biology, Chester Beatty Laboratories, Institute of Cancer Research, 237 Fulham Road, London, SW6 6JB, UK.
- Cancer Research UK Convergence Science Centre, Roderic Hill Building, Imperial College London, London, SW7 2BB, UK.
| |
Collapse
|
26
|
Migliorini E, Guevara-Garcia A, Albiges-Rizo C, Picart C. Learning from BMPs and their biophysical extracellular matrix microenvironment for biomaterial design. Bone 2020; 141:115540. [PMID: 32730925 PMCID: PMC7614069 DOI: 10.1016/j.bone.2020.115540] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 01/19/2023]
Abstract
It is nowadays well-accepted that the extracellular matrix (ECM) is not a simple reservoir for growth factors but is an organization center of their biological activity. In this review, we focus on the ability of the ECM to regulate the biological activity of BMPs. In particular, we survey the role of the ECM components, notably the glycosaminoglycans and fibrillary ECM proteins, which can be promoters or repressors of the biological activities mediated by the BMPs. We examine how a process called mechano-transduction induced by the ECM can affect BMP signaling, including BMP internalization by the cells. We also focus on the spatio-temporal regulation of the BMPs, including their release from the ECM, which enables to modulate their spatial localization as well as their local concentration. We highlight how biomaterials can recapitulate some aspects of the BMPs/ECM interactions and help to answer fundamental questions to reveal previously unknown molecular mechanisms. Finally, the design of new biomaterials inspired by the ECM to better present BMPs is discussed, and their use for a more efficient bone regeneration in vivo is also highlighted.
Collapse
Affiliation(s)
- Elisa Migliorini
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France.
| | - Amaris Guevara-Garcia
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France; Université Grenoble Alpes, Institut for Advances Biosciences, Institute Albert Bonniot, INSERM U1209, CNRS 5309, La Tronche, France
| | - Corinne Albiges-Rizo
- Université Grenoble Alpes, Institut for Advances Biosciences, Institute Albert Bonniot, INSERM U1209, CNRS 5309, La Tronche, France
| | - Catherine Picart
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France.
| |
Collapse
|
27
|
Michel JB. Phylogenic Determinants of Cardiovascular Frailty, Focus on Hemodynamics and Arterial Smooth Muscle Cells. Physiol Rev 2020; 100:1779-1837. [DOI: 10.1152/physrev.00022.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The evolution of the circulatory system from invertebrates to mammals has involved the passage from an open system to a closed in-parallel system via a closed in-series system, accompanying the increasing complexity and efficiency of life’s biological functions. The archaic heart enables pulsatile motion waves of hemolymph in invertebrates, and the in-series circulation in fish occurs with only an endothelium, whereas mural smooth muscle cells appear later. The present review focuses on evolution of the circulatory system. In particular, we address how and why this evolution took place from a closed, flowing, longitudinal conductance at low pressure to a flowing, highly pressurized and bifurcating arterial compartment. However, although arterial pressure was the latest acquired hemodynamic variable, the general teleonomy of the evolution of species is the differentiation of individual organ function, supported by specific fueling allowing and favoring partial metabolic autonomy. This was achieved via the establishment of an active contractile tone in resistance arteries, which permitted the regulation of blood supply to specific organ activities via its localized function-dependent inhibition (active vasodilation). The global resistance to viscous blood flow is the peripheral increase in frictional forces caused by the tonic change in arterial and arteriolar radius, which backscatter as systemic arterial blood pressure. Consequently, the arterial pressure gradient from circulating blood to the adventitial interstitium generates the unidirectional outward radial advective conductance of plasma solutes across the wall of conductance arteries. This hemodynamic evolution was accompanied by important changes in arterial wall structure, supported by smooth muscle cell functional plasticity, including contractility, matrix synthesis and proliferation, endocytosis and phagocytosis, etc. These adaptive phenotypic shifts are due to epigenetic regulation, mainly related to mechanotransduction. These paradigms actively participate in cardio-arterial pathologies such as atheroma, valve disease, heart failure, aneurysms, hypertension, and physiological aging.
Collapse
|
28
|
Nuclear mechanosensing controls MSC osteogenic potential through HDAC epigenetic remodeling. Proc Natl Acad Sci U S A 2020; 117:21258-21266. [PMID: 32817542 PMCID: PMC7474590 DOI: 10.1073/pnas.2006765117] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cells sense mechanical cues from the extracellular matrix to regulate cellular behavior and maintain tissue homeostasis. The nucleus has been implicated as a key mechanosensor and can directly influence chromatin organization, epigenetic modifications, and gene expression. Dysregulation of nuclear mechanosensing has been implicated in several diseases, including bone degeneration. Here, we exploit photostiffening hydrogels to manipulate nuclear mechanosensing in human mesenchymal stem cells (hMSCs) in vitro. Results show that hMSCs respond to matrix stiffening by increasing nuclear tension and causing an increase in histone acetylation via deactivation of histone deacetylases (HDACs). This ultimately induces osteogenic fate commitment. Disrupting nuclear mechanosensing by disconnecting the nucleus from the cytoskeleton up-regulates HDACs and prevents osteogenesis. Resetting HDAC activity back to healthy levels rescues the epigenetic and osteogenic response in hMSCs with pathological nuclear mechanosensing. Notably, bone from patients with osteoarthritis displays similar defective nuclear mechanosensing. Collectively, our results reveal that nuclear mechanosensing controls hMSC osteogenic potential mediated by HDAC epigenetic remodeling and that this cellular mechanism is likely relevant to bone-related diseases.
Collapse
|
29
|
Fischer T, Hayn A, Mierke CT. Effect of Nuclear Stiffness on Cell Mechanics and Migration of Human Breast Cancer Cells. Front Cell Dev Biol 2020; 8:393. [PMID: 32548118 PMCID: PMC7272586 DOI: 10.3389/fcell.2020.00393] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
The migration and invasion of cancer cells through 3D confined extracellular matrices is coupled to cell mechanics and the mechanics of the extracellular matrix. Cell mechanics is mainly determined by both the mechanics of the largest organelle in the cell, the nucleus, and the cytoskeletal architecture of the cell. Hence, cytoskeletal and nuclear mechanics are the major contributors to cell mechanics. Among other factors, steric hindrances of the extracellular matrix confinement are supposed to affect nuclear mechanics and thus also influence cell mechanics. Therefore, we propose that the percentage of invasive cells and their invasion depths into loose and dense 3D extracellular matrices is regulated by both nuclear and cytoskeletal mechanics. In order to investigate the effect of both nuclear and cytoskeletal mechanics on the overall cell mechanics, we firstly altered nuclear mechanics by the chromatin de-condensing reagent Trichostatin A (TSA) and secondly altered cytoskeletal mechanics by addition of actin polymerization inhibitor Latrunculin A and the myosin inhibitor Blebbistatin. In fact, we found that TSA-treated MDA-MB-231 human breast cancer cells increased their invasion depth in dense 3D extracellular matrices, whereas the invasion depths in loose matrices were decreased. Similarly, the invasion depths of TSA-treated MCF-7 human breast cancer cells in dense matrices were significantly increased compared to loose matrices, where the invasion depths were decreased. These results are also valid in the presence of a matrix-metalloproteinase inhibitor GM6001. Using atomic force microscopy (AFM), we found that the nuclear stiffnesses of both MDA-MB-231 and MCF-7 breast cancer cells were pronouncedly higher than their cytoskeletal stiffness, whereas the stiffness of the nucleus of human mammary epithelial cells was decreased compared to their cytoskeleton. TSA treatment reduced cytoskeletal and nuclear stiffness of MCF-7 cells, as expected. However, a softening of the nucleus by TSA treatment may induce a stiffening of the cytoskeleton of MDA-MB-231 cells and subsequently an apparent stiffening of the nucleus. Inhibiting actin polymerization using Latrunculin A revealed a softer nucleus of MDA-MB-231 cells under TSA treatment. This indicates that the actin-dependent cytoskeletal stiffness seems to be influenced by the TSA-induced nuclear stiffness changes. Finally, the combined treatment with TSA and Latrunculin A further justifies the hypothesis of apparent nuclear stiffening, indicating that cytoskeletal mechanics seem to be regulated by nuclear mechanics.
Collapse
Affiliation(s)
- Tony Fischer
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| | - Alexander Hayn
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| | - Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| |
Collapse
|
30
|
Hannezo E, Heisenberg CP. Mechanochemical Feedback Loops in Development and Disease. Cell 2020; 178:12-25. [PMID: 31251912 DOI: 10.1016/j.cell.2019.05.052] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/17/2019] [Accepted: 05/24/2019] [Indexed: 12/31/2022]
Abstract
There is increasing evidence that both mechanical and biochemical signals play important roles in development and disease. The development of complex organisms, in particular, has been proposed to rely on the feedback between mechanical and biochemical patterning events. This feedback occurs at the molecular level via mechanosensation but can also arise as an emergent property of the system at the cellular and tissue level. In recent years, dynamic changes in tissue geometry, flow, rheology, and cell fate specification have emerged as key platforms of mechanochemical feedback loops in multiple processes. Here, we review recent experimental and theoretical advances in understanding how these feedbacks function in development and disease.
Collapse
Affiliation(s)
- Edouard Hannezo
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| | | |
Collapse
|
31
|
Dai EN, Heo S, Mauck RL. "Looping In" Mechanics: Mechanobiologic Regulation of the Nucleus and the Epigenome. Adv Healthc Mater 2020; 9:e2000030. [PMID: 32285630 DOI: 10.1002/adhm.202000030] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
Cells respond to physical cues in their microenvironment. These cues result in changes in cell behavior, some of which are transient, and others of which are permanent. Understanding and leveraging permanent alteration of cell behavior induced by mechanical cues, or "mechanical memories," is an important aim in cell and tissue engineering. Herein, this paper reviews the existing literature outlining how cells may store memories of biophysical cues with a specific focus on the nucleus, the storehouse of information in eukaryotic cells. In particular, this review details mechanically driven adaptations in nuclear structure and genome organization and outlines potential mechanisms by which mechanical memories may be encoded within the structure and organization of the nucleus and chromatin.
Collapse
Affiliation(s)
- Eric N. Dai
- Departments of Orthopaedic Surgery and Bioengineering University of Pennsylvania Philadelphia PA 19104 USA
- Translational Musculoskeletal Research Center Corporal Michael J. Crescenz VA Medical Center Philadelphia PA 19104 USA
| | - Su‐Jin Heo
- Departments of Orthopaedic Surgery and Bioengineering University of Pennsylvania Philadelphia PA 19104 USA
- Translational Musculoskeletal Research Center Corporal Michael J. Crescenz VA Medical Center Philadelphia PA 19104 USA
| | - Robert L. Mauck
- Departments of Orthopaedic Surgery and Bioengineering University of Pennsylvania Philadelphia PA 19104 USA
- Translational Musculoskeletal Research Center Corporal Michael J. Crescenz VA Medical Center Philadelphia PA 19104 USA
- McKay Orthopaedic Research Laboratory University of Pennsylvania Philadelphia PA 19104‐6081 USA
| |
Collapse
|
32
|
Biggs LC, Kim CS, Miroshnikova YA, Wickström SA. Mechanical Forces in the Skin: Roles in Tissue Architecture, Stability, and Function. J Invest Dermatol 2020; 140:284-290. [DOI: 10.1016/j.jid.2019.06.137] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/19/2019] [Accepted: 06/27/2019] [Indexed: 01/08/2023]
|
33
|
Song Y, Soto J, Chen B, Yang L, Li S. Cell engineering: Biophysical regulation of the nucleus. Biomaterials 2020; 234:119743. [PMID: 31962231 DOI: 10.1016/j.biomaterials.2019.119743] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/02/2019] [Accepted: 12/25/2019] [Indexed: 12/12/2022]
Abstract
Cells live in a complex and dynamic microenvironment, and a variety of microenvironmental cues can regulate cell behavior. In addition to biochemical signals, biophysical cues can induce not only immediate intracellular responses, but also long-term effects on phenotypic changes such as stem cell differentiation, immune cell activation and somatic cell reprogramming. Cells respond to mechanical stimuli via an outside-in and inside-out feedback loop, and the cell nucleus plays an important role in this process. The mechanical properties of the nucleus can directly or indirectly modulate mechanotransduction, and the physical coupling of the cell nucleus with the cytoskeleton can affect chromatin structure and regulate the epigenetic state, gene expression and cell function. In this review, we will highlight the recent progress in nuclear biomechanics and mechanobiology in the context of cell engineering, tissue remodeling and disease development.
Collapse
Affiliation(s)
- Yang Song
- Department of Bioengineering, University of California, Los Angeles, CA, USA; School of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Jennifer Soto
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Binru Chen
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Li Yang
- School of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, CA, USA; Department of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
34
|
Bastounis EE, Yeh YT, Theriot JA. Subendothelial stiffness alters endothelial cell traction force generation while exerting a minimal effect on the transcriptome. Sci Rep 2019; 9:18209. [PMID: 31796790 PMCID: PMC6890669 DOI: 10.1038/s41598-019-54336-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023] Open
Abstract
Endothelial cells respond to changes in subendothelial stiffness by altering their migration and mechanics, but whether those responses are due to transcriptional reprogramming remains largely unknown. We measured traction force generation and also performed gene expression profiling for two endothelial cell types grown in monolayers on soft or stiff matrices: primary human umbilical vein endothelial cells (HUVEC) and immortalized human microvascular endothelial cells (HMEC-1). Both cell types respond to changes in subendothelial stiffness by increasing the traction stresses they exert on stiffer as compared to softer matrices, and exhibit a range of altered protein phosphorylation or protein conformational changes previously implicated in mechanotransduction. However, the transcriptome has only a minimal role in this conserved biomechanical response. Only few genes were differentially expressed in each cell type in a stiffness-dependent manner, and none were shared between them. In contrast, thousands of genes were differentially regulated in HUVEC as compared to HMEC-1. HUVEC (but not HMEC-1) upregulate expression of TGF-β2 on stiffer matrices, and also respond to application of exogenous TGF-β2 by enhancing their endogenous TGF-β2 expression and their cell-matrix traction stresses. Altogether, these findings provide insights into the relationship between subendothelial stiffness, endothelial mechanics and variation of the endothelial cell transcriptome, and reveal that subendothelial stiffness, while critically altering endothelial cells’ mechanical behavior, minimally affects their transcriptome.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195-1800, USA
| | - Yi-Ting Yeh
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195-1800, USA.
| |
Collapse
|
35
|
Stanley A, Heo SJ, Mauck RL, Mourkioti F, Shore EM. Elevated BMP and Mechanical Signaling Through YAP1/RhoA Poises FOP Mesenchymal Progenitors for Osteogenesis. J Bone Miner Res 2019; 34:1894-1909. [PMID: 31107558 PMCID: PMC7209824 DOI: 10.1002/jbmr.3760] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/03/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease characterized by the formation of extraskeletal bone, or heterotopic ossification (HO), in soft connective tissues such as skeletal muscle. All familial and sporadic cases with a classic clinical presentation of FOP carry a gain-of-function mutation (R206H; c.617 G > A) in ACVR1, a cell surface receptor that mediates bone morphogenetic protein (BMP) signaling. The BMP signaling pathway is recognized for its chondro/osteogenic-induction potential, and HO in FOP patients forms ectopic but qualitatively normal endochondral bone tissue through misdirected cell fate decisions by tissue-resident mesenchymal stem cells. In addition to biochemical ligand-receptor signaling, mechanical cues from the physical environment are transduced to activate intracellular signaling, a process known as mechanotransduction, and can influence cell fates. Utilizing an established mesenchymal stem cell model of mouse embryonic fibroblasts (MEFs) from the Acvr1R206H/+ mouse model that mimics the human disease, we demonstrated that activation of the mechanotransductive effectors Rho/ROCK and YAP1 are increased in Acvr1R206H/+ cells. We show that on softer substrates, a condition associated with low mechanical signaling, the morphology of Acvr1R206H/+ cells is similar to the morphology of control Acvr1+/+ cells on stiffer substrates, a condition that activates mechanotransduction. We further determined that Acvr1R206H/+ cells are poised for osteogenic differentiation, expressing increased levels of chondro/osteogenic markers compared with Acvr1+/+ cells. We also identified increased YAP1 nuclear localization in Acvr1R206H/+ cells, which can be rescued by either BMP inhibition or Rho antagonism. Our results establish RhoA and YAP1 signaling as modulators of mechanotransduction in FOP and suggest that aberrant mechanical signals, combined with and as a result of the increased BMP pathway signaling through mutant ACVR1, lead to misinterpretation of the cellular microenvironment and a heightened sensitivity to mechanical stimuli that promotes commitment of Acvr1R206H/+ progenitor cells to chondro/osteogenic lineages.
Collapse
Affiliation(s)
- Alexandra Stanley
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Su-jin Heo
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA
- Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, PA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA
| | - Robert L. Mauck
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA
- Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, PA
- Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Departments of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eileen M. Shore
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
36
|
Abstract
Cellular behavior is continuously affected by microenvironmental forces through the process of mechanotransduction, in which mechanical stimuli are rapidly converted to biochemical responses. Mounting evidence suggests that the nucleus itself is a mechanoresponsive element, reacting to cytoskeletal forces and mediating downstream biochemical responses. The nucleus responds through a host of mechanisms, including partial unfolding, conformational changes, and phosphorylation of nuclear envelope proteins; modulation of nuclear import/export; and altered chromatin organization, resulting in transcriptional changes. It is unclear which of these events present direct mechanotransduction processes and which are downstream of other mechanotransduction pathways. We critically review and discuss the current evidence for nuclear mechanotransduction, particularly in the context of stem cell fate, a largely unexplored topic, and in disease, where an improved understanding of nuclear mechanotransduction is beginning to open new treatment avenues. Finally, we discuss innovative technological developments that will allow outstanding questions in the rapidly growing field of nuclear mechanotransduction to be answered.
Collapse
Affiliation(s)
- Melanie Maurer
- Meinig School of Biomedical Engineering and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, USA; ,
| | - Jan Lammerding
- Meinig School of Biomedical Engineering and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, USA; ,
| |
Collapse
|
37
|
Miroshnikova YA, Cohen I, Ezhkova E, Wickström SA. Epigenetic gene regulation, chromatin structure, and force-induced chromatin remodelling in epidermal development and homeostasis. Curr Opin Genet Dev 2019; 55:46-51. [PMID: 31112907 DOI: 10.1016/j.gde.2019.04.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 01/14/2023]
Abstract
The skin epidermis is a constantly renewing stratified epithelium that provides essential protective barrier functions throughout life. Epidermal stratification is governed by a step-wise differentiation program that requires precise spatiotemporal control of gene expression. How epidermal self-renewal and differentiation are regulated remains a fundamental open question. Cell-intrinsic and cell-extrinsic mechanisms that modify chromatin structure and interactions have been identified as key regulators of epidermal differentiation and stratification. Here, we will review the recent advances in our understanding of how chromatin modifiers, tissue-specific transcription factors, and force-induced nuclear remodeling processes function to shape chromatin and to control epidermal tissue development and homeostasis.
Collapse
Affiliation(s)
- Yekaterina A Miroshnikova
- Helsinki Institute of Life Science, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Finland; Wihuri Research Institute, University of Helsinki, Finland; Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Finland; Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Idan Cohen
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Elena Ezhkova
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA.
| | - Sara A Wickström
- Helsinki Institute of Life Science, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Finland; Wihuri Research Institute, University of Helsinki, Finland; Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Finland; Max Planck Institute for Biology of Ageing, Cologne, Germany.
| |
Collapse
|
38
|
Killaars AR, Grim JC, Walker CJ, Hushka EA, Brown TE, Anseth KS. Extended Exposure to Stiff Microenvironments Leads to Persistent Chromatin Remodeling in Human Mesenchymal Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801483. [PMID: 30775233 PMCID: PMC6364489 DOI: 10.1002/advs.201801483] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/16/2018] [Indexed: 05/02/2023]
Abstract
Bone marrow derived human mesenchymal stem cells (hMSCs) are a promising cell source for regenerative therapies; however, ex vivo expansion is often required to achieve clinically useful cells numbers. Recent results reveal that when MSCs are cultured in stiff microenvironments, their regenerative capacity can be altered in a manner that is dependent on time (e.g., a mechanical dosing analogous to a chemical one). It is hypothesized that epigenomic modifications are involved in storing these mechanical cues, regulating gene expression, and ultimately leading to a mechanical memory. Using hydrogels containing an allyl sulfide cross-linker and a radical-mediated addition-fragmentation chain transfer process, in situ softened hMSC-laden hydrogels at different time points are achieved and the effects of short-term and long-term mechanical dosing on epigenetic modifications in hMSCs are quantified. Results show that histone acetylation and chromatin organization adapt rapidly after softening and can be reversible or irreversible depending on time of exposure to stiff microenvironments. Furthermore, epigenetic modulators are differentially expressed depending on the culture history. Collectively, these experiments suggest that epigenetic remodeling can be persistent and might be a memory keeper.
Collapse
Affiliation(s)
- Anouk R. Killaars
- Program of Materials Science and Engineering and BioFrontiers InstituteUniversity of Colorado BoulderJennie Smoly Caruthers Biotechnology Building, 3415 Colorado AveBoulderCO80303USA
| | - Joseph C. Grim
- Department of Chemical and Biological Engineering and BioFrontiers InstituteUniversity of Colorado BoulderJennie Smoly Caruthers Biotechnology Building, 3415 Colorado AveBoulderCO80303USA
| | - Cierra J. Walker
- Program of Materials Science and Engineering and BioFrontiers InstituteUniversity of Colorado BoulderJennie Smoly Caruthers Biotechnology Building, 3415 Colorado AveBoulderCO80303USA
| | - Ella A. Hushka
- Department of Chemical and Biological Engineering and BioFrontiers InstituteUniversity of Colorado BoulderJennie Smoly Caruthers Biotechnology Building, 3415 Colorado AveBoulderCO80303USA
| | - Tobin E. Brown
- Department of Chemical and Biological Engineering and BioFrontiers InstituteUniversity of Colorado BoulderJennie Smoly Caruthers Biotechnology Building, 3415 Colorado AveBoulderCO80303USA
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering and BioFrontiers InstituteUniversity of Colorado BoulderJennie Smoly Caruthers Biotechnology Building, 3415 Colorado AveBoulderCO80303USA
| |
Collapse
|
39
|
Haupt J, Stanley A, McLeod CM, Cosgrove BD, Culbert AL, Wang L, Mourkioti F, Mauck RL, Shore EM. ACVR1 R206H FOP mutation alters mechanosensing and tissue stiffness during heterotopic ossification. Mol Biol Cell 2018; 30:17-29. [PMID: 30379592 PMCID: PMC6337906 DOI: 10.1091/mbc.e18-05-0311] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
An activating bone morphogenetic proteins (BMP) type I receptor ACVR1 (ACVR1R206H) mutation enhances BMP pathway signaling and causes the rare genetic disorder of heterotopic (extraskeletal) bone formation fibrodysplasia ossificans progressiva. Heterotopic ossification frequently occurs following injury as cells aberrantly differentiate during tissue repair. Biomechanical signals from the tissue microenvironment and cellular responses to these physical cues, such as stiffness and rigidity, are important determinants of cell differentiation and are modulated by BMP signaling. We used an Acvr1R206H/+ mouse model of injury-induced heterotopic ossification to examine the fibroproliferative tissue preceding heterotopic bone and identified pathologic stiffening at this stage of repair. In response to microenvironment stiffness, in vitro assays showed that Acvr1R206H/+ cells inappropriately sense their environment, responding to soft substrates with a spread morphology similar to wild-type cells on stiff substrates and to cells undergoing osteoblastogenesis. Increased activation of RhoA and its downstream effectors demonstrated increased mechanosignaling. Nuclear localization of the pro-osteoblastic factor RUNX2 on soft and stiff substrates suggests a predisposition to this cell fate. Our data support that increased BMP signaling in Acvr1R206H/+ cells alters the tissue microenvironment and results in misinterpretation of the tissue microenvironment through altered sensitivity to mechanical stimuli that lowers the threshold for commitment to chondro/osteogenic lineages.
Collapse
Affiliation(s)
- Julia Haupt
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104.,Department of Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Alexandra Stanley
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104.,Department of Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Claire M McLeod
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104.,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Brian D Cosgrove
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104.,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Andria L Culbert
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104.,Department of Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Linda Wang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104.,Department of Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104.,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - Robert L Mauck
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104.,Department of Mechanical Engineering and Applied Mechanics, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104.,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Eileen M Shore
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104.,Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104.,Department of Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
40
|
Damodaran K, Venkatachalapathy S, Alisafaei F, Radhakrishnan AV, Sharma Jokhun D, Shenoy VB, Shivashankar GV. Compressive force induces reversible chromatin condensation and cell geometry-dependent transcriptional response. Mol Biol Cell 2018; 29:3039-3051. [PMID: 30256731 PMCID: PMC6333178 DOI: 10.1091/mbc.e18-04-0256] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Fibroblasts exhibit heterogeneous cell geometries in tissues and integrate both mechanical and biochemical signals in their local microenvironment to regulate genomic programs via chromatin remodelling. While in connective tissues fibroblasts experience tensile and compressive forces (CFs), the role of compressive forces in regulating cell behavior and, in particular, the impact of cell geometry in modulating transcriptional response to such extrinsic mechanical forces is unclear. Here we show that CF on geometrically well-defined mouse fibroblast cells reduces actomyosin contractility and shuttles histone deacetylase 3 (HDAC3) into the nucleus. HDAC3 then triggers an increase in the heterochromatin content by initiating removal of acetylation marks on the histone tails. This suggests that, in response to CF, fibroblasts condense their chromatin and enter into a transcriptionally less active and quiescent states as also revealed by transcriptome analysis. On removal of CF, the alteration in chromatin condensation was reversed. We also present a quantitative model linking CF-dependent changes in actomyosin contractility leading to chromatin condensation. Further, transcriptome analysis also revealed that the transcriptional response of cells to CF was geometry dependent. Collectively, our results suggest that CFs induce chromatin condensation and geometry-dependent differential transcriptional response in fibroblasts that allows maintenance of tissue homeostasis.
Collapse
Affiliation(s)
- Karthik Damodaran
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, 117411, Singapore
| | - Saradha Venkatachalapathy
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, 117411, Singapore
| | - Farid Alisafaei
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
| | - A V Radhakrishnan
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, 117411, Singapore
| | - Doorgesh Sharma Jokhun
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, 117411, Singapore
| | - Vivek B Shenoy
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
| | - G V Shivashankar
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, 117411, Singapore.,FIRC Institute for Molecular Oncology (IFOM), 20139 Milan, Italy
| |
Collapse
|
41
|
Heo SJ, Szczesny SE, Kim DH, Saleh KS, Mauck RL. Expansion of mesenchymal stem cells on electrospun scaffolds maintains stemness, mechano-responsivity, and differentiation potential. J Orthop Res 2018; 36:808-815. [PMID: 29027711 PMCID: PMC5839953 DOI: 10.1002/jor.23772] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 10/06/2017] [Indexed: 02/04/2023]
Abstract
Mesenchymal stem cells (MSCs) hold great promise for regenerative therapies and tissue engineering applications given their multipotential differentiation capacity. However, MSC isolation and expansion are typically performed on super-physiologically stiff tissue culture plastic (TCP), which may alter their behavior and lead to unintended consequences upon implantation. In contrast, electrospun nanofibrous scaffolds possess physical and mechanical properties that are similar to that of native tissue. In this study, we investigated whether isolation and expansion of juvenile bovine MSCs directly onto electrospun nanofibrous scaffolds better preserves MSC phenotype and stemness compared to TCP. Our data show that culture of MSCs on electrospun scaffolds reduces proliferation, decreases cellular senescence, and better maintains stemness compared to cells isolated and expanded on TCP, likely due to a reduction in cell contractility. Furthermore, in contrast to electrospun scaffolds, TCP biased MSCs towards a fibrotic phenotype that persisted even after the cells were reseeded onto a different substrate. Cells pre-cultured on electrospun scaffolds exhibited a heightened response to mechanical stimuli and greater chondrogenesis in methacrylated hyaluronic acid hydrogels. These data suggest that alternative substrates that better approximate the native cell environment should be used to preserve endogenous MSC behavior and may improve their success in tissue engineering applications. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:808-815, 2018.
Collapse
Affiliation(s)
- Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Spencer E. Szczesny
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Dong Hwa Kim
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Kamiel S. Saleh
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA,Address for Correspondence: Robert L. Mauck, Ph.D., Mary Black Ralston Professor of Orthopaedic Surgery, Professor of Bioengineering, McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, Phone: (215) 898-3294, Fax: (215) 573-2133,
| |
Collapse
|
42
|
Cheng B, Lin M, Huang G, Li Y, Ji B, Genin GM, Deshpande VS, Lu TJ, Xu F. Cellular mechanosensing of the biophysical microenvironment: A review of mathematical models of biophysical regulation of cell responses. Phys Life Rev 2017; 22-23:88-119. [PMID: 28688729 PMCID: PMC5712490 DOI: 10.1016/j.plrev.2017.06.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 06/14/2017] [Indexed: 12/11/2022]
Abstract
Cells in vivo reside within complex microenvironments composed of both biochemical and biophysical cues. The dynamic feedback between cells and their microenvironments hinges upon biophysical cues that regulate critical cellular behaviors. Understanding this regulation from sensing to reaction to feedback is therefore critical, and a large effort is afoot to identify and mathematically model the fundamental mechanobiological mechanisms underlying this regulation. This review provides a critical perspective on recent progress in mathematical models for the responses of cells to the biophysical cues in their microenvironments, including dynamic strain, osmotic shock, fluid shear stress, mechanical force, matrix rigidity, porosity, and matrix shape. The review highlights key successes and failings of existing models, and discusses future opportunities and challenges in the field.
Collapse
Affiliation(s)
- Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Guoyou Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuhui Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Baohua Ji
- Biomechanics and Biomaterials Laboratory, Department of Applied Mechanics, Beijing Institute of Technology, Beijing, China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; Department of Mechanical Engineering & Materials Science, and NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis 63130, MO, USA
| | - Vikram S Deshpande
- Department of Engineering, University of Cambridge, Cambridge CB2 1PZ, United Kingdom
| | - Tian Jian Lu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
43
|
Abstract
Exogenous mechanical forces are transmitted through the cell and to the nucleus, initiating mechanotransductive signaling cascades with profound effects on cellular function and stem cell fate. A growing body of evidence has shown that the force sensing and force-responsive elements of the nucleus adapt to these mechanotransductive events, tuning their response to future mechanical input. The mechanisms underlying this “mechano-adaptation” are only just beginning to be elucidated, and it remains poorly understood how these components act and adapt in tandem to drive stem cell differentiation. Here, we review the evidence on how the stem cell nucleus responds and adapts to physical forces, and provide a perspective on how this mechano-adaptation may function to drive and enforce stem cell differentiation.
Collapse
Affiliation(s)
- Su-Jin Heo
- a McKay Orthopaedic Research Laboratory , Department of Orthopaedic Surgery , Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA.,b Department of Bioengineering , School of Engineering and Applied Science, University of Pennsylvania , Philadelphia , PA , USA
| | - Brian D Cosgrove
- a McKay Orthopaedic Research Laboratory , Department of Orthopaedic Surgery , Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA.,b Department of Bioengineering , School of Engineering and Applied Science, University of Pennsylvania , Philadelphia , PA , USA
| | - Eric N Dai
- a McKay Orthopaedic Research Laboratory , Department of Orthopaedic Surgery , Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA.,b Department of Bioengineering , School of Engineering and Applied Science, University of Pennsylvania , Philadelphia , PA , USA
| | - Robert L Mauck
- a McKay Orthopaedic Research Laboratory , Department of Orthopaedic Surgery , Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA.,b Department of Bioengineering , School of Engineering and Applied Science, University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
44
|
Uhler C, Shivashankar GV. Regulation of genome organization and gene expression by nuclear mechanotransduction. Nat Rev Mol Cell Biol 2017; 18:717-727. [PMID: 29044247 DOI: 10.1038/nrm.2017.101] [Citation(s) in RCA: 252] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is well established that cells sense chemical signals from their local microenvironment and transduce them to the nucleus to regulate gene expression programmes. Although a number of experiments have shown that mechanical cues can also modulate gene expression, the underlying mechanisms are far from clear. Nevertheless, we are now beginning to understand how mechanical cues are transduced to the nucleus and how they influence nuclear mechanics, genome organization and transcription. In particular, recent progress in super-resolution imaging, in genome-wide application of RNA sequencing, chromatin immunoprecipitation and chromosome conformation capture and in theoretical modelling of 3D genome organization enables the exploration of the relationship between cell mechanics, 3D chromatin configurations and transcription, thereby shedding new light on how mechanical forces regulate gene expression.
Collapse
Affiliation(s)
- Caroline Uhler
- Department of Electrical Engineering and Computer Science, Laboratory of Information and Decision Systems, Institute for Data, Systems and Society, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| | - G V Shivashankar
- Mechanobiology Institute, National University of Singapore, 119077 Singapore.,Italian Foundation for Cancer Research (FIRC) Institute of Molecular Oncology (IFOM), Milan 20139, Italy
| |
Collapse
|
45
|
Thorpe SD, Lee DA. Dynamic regulation of nuclear architecture and mechanics-a rheostatic role for the nucleus in tailoring cellular mechanosensitivity. Nucleus 2017; 8:287-300. [PMID: 28152338 PMCID: PMC5499908 DOI: 10.1080/19491034.2017.1285988] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Nuclear architecture, a function of both chromatin and nucleoskeleton structure, is known to change with stem cell differentiation and differs between various somatic cell types. These changes in nuclear architecture are associated with the regulation of gene expression and genome function in a cell-type specific manner. Biophysical stimuli are known effectors of differentiation and also elicit stimuli-specific changes in nuclear architecture. This occurs via the process of mechanotransduction whereby extracellular mechanical forces activate several well characterized signaling cascades of cytoplasmic origin, and potentially some recently elucidated signaling cascades originating in the nucleus. Recent work has demonstrated changes in nuclear mechanics both with pluripotency state in embryonic stem cells, and with differentiation progression in adult mesenchymal stem cells. This review explores the interplay between cytoplasmic and nuclear mechanosensitivity, highlighting a role for the nucleus as a rheostat in tuning the cellular mechano-response.
Collapse
Affiliation(s)
- Stephen D Thorpe
- a Institute of Bioengineering, School of Engineering and Materials Science , Queen Mary University of London , London , UK
| | - David A Lee
- a Institute of Bioengineering, School of Engineering and Materials Science , Queen Mary University of London , London , UK
| |
Collapse
|
46
|
Miroshnikova YA, Nava MM, Wickström SA. Emerging roles of mechanical forces in chromatin regulation. J Cell Sci 2017. [DOI: 10.1242/jcs.202192] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
ABSTRACT
Cells are constantly subjected to a spectrum of mechanical cues, such as shear stress, compression, differential tissue rigidity and strain, to which they adapt by engaging mechanisms of mechanotransduction. While the central role of cell adhesion receptors in this process is established, it has only recently been appreciated that mechanical cues reach far beyond the plasma membrane and the cytoskeleton, and are directly transmitted to the nucleus. Furthermore, changes in the mechanical properties of the perinuclear cytoskeleton, nuclear lamina and chromatin are critical for cellular responses and adaptation to external mechanical cues. In that respect, dynamic changes in the nuclear lamina and the surrounding cytoskeleton modify mechanical properties of the nucleus, thereby protecting genetic material from damage. The importance of this mechanism is highlighted by debilitating genetic diseases, termed laminopathies, that result from impaired mechanoresistance of the nuclear lamina. What has been less evident, and represents one of the exciting emerging concepts, is that chromatin itself is an active rheological element of the nucleus, which undergoes dynamic changes upon application of force, thereby facilitating cellular adaption to differential force environments. This Review aims to highlight these emerging concepts by discussing the latest literature in this area and by proposing an integrative model of cytoskeletal and chromatin-mediated responses to mechanical stress.
Collapse
Affiliation(s)
| | - Michele M. Nava
- Paul Gerson Unna Group ‘Skin Homeostasis and Ageing’, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Sara A. Wickström
- Paul Gerson Unna Group ‘Skin Homeostasis and Ageing’, Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne 50931, Germany
| |
Collapse
|
47
|
Heo SJ, Driscoll TP, Thorpe SD, Nerurkar NL, Baker BM, Yang MT, Chen CS, Lee DA, Mauck RL. Differentiation alters stem cell nuclear architecture, mechanics, and mechano-sensitivity. eLife 2016; 5. [PMID: 27901466 PMCID: PMC5148611 DOI: 10.7554/elife.18207] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cell (MSC) differentiation is mediated by soluble and physical cues. In this study, we investigated differentiation-induced transformations in MSC cellular and nuclear biophysical properties and queried their role in mechanosensation. Our data show that nuclei in differentiated bovine and human MSCs stiffen and become resistant to deformation. This attenuated nuclear deformation was governed by restructuring of Lamin A/C and increased heterochromatin content. This change in nuclear stiffness sensitized MSCs to mechanical-loading-induced calcium signaling and differentiated marker expression. This sensitization was reversed when the 'stiff' differentiated nucleus was softened and was enhanced when the 'soft' undifferentiated nucleus was stiffened through pharmacologic treatment. Interestingly, dynamic loading of undifferentiated MSCs, in the absence of soluble differentiation factors, stiffened and condensed the nucleus, and increased mechanosensitivity more rapidly than soluble factors. These data suggest that the nucleus acts as a mechanostat to modulate cellular mechanosensation during differentiation.
Collapse
Affiliation(s)
- Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States.,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Pennsylvania, United States
| | - Tristan P Driscoll
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States.,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Pennsylvania, United States
| | - Stephen D Thorpe
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Nandan L Nerurkar
- Department of Genetics, Harvard Medical School, Harvard University, Boston, United States
| | - Brendon M Baker
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, United States
| | - Michael T Yang
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, United States
| | - David A Lee
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States.,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Pennsylvania, United States
| |
Collapse
|