1
|
Rao R, Yang H, Qiu K, Xu M, Liu H, Shen J, Wang W, Nie R, Chen H, Jiang H. Mechanical confinement triggers spreading and migration of immobile cells by deforming nucleus. Biomaterials 2025; 320:123209. [PMID: 40049023 DOI: 10.1016/j.biomaterials.2025.123209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/18/2025] [Accepted: 02/23/2025] [Indexed: 04/06/2025]
Abstract
Cells in vivo are often subject to the challenge of spatial confinement from neighboring cells and extracellular matrix (ECM) that are usually adhesive and deformable. Here, we showed that confinement makes initially quiescent round cells on soft adhesive substrates spread and migrate, exhibiting a phenotype similar to that of cells on unconfined stiff substrates. Interestingly, the confinement-induced cell spreading and migration exist widely in many cell types, and depend on formins, cell contractility and endonuclear YAP-TEAD interaction. Finally, we demonstrated the nucleus is a mechanosensor independent of ECM rigidity, and its flattening alone is sufficient to trigger YAP nuclear translocation, assembly of focal adhesions and stress fibers, cell spreading and migration. Thus, our findings revealed a new inside-out mechanism through which the nucleus directly detects and responds to external mechanical confinement, and could have important implications for cell migration in crowded micro-environments during cancer metastasis, wound healing and embryonic development.
Collapse
Affiliation(s)
- Ran Rao
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China; Current Address: Department of Pathology, the First Affilliated Hospital of Anhui Medical University, Hefei, 230001, Anhui, China
| | - Haoxiang Yang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Kailong Qiu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Min Xu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Hao Liu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Jinghao Shen
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Weihao Wang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Runjie Nie
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Huan Chen
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Hongyuan Jiang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China; CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230026, Anhui, China.
| |
Collapse
|
2
|
Hollander S, Guo Y, Wolfenson H, Zaritsky A. Spatiotemporal analysis of F-actin polymerization with micropillar arrays reveals synchronization between adhesion sites. Mol Biol Cell 2024; 35:br23. [PMID: 39441710 PMCID: PMC11656478 DOI: 10.1091/mbc.e24-06-0276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
We repurposed micropillar arrays to quantify spatiotemporal inter-adhesion communication. Following the observation that integrin adhesions formed around pillar tops we relied on the precise repetitive spatial control of the pillars to reliably monitor F-actin dynamics in mouse embryonic fibroblasts as a model for spatiotemporal adhesion-related intracellular signaling. Using correlation-based analyses, we revealed localized information flows propagating between adjacent pillars that were integrated over space and time to synchronize the adhesion dynamics within the entire cell. Probing the mechanical regulation, we discovered that stiffer pillars or partial actomyosin contractility inhibition enhances inter-adhesion F-actin synchronization, and that inhibition of Arp2/3, but not formin, reduces synchronization. Our results suggest that adhesions can communicate and highlight the potential of using micropillar arrays as a tool to measure spatiotemporal intracellular signaling.
Collapse
Affiliation(s)
- Sarit Hollander
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Yuanning Guo
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Assaf Zaritsky
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
3
|
Zhang Y, Rao Y, Lu J, Wang J, Ker DFE, Zhou J, Wang DM. The influence of biophysical niche on tumor-associated macrophages in liver cancer. Hepatol Commun 2024; 8:e0569. [PMID: 39470328 PMCID: PMC11524744 DOI: 10.1097/hc9.0000000000000569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/30/2024] [Indexed: 10/30/2024] Open
Abstract
HCC, the most common type of primary liver cancer, is a leading cause of cancer-related mortality worldwide. Although the advancement of immunotherapies by immune checkpoint inhibitors (ICIs) that target programmed cell death 1 or programmed cell death 1-ligand 1 has revolutionized the treatment for HCC, the majority is still not beneficial. Accumulating evidence has pointed out that the potent immunosuppressive tumor microenvironment in HCC poses a great challenge to ICI therapeutic efficacy. As a key component in tumor microenvironment, tumor-associated macrophages (TAMs) play vital roles in HCC development, progression, and ICI low responsiveness. Mechanistically, TAM can promote cancer invasion and metastasis, angiogenesis, epithelial-mesenchymal transition, maintenance of stemness, and most importantly, immunosuppression. Targeting TAMs, therefore, represents an opportunity to enhance the ICI therapeutic efficacy in patients with HCC. While previous research has primarily focused on biochemical cues influencing macrophages, emerging evidence highlights the critical role of biophysical signals, such as substrate stiffness, topography, and external forces. In this review, we summarize the influence of biophysical characteristics within the tumor microenvironment that regulate the phenotype and function of TAMs in HCC pathogenesis and progression. We also explore the possible mechanisms and discuss the potential of manipulating biophysical cues in regulating TAM for HCC therapy. By gaining a deeper understanding of how macrophages sense and respond to mechanical forces, we may potentially usher in a path toward a curative approach for combinatory cancer immunotherapies.
Collapse
Affiliation(s)
- Ying Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiahuan Lu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiyu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dai Fei Elmer Ker
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR, China
| | - Jingying Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| |
Collapse
|
4
|
Qi B, Li Y, Zhao J, Zhang J, Zhang X, Chen G, Yang Z. Regulating Bacterial Culture through Tailored Silk Inverse Opal Scaffolds. Macromol Biosci 2024; 24:e2400238. [PMID: 38843881 DOI: 10.1002/mabi.202400238] [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: 05/15/2024] [Revised: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Controlling the growth of microbial consortia is of great significance in the biomedical field. Selective bacterial growth is achieved by fabricating silk inverse opal (SIO) scaffolds with varying pore sizes ranging from 0.3 to 4.5 µm. Pore size significantly influences the growth dynamics of bacteria in both single and mixed-strain cultures. Specially, the SIO-4.5 µm scaffold is observed to be more favorable for cultivating S. aureus, whereas the SIO-0.3 µm scaffold is more suitable for cultivating E. coli and P. aeruginosa. By adjusting the secondary conformation of silk fibroin, the stiffness of the SIO substrate will be altered, which results in the increase of bacteria on the SIO by 16 times compared with that on the silk fibroin film. Manipulating the pore size allows for the adjustment of the S. aureus to P. aeruginosa ratio from 0.8 to 9.3, highlighting the potential of this approach in regulating bacterial culture.
Collapse
Affiliation(s)
- Bei Qi
- School of Physical Science and Technology, Jiangsu Key Laboratory of Frontier Material Physics and Devices, Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, China
| | - Yitan Li
- National Engineering Research Center for Colloidal Materials, School of Chemistry and Chemical Engineering, Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Junyan Zhao
- College of Chemistry, Chemical Engineering and Materials Science, Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, China
| | - Jiapeng Zhang
- School of Physical Science and Technology, Jiangsu Key Laboratory of Frontier Material Physics and Devices, Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, China
| | - Xiaohua Zhang
- School of Physical Science and Technology, Jiangsu Key Laboratory of Frontier Material Physics and Devices, Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, China
| | - Gaojian Chen
- College of Chemistry, Chemical Engineering and Materials Science, Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, China
| | - Zhaohui Yang
- School of Physical Science and Technology, Jiangsu Key Laboratory of Frontier Material Physics and Devices, Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, China
| |
Collapse
|
5
|
Burgess JK, Gosens R. Mechanotransduction and the extracellular matrix: Key drivers of lung pathologies and drug responsiveness. Biochem Pharmacol 2024; 228:116255. [PMID: 38705536 DOI: 10.1016/j.bcp.2024.116255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
The lung is a biomechanically active organ, with multiscale mechanical forces impacting the organ, tissue and cellular responses within this microenvironment. In chronic lung diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and others, the structure of the lung is drastically altered impeding gas exchange. These changes are, in part, reflected in alterations in the composition, amount and organization of the extracellular matrix within the different lung compartments. The transmission of mechanical forces within lung tissue are broadcast by this complex mix of extracellular matrix components, in particular the collagens, elastin and proteoglycans and the crosslinking of these components. At both a macro and a micro level, the mechanical properties of the microenvironment have a key regulatory role in ascertaining cellular responses and the function of the lung. Cells adhere to, and receive signals from, the extracellular matrix through a number of different surface receptors and complexes which are important for mechanotransduction. This review summarizes the multiscale mechanics in the lung and how the mechanical environment changes in lung disease and aging. We then examine the role of mechanotransduction in driving cell signaling events in lung diseases and finish with a future perspective of the need to consider how such forces may impact pharmacological responsiveness in lung diseases.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| | - Reinoud Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
6
|
Elgin S, Silberman E, Shapira A, Dvir T. Customizable Hydrogel Coating of ECM-Based Microtissues for Improved Cell Retention and Tissue Integrity. Gels 2024; 10:515. [PMID: 39195044 DOI: 10.3390/gels10080515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 08/29/2024] Open
Abstract
Overcoming the oxygen diffusion limit of approximately 200 µm remains one of the most significant and intractable challenges to be overcome in tissue engineering. The fabrication of hydrogel microtissues and their assembly into larger structures may provide a solution, though these constructs are not without their own drawbacks; namely, these hydrogels are rapidly degraded in vivo, and cells delivered via microtissues are quickly expelled from the area of action. Here, we report the development of an easily customized protocol for creating a protective, biocompatible hydrogel barrier around microtissues. We show that calcium carbonate nanoparticles embedded within an ECM-based microtissue diffuse outwards and, when then exposed to a solution of alginate, can be used to generate a coated layer around the tissue. We further show that this technique can be fine-tuned by adjusting numerous parameters, granting us full control over the thickness of the hydrogel coating layer. The microtissues' protective hydrogel functioned as hypothesized in both in vitro and in vivo testing by preventing the cells inside the tissue from escaping and protecting the microdroplets against external degradation. This technology may provide microtissues with customized properties for use as sources of regenerative therapies.
Collapse
Affiliation(s)
- Shani Elgin
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eric Silberman
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- The Sagol Center for Regenerative Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Center for Nanoscience & Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Assaf Shapira
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- The Sagol Center for Regenerative Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Center for Nanoscience & Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tal Dvir
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- The Sagol Center for Regenerative Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Center for Nanoscience & Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School for Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
7
|
Jain K, Pandey A, Wang H, Chung T, Nemati A, Kanchanawong P, Sheetz MP, Cai H, Changede R. TiO 2 Nano-Biopatterning Reveals Optimal Ligand Presentation for Cell-Matrix Adhesion Formation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309284. [PMID: 38340044 PMCID: PMC11126362 DOI: 10.1002/adma.202309284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Nanoscale organization of transmembrane receptors is critical for cellular functions, enabled by the nanoscale engineering of bioligand presentation. Previously, a spatial threshold of ≤60 nm for integrin binding ligands in cell-matrix adhesion is demonstrated using monoliganded gold nanoparticles. However, the ligand geometric arrangement is limited to hexagonal arrays of monoligands, while plasmonic quenching limits further investigation by fluorescence-based high-resolution imaging. Here, these limitations are overcome with dielectric TiO2 nanopatterns, eliminating fluorescence quenching, thus enabling super-resolution fluorescence microscopy on nanopatterns. By dual-color super-resolution imaging, high precision and consistency among nanopatterns, bioligands, and integrin nanoclusters are observed, validating the high quality and integrity of both nanopattern functionalization and passivation. By screening TiO2 nanodiscs with various diameters, an increase in fibroblast cell adhesion, spreading area, and Yes-associated protein (YAP) nuclear localization on 100 nm diameter compared with smaller diameters was observed. Focal adhesion kinase is identified as the regulatory signal. These findings explore the optimal ligand presentation when the minimal requirements are sufficiently fulfilled in the heterogenous extracellular matrix network of isolated binding regions with abundant ligands. Integration of high-fidelity nano-biopatterning with super-resolution imaging allows precise quantitative studies to address early signaling events in response to receptor clustering and their nanoscale organization.
Collapse
Affiliation(s)
- Kashish Jain
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Ashish Pandey
- Tech4Health Institute and Department of Radiology, NYU Langone Health, New York, NY, USA
| | - Hao Wang
- Tech4Health Institute and Department of Radiology, NYU Langone Health, New York, NY, USA
| | - Taerin Chung
- Tech4Health Institute and Department of Radiology, NYU Langone Health, New York, NY, USA
| | - Arash Nemati
- Tech4Health Institute and Department of Radiology, NYU Langone Health, New York, NY, USA
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Michael P. Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Molecular Mechanomedicine Program, Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, TX, USA
| | - Haogang Cai
- Tech4Health Institute and Department of Radiology, NYU Langone Health, New York, NY, USA
- Department of Biomedical Engineering, New York University, Brooklyn, NY, USA
| | - Rishita Changede
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- TeOra Pte. Ltd, Singapore, Singapore
| |
Collapse
|
8
|
Shi L, Nadjar-Boger E, Jafarinia H, Carlier A, Wolfenson H. YAP mediates apoptosis through failed integrin adhesion reinforcement. Cell Rep 2024; 43:113811. [PMID: 38393944 DOI: 10.1016/j.celrep.2024.113811] [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: 06/19/2023] [Revised: 12/26/2023] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Extracellular matrix (ECM) rigidity is a major effector of cell fate decisions. Whereas cell proliferation on stiff matrices, wherein Yes-associated protein (YAP) plays a pivotal role, is well documented, activation of apoptosis in response to soft matrices is poorly understood. Here, we show that YAP drives the apoptotic decision as well. We find that in cells on soft matrices, YAP is recruited to small adhesions, phosphorylated at the Y357 residue, and translocated into the nucleus, ultimately leading to apoptosis. In contrast, Y357 phosphorylation levels are dramatically low in large adhesions on stiff matrices. Furthermore, mild attenuation of actomyosin contractility allows adhesion growth on soft matrices, leading to reduced Y357 phosphorylation levels and resulting in cell growth. These findings indicate that failed adhesion reinforcement drives rigidity-dependent apoptosis through YAP and that this decision is not determined solely by ECM rigidity but rather by the balance between cellular forces and ECM rigidity.
Collapse
Affiliation(s)
- Lidan Shi
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Elisabeth Nadjar-Boger
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Hamidreza Jafarinia
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
9
|
Mittal N, Michels EB, Massey AE, Qiu Y, Royer-Weeden SP, Smith BR, Cartagena-Rivera AX, Han SJ. Myosin-independent stiffness sensing by fibroblasts is regulated by the viscoelasticity of flowing actin. COMMUNICATIONS MATERIALS 2024; 5:6. [PMID: 38741699 PMCID: PMC11090405 DOI: 10.1038/s43246-024-00444-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 01/02/2024] [Indexed: 05/16/2024]
Abstract
The stiffness of the extracellular matrix induces differential tension within integrin-based adhesions, triggering differential mechanoresponses. However, it has been unclear if the stiffness-dependent differential tension is induced solely by myosin activity. Here, we report that in the absence of myosin contractility, 3T3 fibroblasts still transmit stiffness-dependent differential levels of traction. This myosin-independent differential traction is regulated by polymerizing actin assisted by actin nucleators Arp2/3 and formin where formin has a stronger contribution than Arp2/3 to both traction and actin flow. Intriguingly, despite only slight changes in F-actin flow speed observed in cells with the combined inhibition of Arp2/3 and myosin compared to cells with sole myosin inhibition, they show a 4-times reduction in traction than cells with myosin-only inhibition. Our analyses indicate that traditional models based on rigid F-actin are inadequate for capturing such dramatic force reduction with similar actin flow. Instead, incorporating the F-actin network's viscoelastic properties is crucial. Our new model including the F-actin viscoelasticity reveals that Arp2/3 and formin enhance stiffness sensitivity by mechanically reinforcing the F-actin network, thereby facilitating more effective transmission of flow-induced forces. This model is validated by cell stiffness measurement with atomic force microscopy and experimental observation of model-predicted stiffness-dependent actin flow fluctuation.
Collapse
Affiliation(s)
- Nikhil Mittal
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
- Health Research Institute, Michigan Technological University, Houghton, MI, USA
| | - Etienne B. Michels
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
| | - Andrew E. Massey
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Yunxiu Qiu
- Department of Biomedical Engineering, Michigan State University, Lansing, MI, USA
| | - Shaina P. Royer-Weeden
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
| | - Bryan R. Smith
- Department of Biomedical Engineering, Michigan State University, Lansing, MI, USA
| | - Alexander X. Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Sangyoon J. Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, USA
- Health Research Institute, Michigan Technological University, Houghton, MI, USA
- Department of Mechanical Engineering and Engineering Mechanics, Michigan Technological University, Houghton, MI, USA
| |
Collapse
|
10
|
Griveau L, Bouvet M, Christin E, Paret C, Lecoq L, Radix S, Laumonier T, Sohier J, Gache V. Synthetic injectable and porous hydrogels for the formation of skeletal muscle fibers: Novel perspectives for the acellular repair of substantial volumetric muscle loss. J Tissue Eng 2024; 15:20417314241283148. [PMID: 39502329 PMCID: PMC11536390 DOI: 10.1177/20417314241283148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/28/2024] [Indexed: 11/08/2024] Open
Abstract
In severe skeletal muscle damage, muscle tissue regeneration process has to face the loss of resident muscle stem cells (MuSCs) and the lack of connective tissue necessary to guide the regeneration process. Biocompatible and standardized 3D structures that can be injected to the muscle injury site, conforming to the defect shape while actively guiding the repair process, holds great promise for skeletal muscle tissue regeneration. In this study, we explore the use of an injectable and porous lysine dendrimer/polyethylene glycol (DGL/PEG) hydrogel as an acellular support for skeletal muscle regeneration. We adjusted the DGL/PEG composition to achieve a stiffness conducive to the attachment and proliferation of murine immortalized myoblasts and human primary muscle stems cells, sustaining the formation and maturation of muscle fibers in vitro. We then evaluated the potential of one selected "myogenic-porous hydrogel" as a supportive structure for muscle repair in a large tibialis anterior muscle defect in rats. This injectable and porous formulation filled the defect, promoting rapid cellularization with the presence of endothelial cells, macrophages, and myoblasts, thereby supporting neo-myogenesis more specifically at the interface between the wound edges and the hydrogel. The selected porous DGL/PEG hydrogel acted as a guiding scaffold at the periphery of the defect, facilitating the formation and anchorage of aligned muscle fibers 21 days after injury. Overall, our results indicate DGL/PEG porous injectable hydrogel potential to create a pro-regenerative environment for muscle cells after large skeletal muscle injuries, paving the way for acellular treatment in regenerative muscle medicine.
Collapse
Affiliation(s)
- Louise Griveau
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS, University of Lyon, Claude Bernard University Lyon 1, UMR5305 LBTI, Lyon, France
| | - Marion Bouvet
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS, University of Lyon, Claude Bernard University Lyon 1, UMR5305 LBTI, Lyon, France
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - Emilie Christin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - Cloé Paret
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - Lauriane Lecoq
- Molecular Microbiology and Structural Biochemistry, Labex Ecofect, UMR 5086 CNRS/Université de Lyon, Lyon, France
| | - Sylvie Radix
- Universite Claude Bernard Lyon 1, CNRS UMR 5246 Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB, Lyon, France
| | - Thomas Laumonier
- Department of Orthopedic Surgery & Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland
| | - Jerome Sohier
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS, University of Lyon, Claude Bernard University Lyon 1, UMR5305 LBTI, Lyon, France
| | - Vincent Gache
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, INSERM U1513, CNRS UMR 5261, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| |
Collapse
|
11
|
Cheng L, Yue H, Zhang H, Liu Q, Du L, Liu X, Xie J, Shen Y. The influence of microenvironment stiffness on endothelial cell fate: Implication for occurrence and progression of atherosclerosis. Life Sci 2023; 334:122233. [PMID: 37918628 DOI: 10.1016/j.lfs.2023.122233] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
Atherosclerosis, the primary cause of cardiovascular diseases (CVDs), is characterized by phenotypic changes in fibrous proliferation, chronic inflammation and lipid accumulation mediated by vascular endothelial cells (ECs) and vascular smooth muscle cells (SMCs) which are correlated with the stiffening and ectopic remodeling of local extracellular matrix (ECM). The native residents, ECs and SMCs, are not only affected by various chemical factors including inflammatory mediators and chemokines, but also by a range of physical stimuli, such as shear stress and ECM stiffness, presented in the microenvironmental niche. Especially, ECs, as a semi-selective barrier, can sense mechanical forces, respond quickly to changes in mechanical loading and provide context-specific adaptive responses to restore homeostasis. However, blood arteries undergo stiffening and lose their elasticity with age. Reports have shown that the ECM stiffening could influence EC fate by changing the cell adhesion, spreading, proliferation, cell to cell contact, migration and even communication with SMCs. The cell behaviour changes mediated by ECM stiffening are dependent on the activation of a signaling cascade of mechanoperception and mechanotransduction. Although the substantial evidence directly indicates the importance of ECM stiffening on the native ECs, the understanding about this complex interplay is still largely limited. In this review, we systematically summarize the roles of ECM stiffening on the behaviours of endothelial cells and elucidate the underlying details in biological mechanism, aiming to provide the process of how ECs integrate ECM mechanics and the highlights for bioaffinity of tissue-specific engineered scaffolds.
Collapse
Affiliation(s)
- Lin Cheng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Hongyan Yue
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Huaiyi Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Qiao Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Lingyu Du
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; JinFeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
12
|
Brondolin M, Herzog D, Sultan S, Warburton F, Vigilante A, Knight RD. Migration and differentiation of muscle stem cells are coupled by RhoA signalling during regeneration. Open Biol 2023; 13:230037. [PMID: 37726092 PMCID: PMC10508982 DOI: 10.1098/rsob.230037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023] Open
Abstract
Skeletal muscle is highly regenerative and is mediated by a population of migratory adult muscle stem cells (muSCs). Effective muscle regeneration requires a spatio-temporally regulated response of the muSC population to generate sufficient muscle progenitor cells that then differentiate at the appropriate time. The relationship between muSC migration and cell fate is poorly understood and it is not clear how forces experienced by migrating cells affect cell behaviour. We have used zebrafish to understand the relationship between muSC cell adhesion, behaviour and fate in vivo. Imaging of pax7-expressing muSCs as they respond to focal injuries in trunk muscle reveals that they migrate by protrusive-based means. By carefully characterizing their behaviour in response to injury we find that they employ an adhesion-dependent mode of migration that is regulated by the RhoA kinase ROCK. Impaired ROCK activity results in reduced expression of cell cycle genes and increased differentiation in regenerating muscle. This correlates with changes to focal adhesion dynamics and migration, revealing that ROCK inhibition alters the interaction of muSCs to their local environment. We propose that muSC migration and differentiation are coupled processes that respond to changes in force from the environment mediated by RhoA signalling.
Collapse
Affiliation(s)
- Mirco Brondolin
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, London SE1 9RT, UK
| | - Dylan Herzog
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, London SE1 9RT, UK
| | - Sami Sultan
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, London SE1 9RT, UK
| | - Fiona Warburton
- Oral Clinical Research Unit, King's College London, London, London SE1 9RT, UK
| | | | - Robert D. Knight
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, London SE1 9RT, UK
| |
Collapse
|
13
|
Saraswathibhatla A, Indana D, Chaudhuri O. Cell-extracellular matrix mechanotransduction in 3D. Nat Rev Mol Cell Biol 2023; 24:495-516. [PMID: 36849594 PMCID: PMC10656994 DOI: 10.1038/s41580-023-00583-1] [Citation(s) in RCA: 265] [Impact Index Per Article: 132.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 03/01/2023]
Abstract
Mechanical properties of extracellular matrices (ECMs) regulate essential cell behaviours, including differentiation, migration and proliferation, through mechanotransduction. Studies of cell-ECM mechanotransduction have largely focused on cells cultured in 2D, on top of elastic substrates with a range of stiffnesses. However, cells often interact with ECMs in vivo in a 3D context, and cell-ECM interactions and mechanisms of mechanotransduction in 3D can differ from those in 2D. The ECM exhibits various structural features as well as complex mechanical properties. In 3D, mechanical confinement by the surrounding ECM restricts changes in cell volume and cell shape but allows cells to generate force on the matrix by extending protrusions and regulating cell volume as well as through actomyosin-based contractility. Furthermore, cell-matrix interactions are dynamic owing to matrix remodelling. Accordingly, ECM stiffness, viscoelasticity and degradability often play a critical role in regulating cell behaviours in 3D. Mechanisms of 3D mechanotransduction include traditional integrin-mediated pathways that sense mechanical properties and more recently described mechanosensitive ion channel-mediated pathways that sense 3D confinement, with both converging on the nucleus for downstream control of transcription and phenotype. Mechanotransduction is involved in tissues from development to cancer and is being increasingly harnessed towards mechanotherapy. Here we discuss recent progress in our understanding of cell-ECM mechanotransduction in 3D.
Collapse
Affiliation(s)
| | - Dhiraj Indana
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
| |
Collapse
|
14
|
Jones DL, Hallström GF, Jiang X, Locke RC, Evans MK, Bonnevie ED, Srikumar A, Leahy TP, Nijsure MP, Boerckel JD, Mauck RL, Dyment NA. Mechanoepigenetic regulation of extracellular matrix homeostasis via Yap and Taz. Proc Natl Acad Sci U S A 2023; 120:e2211947120. [PMID: 37216538 PMCID: PMC10235980 DOI: 10.1073/pnas.2211947120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Cells integrate mechanical cues to direct fate specification to maintain tissue function and homeostasis. While disruption of these cues is known to lead to aberrant cell behavior and chronic diseases, such as tendinopathies, the underlying mechanisms by which mechanical signals maintain cell function are not well understood. Here, we show using a model of tendon de-tensioning that loss of tensile cues in vivo acutely changes nuclear morphology, positioning, and expression of catabolic gene programs, resulting in subsequent weakening of the tendon. In vitro studies using paired ATAC/RNAseq demonstrate that the loss of cellular tension rapidly reduces chromatin accessibility in the vicinity of Yap/Taz genomic targets while also increasing expression of genes involved in matrix catabolism. Concordantly, the depletion of Yap/Taz elevates matrix catabolic expression. Conversely, overexpression of Yap results in a reduction of chromatin accessibility at matrix catabolic gene loci, while also reducing transcriptional levels. The overexpression of Yap not only prevents the induction of this broad catabolic program following a loss of cellular tension, but also preserves the underlying chromatin state from force-induced alterations. Taken together, these results provide novel mechanistic details by which mechanoepigenetic signals regulate tendon cell function through a Yap/Taz axis.
Collapse
Affiliation(s)
- Dakota L. Jones
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
| | - Grey F. Hallström
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
| | - Xi Jiang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
| | - Ryan C. Locke
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Translational Musculoskeletal Research Center, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA19104
| | - Mary Kate Evans
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Edward D. Bonnevie
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
- Translational Musculoskeletal Research Center, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA19104
| | - Anjana Srikumar
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
| | - Thomas P. Leahy
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Madhura P. Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA19104
| | - Joel D. Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA19104
| | - Robert L. Mauck
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Translational Musculoskeletal Research Center, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA19104
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA19104
| | - Nathaniel A. Dyment
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
15
|
Higgins G, Higgins F, Peres J, Lang DM, Abdalrahman T, Zaman MH, Prince S, Franz T. Intracellular mechanics and TBX3 expression jointly dictate the spreading mode of melanoma cells in 3D environments. Exp Cell Res 2023; 428:113633. [PMID: 37172754 DOI: 10.1016/j.yexcr.2023.113633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Cell stiffness and T-box transcription factor 3 (TBX3) expression have been identified as biomarkers of melanoma metastasis in 2D environments. This study aimed to determine how mechanical and biochemical properties of melanoma cells change during cluster formation in 3D environments. Vertical growth phase (VGP) and metastatic (MET) melanoma cells were embedded in 3D collagen matrices of 2 and 4 mg/ml collagen concentrations, representing low and high matrix stiffness. Mitochondrial fluctuation, intracellular stiffness, and TBX3 expression were quantified before and during cluster formation. In isolated cells, mitochondrial fluctuation decreased and intracellular stiffness increased with increase in disease stage from VGP to MET and increased matrix stiffness. TBX3 was highly expressed in soft matrices but diminished in stiff matrices for VGP and MET cells. Cluster formation of VGP cells was excessive in soft matrices but limited in stiff matrices, whereas for MET cells it was limited in soft and stiff matrices. In soft matrices, VGP cells did not change the intracellular properties, whereas MET cells exhibited increased mitochondrial fluctuation and decreased TBX3 expression. In stiff matrices, mitochondrial fluctuation and TBX3 expression increased in VGP and MET, and intracellular stiffness increased in VGP but decreased in MET cells. The findings suggest that soft extracellular environments are more favourable for tumour growth, and high TBX3 levels mediate collective cell migration and tumour growth in the earlier VGP disease stage but play a lesser role in the later metastatic stage of melanoma.
Collapse
Affiliation(s)
- Ghodeejah Higgins
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Faatiemah Higgins
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Jade Peres
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Dirk M Lang
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Tamer Abdalrahman
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Muhammad H Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Sharon Prince
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Thomas Franz
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa; Bioengineering Science Research Group, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
16
|
Sumey JL, Johnston PC, Harrell AM, Caliari SR. Hydrogel mechanics regulate fibroblast DNA methylation and chromatin condensation. Biomater Sci 2023; 11:2886-2897. [PMID: 36880435 PMCID: PMC10329270 DOI: 10.1039/d2bm02058k] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Cellular mechanotransduction plays a central role in fibroblast activation during fibrotic disease progression, leading to increased tissue stiffness and reduced organ function. While the role of epigenetics in disease mechanotransduction has begun to be appreciated, little is known about how substrate mechanics, particularly the timing of mechanical inputs, regulate epigenetic changes such as DNA methylation and chromatin reorganization during fibroblast activation. In this work, we engineered a hyaluronic acid hydrogel platform with independently tunable stiffness and viscoelasticity to model normal (storage modulus, G' ∼ 0.5 kPa, loss modulus, G'' ∼ 0.05 kPa) to increasingly fibrotic (G' ∼ 2.5 and 8 kPa, G'' ∼ 0.05 kPa) lung mechanics. Human lung fibroblasts exhibited increased spreading and nuclear localization of myocardin-related transcription factor-A (MRTF-A) with increasing substrate stiffness within 1 day, with these trends holding steady for longer cultures. However, fibroblasts displayed time-dependent changes in global DNA methylation and chromatin organization. Fibroblasts initially displayed increased DNA methylation and chromatin decondensation on stiffer hydrogels, but both of these measures decreased with longer culture times. To investigate how culture time affected the responsiveness of fibroblast nuclear remodeling to mechanical signals, we engineered hydrogels amenable to in situ secondary crosslinking, enabling a transition from a compliant substrate mimicking normal tissue to a stiffer substrate resembling fibrotic tissue. When stiffening was initiated after only 1 day of culture, fibroblasts rapidly responded and displayed increased DNA methylation and chromatin decondensation, similar to fibroblasts on static stiffer hydrogels. Conversely, when fibroblasts experienced later stiffening at day 7, they showed no changes in DNA methylation and chromatin condensation, suggesting the induction of a persistent fibroblast phenotype. These results highlight the time-dependent nuclear changes associated with fibroblast activation in response to dynamic mechanical perturbations and may provide mechanisms to target for controlling fibroblast activation.
Collapse
Affiliation(s)
- Jenna L Sumey
- Department of Chemical Engineering, University of Virginia, USA.
| | | | | | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, USA.
- Department of Biomedical Engineering, University of Virginia, USA
| |
Collapse
|
17
|
Srivastava P, Romanazzo S, Kopecky C, Nemec S, Ireland J, Molley TG, Lin K, Jayathilaka PB, Pandzic E, Yeola A, Chandrakanthan V, Pimanda J, Kilian K. Defined Microenvironments Trigger In Vitro Gastrulation in Human Pluripotent Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203614. [PMID: 36519269 PMCID: PMC9929265 DOI: 10.1002/advs.202203614] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/19/2022] [Indexed: 06/17/2023]
Abstract
Gastrulation is a stage in embryo development where three germ layers arise to dictate the human body plan. In vitro models of gastrulation have been demonstrated by treating pluripotent stem cells with soluble morphogens to trigger differentiation. However, in vivo gastrulation is a multistage process coordinated through feedback between soluble gradients and biophysical forces, with the multipotent epiblast transforming to the primitive streak followed by germ layer segregation. Here, the authors show how constraining pluripotent stem cells to hydrogel islands triggers morphogenesis that mirrors the stages preceding in vivo gastrulation, without the need for exogenous supplements. Within hours of initial seeding, cells display a contractile phenotype at the boundary, which leads to enhanced proliferation, yes-associated protein (YAP) translocation, epithelial to mesenchymal transition, and emergence of SRY-box transcription factor 17 (SOX17)+ T/BRACHYURY+ cells. Molecular profiling and pathway analysis reveals a role for mechanotransduction-coupled wingless-type (WNT) signaling in orchestrating differentiation, which bears similarities to processes observed in whole organism models of development. After two days, the colonies form multilayered aggregates, which can be removed for further growth and differentiation. This approach demonstrates how materials alone can initiate gastrulation, thereby providing in vitro models of development and a tool to support organoid bioengineering efforts.
Collapse
Affiliation(s)
- Pallavi Srivastava
- School of ChemistryAustralian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
- School of Biomedical SciencesUniversity of New South WalesSydneyNSW2052Australia
- Adult Cancer ProgramSchool of Clinical Medicine, Lowy Cancer Research CentreUNSW SydneySydneyNSW2052Australia
| | - Sara Romanazzo
- School of ChemistryAustralian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
| | - Chantal Kopecky
- School of ChemistryAustralian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
- Adult Cancer ProgramSchool of Clinical Medicine, Lowy Cancer Research CentreUNSW SydneySydneyNSW2052Australia
| | - Stephanie Nemec
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Jake Ireland
- School of ChemistryAustralian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
| | - Thomas G. Molley
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Kang Lin
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Pavithra B. Jayathilaka
- School of ChemistryAustralian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
| | - Elvis Pandzic
- Katharina Gaus Light Microscopy FacilityMark Wainwright Analytical CentreUniversity of New South WalesSydneyNSW2052Australia
| | - Avani Yeola
- Adult Cancer ProgramSchool of Clinical Medicine, Lowy Cancer Research CentreUNSW SydneySydneyNSW2052Australia
| | - Vashe Chandrakanthan
- School of Biomedical SciencesUniversity of New South WalesSydneyNSW2052Australia
- Adult Cancer ProgramSchool of Clinical Medicine, Lowy Cancer Research CentreUNSW SydneySydneyNSW2052Australia
| | - John Pimanda
- School of Biomedical SciencesUniversity of New South WalesSydneyNSW2052Australia
- Adult Cancer ProgramSchool of Clinical Medicine, Lowy Cancer Research CentreUNSW SydneySydneyNSW2052Australia
- Department of HaematologyPrince of Wales HospitalRandwickNSW2031Australia
| | - Kristopher Kilian
- School of ChemistryAustralian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
- Adult Cancer ProgramSchool of Clinical Medicine, Lowy Cancer Research CentreUNSW SydneySydneyNSW2052Australia
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
| |
Collapse
|
18
|
Dashtarzheneh AK, Afrashtehpour A, Ramesh BS, Loizidou M. Harvestable tumour spheroids initiated in a gelatin-carboxymethyl cellulose hydrogel for cancer targeting and imaging with fluorescent gold nanoclusters. IN VITRO MODELS 2022; 1:437-446. [PMID: 39872615 PMCID: PMC11756458 DOI: 10.1007/s44164-022-00033-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/11/2022] [Accepted: 08/18/2022] [Indexed: 01/30/2025]
Abstract
Cancer cell spheroids are the simplest 3D in vitro cancer models and have been extensively used for cancer research. More recently, models have been becoming complex, with the introduction of a matrix and non-cancer cell types to mimic specific tumour aspects. However, applying drugs or agents in matrix-embedded cancer spheroids can be problematic. Most matrices can impede and also bind drugs or visualizing agents non-specifically, in the vicinity of the embedded spheroids. This may interfere with imaging or further analysis without breaking apart the 3D model into its constituents. Here, we developed a combined gelatin-carboxymethyl cellulose (G-CMC) hydrogel for initiating cancer spheroids that enabled intact harvesting pre/post treatment for further investigation, such as targeting and imaging. We combined CMC (1.25%) and gelatin (2.5%) at 25 °C and initiated polymerisation after autoclaving (121 °C) to obtain a mechanical strength (sheer stress) of 38 Pas versus 1.28 Pas for CMC alone. These matrix conditions facilitated separation of the spheroids from the G-CMC, using low centrifugation (100 g). We described growth of colorectal and breast cancer spheroids within the G-CMC matrix (with average diameters of 220 mm and 180 μm for representative cell lines HT29 and MCF7 at 10 days, respectively). As the cancer cells express the surface biomarker calreticulin (CRT), we manufactured anti-calreticulin IgG (anti-CRT) conjugated to fluorescent gold nanoclusters (anti-CRT-AuNC) as a probe. We harvested cancer spheroids and incubated live with the nanoclusters. Imaging demonstrated strong binding of CRT-targeted AuNCs compared to control AuNCs. This novel model preserves cancer spheroid integrity upon isolation and is well suited for targeted imaging and drug delivery of cancer in 3D.
Collapse
Affiliation(s)
- Ashkan Kamali Dashtarzheneh
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London, UK
| | - Amir Afrashtehpour
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London, UK
| | - Bala Subramaniyam Ramesh
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London, UK
| | - Marilena Loizidou
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London, UK
| |
Collapse
|
19
|
Rackow AR, Nagel DJ, Zapas G, Clough RS, Sime PJ, Kottmann RM. The Novel Small Molecule BTB Inhibits Pro-Fibrotic Fibroblast Behavior though Inhibition of RhoA Activity. Int J Mol Sci 2022; 23:11946. [PMID: 36233248 PMCID: PMC9569993 DOI: 10.3390/ijms231911946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, chronic, interstitial lung disease with a poor prognosis. Although specific anti-fibrotic medications are now available, the median survival time following diagnosis remains very low, and new therapies are urgently needed. To uncover novel therapeutic targets, we examined how biochemical properties of the fibrotic lung are different from the healthy lung. Previous work identified lactate as a metabolite that is upregulated in IPF lung tissue. Importantly, inhibition of the enzyme responsible for lactate production prevents fibrosis in vivo. Further studies revealed that fibrotic lesions of the lung experience a significant decline in tissue pH, likely due to the overproduction of lactate. It is not entirely clear how cells in the lung respond to changes in extracellular pH, but a family of proton sensing G-protein coupled receptors has been shown to be activated by reductions in extracellular pH. This work examines the expression profiles of proton sensing GPCRs in non-fibrotic and IPF-derived primary human lung fibroblasts. We identify TDAG8 as a proton sensing GPCR that is upregulated in IPF fibroblasts and that knockdown of TDAG8 dampens myofibroblast differentiation. To our surprise, BTB, a proposed positive allosteric modulator of TDAG8, inhibits myofibroblast differentiation. Our data suggest that BTB does not require TDAG8 to inhibit myofibroblast differentiation, but rather inhibits myofibroblast differentiation through suppression of RhoA mediated signaling. Our work highlights the therapeutic potential of BTB as an anti-fibrotic treatment and expands upon the importance of RhoA-mediated signaling pathways in the context of myofibroblast differentiation. Furthermore, this works also suggests that TDAG8 inhibition may have therapeutic relevance in the treatment of IPF.
Collapse
Affiliation(s)
- Ashley R. Rackow
- Division of Pulmonary Disease and Critical Care Medicine, University of Rochester Medical Center Rochester, Rochester, NY 14642, USA
| | - David J. Nagel
- Division of Pulmonary Disease and Critical Care Medicine, University of Rochester Medical Center Rochester, Rochester, NY 14642, USA
| | - Gregory Zapas
- Division of Pulmonary Disease and Critical Care Medicine, University of Rochester Medical Center Rochester, Rochester, NY 14642, USA
| | - Ryan S. Clough
- Department of Human Genetics, University of Utah Salt Lake City, Salt Lake City, UT 84112, USA
| | - Patricia J. Sime
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University Richmond, Richmond, VA 23284, USA
| | - R. Matthew Kottmann
- Division of Pulmonary Disease and Critical Care Medicine, University of Rochester Medical Center Rochester, Rochester, NY 14642, USA
| |
Collapse
|
20
|
Han Y, Cao Y, Lei H. Dynamic Covalent Hydrogels: Strong yet Dynamic. Gels 2022; 8:577. [PMID: 36135289 PMCID: PMC9498565 DOI: 10.3390/gels8090577] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/23/2022] Open
Abstract
Hydrogels are crosslinked polymer networks with time-dependent mechanical response. The overall mechanical properties are correlated with the dynamics of the crosslinks. Generally, hydrogels crosslinked by permanent chemical crosslinks are strong but static, while hydrogels crosslinked by physical interactions are weak but dynamic. It is highly desirable to create synthetic hydrogels that possess strong mechanical stability yet remain dynamic for various applications, such as drug delivery cargos, tissue engineering scaffolds, and shape-memory materials. Recently, with the introduction of dynamic covalent chemistry, the seemingly conflicting mechanical properties, i.e., stability and dynamics, have been successfully combined in the same hydrogels. Dynamic covalent bonds are mechanically stable yet still capable of exchanging, dissociating, or switching in response to external stimuli, empowering the hydrogels with self-healing properties, injectability and suitability for postprocessing and additive manufacturing. Here in this review, we first summarize the common dynamic covalent bonds used in hydrogel networks based on various chemical reaction mechanisms and the mechanical strength of these bonds at the single molecule level. Next, we discuss how dynamic covalent chemistry makes hydrogel materials more dynamic from the materials perspective. Furthermore, we highlight the challenges and future perspectives of dynamic covalent hydrogels.
Collapse
Affiliation(s)
- Yueying Han
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| | - Hai Lei
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing 210093, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| |
Collapse
|
21
|
Yan XR, Li J, Na XM, Li T, Xia YF, Zhou WQ, Ma GH. Mesenchymal Stem Cells Proliferation on Konjac Glucomannan Microcarriers: Effect of Rigidity. CHINESE JOURNAL OF POLYMER SCIENCE 2022. [DOI: 10.1007/s10118-022-2800-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
22
|
Konar S, Bolam SM, Coleman B, Dalbeth N, McGlashan SR, Leung S, Cornish J, Naot D, Musson DS. Changes in Physiological Tendon Substrate Stiffness Have Moderate Effects on Tendon-Derived Cell Growth and Immune Cell Activation. Front Bioeng Biotechnol 2022; 10:800748. [PMID: 35295642 PMCID: PMC8918575 DOI: 10.3389/fbioe.2022.800748] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/07/2022] [Indexed: 11/29/2022] Open
Abstract
Tendinopathy is characterised by pathological changes in tendon matrix composition, architecture, and stiffness, alterations in tendon resident cell characteristics, and fibrosis, with inflammation also emerging as an important factor in tendinopathy progression. The sequence of pathological changes in tendinopathy and the cellular effects of the deteriorating matrix are largely unknown. This study investigated the effects of substrate stiffness on tendon-derived cells (TDCs) and THP-1 macrophages using PDMS substrates representing physiological tendon stiffness (1.88 MPa), a stiff gel (3.17 MPa) and a soft gel (0.61 MPa). Human TDCs were cultured on the different gel substrates and on tissue culture plastic. Cell growth was determined by alamarBlue™ assay, cell morphology was analysed in f-actin labelled cells, and phenotypic markers were analysed by real-time PCR. We found that in comparison to TDCs growing on gels with physiological stiffness, cell growth increased on soft gels at 48 h (23%, p = 0.003). Cell morphology was similar on all three gels. SCX expression was slightly reduced on the soft gels (1.4-fold lower, p = 0.026) and COL1A1 expression increased on the stiff gels (2.2-fold, p = 0.041). Culturing THP-1 macrophages on soft gels induced increased expression of IL1B (2-fold, p = 0.018), and IL8 expression was inhibited on the stiffer gels (1.9-fold, p = 0.012). We also found that culturing TDCs on plastic increased cell growth, altered cell morphology, and inhibited the expression of SCX, SOX9, MMP3, and COL3. We conclude that TDCs and macrophages respond to changes in matrix stiffness. The magnitude of responses measured in TDCs were minor on the range of substrate stiffness tested by the gels. Changes in THP-1 macrophages suggested a more inflammatory phenotype on substrates with non-physiological stiffness. Although cell response to subtle variations in matrix stiffness was moderate, it is possible that these alterations may contribute to the onset and progression of tendinopathy.
Collapse
Affiliation(s)
- Subhajit Konar
- Department of Nutrition and Dietetics, University of Auckland, Auckland, New Zealand
| | - Scott M. Bolam
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Brendan Coleman
- Department of Orthopaedics, Middlemore Hospital, Auckland, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Sue R. McGlashan
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Sophia Leung
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Jillian Cornish
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Dorit Naot
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - David S. Musson
- Department of Nutrition and Dietetics, University of Auckland, Auckland, New Zealand
- *Correspondence: David S. Musson,
| |
Collapse
|
23
|
Spiaggia G, Taladriz-Blanco P, Septiadi D, Ortuso RD, Lee A, Trappe V, Rothen-Rutishauser B, Petri-Fink A. Aligned and Oriented Collagen Nanocomposite Fibers as Substrates to Activate Fibroblasts. ACS APPLIED BIO MATERIALS 2021; 4:8316-8324. [PMID: 35005948 DOI: 10.1021/acsabm.1c00844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Purified collagen possesses weak mechanical properties, hindering its broad application in tissue engineering. Strategies based on manipulating the hydrogel to induce fiber formation or incorporate nanomaterials have been proposed to overcome this issue. Herein, we use a microfluidic device to fabricate, for the first time, collagen hydrogels with aligned and oriented fibers doped with gold nanoparticles and carbon nanotubes. Results based on rheology, atomic force microscopy, and scanning electron microscopy reveal the formation of aligned and oriented collagen fibers possessing greater rigidity and stiffness on the doped hydrogels in comparison with native collagen. The mechanical properties of the hydrogels increased with the nanomaterial loading percentage and the stiffest formulations were those prepared in the presence of carbon nanotubes. We further evaluate the in vitro response of NIH-3T3 fibroblasts to the change in stiffness. The cells were found to be viable on all substrates with directional cell growth observed for the carbon nanotube-doped collagen fibers. No significant differences in the cell area, aspect ratio, and intensification of focal adhesions driven by the increase in stiffness were noted. Nonetheless, fibroblast proliferation and secretion of TGF-β1 were greater on the hydrogels doped with carbon nanotubes. This nanomaterial-collagen composite provides unique features for cell and tissue substrate applications.
Collapse
Affiliation(s)
- Giovanni Spiaggia
- Adolphe Merkle Institute, Université de Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Patricia Taladriz-Blanco
- Adolphe Merkle Institute, Université de Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Dedy Septiadi
- Adolphe Merkle Institute, Université de Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Roberto Diego Ortuso
- Adolphe Merkle Institute, Université de Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Aaron Lee
- Adolphe Merkle Institute, Université de Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Veronique Trappe
- Department of Physics, University of Fribourg, Chemin du Musée 3, 1700 Fribourg, Switzerland
| | | | - Alke Petri-Fink
- Adolphe Merkle Institute, Université de Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.,Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| |
Collapse
|
24
|
Modulation of stem cell response using biodegradable polyester films with different stiffness. BIOMEDICAL ENGINEERING ADVANCES 2021. [DOI: 10.1016/j.bea.2021.100007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
25
|
Hui E, Sumey JL, Caliari SR. Click-functionalized hydrogel design for mechanobiology investigations. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2021; 6:670-707. [PMID: 36338897 PMCID: PMC9631920 DOI: 10.1039/d1me00049g] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The advancement of click-functionalized hydrogels in recent years has coincided with rapid growth in the fields of mechanobiology, tissue engineering, and regenerative medicine. Click chemistries represent a group of reactions that possess high reactivity and specificity, are cytocompatible, and generally proceed under physiologic conditions. Most notably, the high level of tunability afforded by these reactions enables the design of user-controlled and tissue-mimicking hydrogels in which the influence of important physical and biochemical cues on normal and aberrant cellular behaviors can be independently assessed. Several critical tissue properties, including stiffness, viscoelasticity, and biomolecule presentation, are known to regulate cell mechanobiology in the context of development, wound repair, and disease. However, many questions still remain about how the individual and combined effects of these instructive properties regulate the cellular and molecular mechanisms governing physiologic and pathologic processes. In this review, we discuss several click chemistries that have been adopted to design dynamic and instructive hydrogels for mechanobiology investigations. We also chart a path forward for how click hydrogels can help reveal important insights about complex tissue microenvironments.
Collapse
Affiliation(s)
- Erica Hui
- Department of Chemical Engineering, University of Virginia, 102 Engineer's Way, Charlottesville, Virginia 22904, USA
| | - Jenna L Sumey
- Department of Chemical Engineering, University of Virginia, 102 Engineer's Way, Charlottesville, Virginia 22904, USA
| | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, 102 Engineer's Way, Charlottesville, Virginia 22904, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, USA
| |
Collapse
|
26
|
Zamprogno P, Thoma G, Cencen V, Ferrari D, Putz B, Michler J, Fantner GE, Guenat OT. Mechanical Properties of Soft Biological Membranes for Organ-on-a-Chip Assessed by Bulge Test and AFM. ACS Biomater Sci Eng 2021; 7:2990-2997. [PMID: 33651947 DOI: 10.1021/acsbiomaterials.0c00515] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Advanced in vitro models called "organ-on-a-chip" can mimic the specific cellular environment found in various tissues. Many of these models include a thin, sometimes flexible, membrane aimed at mimicking the extracellular matrix (ECM) scaffold of in vivo barriers. These membranes are often made of polydimethylsiloxane (PDMS), a silicone rubber that poorly mimics the chemical and physical properties of the basal membrane. However, the ECM and its mechanical properties play a key role in the homeostasis of a tissue. Here, we report about biological membranes with a composition and mechanical properties similar to those found in vivo. Two types of collagen-elastin (CE) membranes were produced: vitrified and nonvitrified (called "hydrogel membrane"). Their mechanical properties were characterized using the bulge test method. The results were compared using atomic force microscopy (AFM), a standard technique used to evaluate the Young's modulus of soft materials at the nanoscale. Our results show that CE membranes with stiffnesses ranging from several hundred of kPa down to 1 kPa can be produced by tuning the CE ratio, the production mode (vitrified or not), and/or certain parameters such as temperature. The Young's modulus can easily be determined using the bulge test. This method is a robust and reproducible to determine membrane stiffness, even for soft membranes, which are more difficult to assess by AFM. Assessment of the impact of substrate stiffness on the spread of human fibroblasts on these surfaces showed that cell spread is lower on softer surfaces than on stiffer surfaces.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Giuditta Thoma
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Veronika Cencen
- Laboratory for Bio- and Nano- Instrumentation, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Dario Ferrari
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Barbara Putz
- Laboratory for Mechanics of Materials and Nanostructures, EMPA Swiss Federal Laboratories for Materials Science and Technology, Thun 3602, Switzerland
| | - Johann Michler
- Laboratory for Mechanics of Materials and Nanostructures, EMPA Swiss Federal Laboratories for Materials Science and Technology, Thun 3602, Switzerland
| | - Georg E Fantner
- Laboratory for Bio- and Nano- Instrumentation, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern 3008, Switzerland.,Department of General Thoracic Surgery, University Hospital of Bern, Bern 3008, Switzerland
| |
Collapse
|
27
|
Jones DL, Meridew JA, Link PA, Ducharme MT, Lydon KL, Choi KM, Caporarello N, Tan Q, Diaz Espinosa AM, Xiong Y, Lee JH, Ye Z, Yan H, Ordog T, Ligresti G, Varelas X, Tschumperlin DJ. ZNF416 is a pivotal transcriptional regulator of fibroblast mechanoactivation. J Cell Biol 2021; 220:211825. [PMID: 33625469 PMCID: PMC7918622 DOI: 10.1083/jcb.202007152] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/13/2020] [Accepted: 01/26/2021] [Indexed: 01/18/2023] Open
Abstract
Matrix stiffness is a central regulator of fibroblast function. However, the transcriptional mechanisms linking matrix stiffness to changes in fibroblast phenotype are incompletely understood. Here, we evaluated the effect of matrix stiffness on genome-wide chromatin accessibility in freshly isolated lung fibroblasts using ATAC-seq. We found higher matrix stiffness profoundly increased global chromatin accessibility relative to lower matrix stiffness, and these alterations were in close genomic proximity to known profibrotic gene programs. Motif analysis of these regulated genomic loci identified ZNF416 as a putative mediator of fibroblast stiffness responses. Genome occupancy analysis using ChIP-seq confirmed that ZNF416 occupies a broad range of genes implicated in fibroblast activation and tissue fibrosis, with relatively little overlap in genomic occupancy with other mechanoresponsive and profibrotic transcriptional regulators. Using loss- and gain-of-function studies, we demonstrated that ZNF416 plays a critical role in fibroblast proliferation, extracellular matrix synthesis, and contractile function. Together, these observations identify ZNF416 as novel mechano-activated transcriptional regulator of fibroblast biology.
Collapse
Affiliation(s)
- Dakota L Jones
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Jeffrey A Meridew
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Patrick A Link
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Merrick T Ducharme
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Katherine L Lydon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Kyoung M Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Nunzia Caporarello
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Qi Tan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | | | - Yuning Xiong
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Jeong-Heon Lee
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Zhenqing Ye
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Huihuang Yan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Tamas Ordog
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | | | | | | |
Collapse
|
28
|
Islam MS, Molley TG, Ireland J, Kruzic JJ, Kilian KA. Magnetic Nanocomposite Hydrogels for Directing Myofibroblast Activity in Adipose‐Derived Stem Cells. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Md Shariful Islam
- School of Materials Science and Engineering The University of New South Wales Sydney NSW 2052 Australia
| | - Thomas G. Molley
- School of Materials Science and Engineering The University of New South Wales Sydney NSW 2052 Australia
| | - Jake Ireland
- School of Chemistry Australian Centre for Nanomedicine The University of New South Wales Sydney NSW 2052 Australia
| | - Jamie J. Kruzic
- School of Mechanical and Manufacturing Engineering The University of New South Wales Sydney NSW 2052 Australia
| | - Kristopher A. Kilian
- School of Materials Science and Engineering The University of New South Wales Sydney NSW 2052 Australia
- School of Chemistry Australian Centre for Nanomedicine The University of New South Wales Sydney NSW 2052 Australia
| |
Collapse
|
29
|
Liu J, Liu Z, Chen K, Chen W, Fang X, Li M, Zhou X, Ding N, Lei H, Guo C, Qian T, Wang Y, Liu L, Chen Y, Zhao H, Sun Y, Deng Y, Wu C. Kindlin-2 promotes rear focal adhesion disassembly and directional persistence during cell migration. J Cell Sci 2021; 134:jcs244616. [PMID: 33277381 DOI: 10.1242/jcs.244616] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 11/22/2020] [Indexed: 01/13/2023] Open
Abstract
Cell migration involves front-to-rear asymmetric focal adhesion (FA) dynamics, which facilitates trailing edge detachment and directional persistence. Here, we show that kindlin-2 is crucial for FA sliding and disassembly in migrating cells. Loss of kindlin-2 markedly reduced FA number and selectively impaired rear FA sliding and disassembly, resulting in defective rear retraction and reduced directional persistence during cell migration. Kindlin-2-deficient cells failed to develop serum-induced actomyosin-dependent tension at FAs. At the molecular level, kindlin-2 directly interacted with myosin light chain kinase (MYLK, hereafter referred to as MLCK), which was enhanced in response to serum stimulation. Serum deprivation inhibited rear FA disassembly, which was released in response to serum stimulation. Overexpression of the MLCK-binding kindlin-2 F0F1 fragment (amino acid residues 1-167), which inhibits the interaction of endogenous kindlin-2 with MLCK, phenocopied kindlin-2 deficiency-induced migration defects. Inhibition of MLCK, like loss of kindlin-2, also impaired trailing-edge detachment, rear FA disassembly and directional persistence. These results suggest a role of kindlin-2 in promoting actomyosin contractility at FAs, leading to increased rear FA sliding and disassembly, and directional persistence during cell migration.
Collapse
Affiliation(s)
- Jie Liu
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhongzhen Liu
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Keng Chen
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wei Chen
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiyuan Fang
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Meng Li
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xuening Zhou
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ning Ding
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Huan Lei
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chen Guo
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tao Qian
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yilin Wang
- Core Research Facilities, Southern University of Science and Technology, Shenzhen 518055, China
| | - Lin Liu
- Department of Cell Biology and Genetics, College of Life Sciences, Nan Kai University, Tianjin, 300071, China
| | - Yonglong Chen
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hui Zhao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Ying Sun
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yi Deng
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chuanyue Wu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
30
|
dos Santos Á, Cook AW, Gough RE, Schilling M, Olszok N, Brown I, Wang L, Aaron J, Martin-Fernandez ML, Rehfeldt F, Toseland CP. DNA damage alters nuclear mechanics through chromatin reorganization. Nucleic Acids Res 2020; 49:340-353. [PMID: 33330932 PMCID: PMC7797048 DOI: 10.1093/nar/gkaa1202] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/13/2020] [Accepted: 11/25/2020] [Indexed: 12/30/2022] Open
Abstract
DNA double-strand breaks drive genomic instability. However, it remains unknown how these processes may affect the biomechanical properties of the nucleus and what role nuclear mechanics play in DNA damage and repair efficiency. Here, we have used Atomic Force Microscopy to investigate nuclear mechanical changes, arising from externally induced DNA damage. We found that nuclear stiffness is significantly reduced after cisplatin treatment, as a consequence of DNA damage signalling. This softening was linked to global chromatin decondensation, which improves molecular diffusion within the organelle. We propose that this can increase recruitment for repair factors. Interestingly, we also found that reduction of nuclear tension, through cytoskeletal relaxation, has a protective role to the cell and reduces accumulation of DNA damage. Overall, these changes protect against further genomic instability and promote DNA repair. We propose that these processes may underpin the development of drug resistance.
Collapse
Affiliation(s)
- Ália dos Santos
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Alexander W Cook
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Rosemarie E Gough
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Martin Schilling
- University of Göttingen, 3rd Institute of Physics—Biophysics, Göttingen 37077, Germany
| | - Nora A Olszok
- University of Göttingen, 3rd Institute of Physics—Biophysics, Göttingen 37077, Germany
| | - Ian Brown
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Lin Wang
- Central Laser Facility, Research Complex at Harwell, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Harwell, Didcot, Oxford OX11 0QX, UK
| | - Jesse Aaron
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA 20147, USA
| | - Marisa L Martin-Fernandez
- Central Laser Facility, Research Complex at Harwell, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Harwell, Didcot, Oxford OX11 0QX, UK
| | - Florian Rehfeldt
- Correspondence may also be addressed to Florian Rehfeldt. Tel: +49 921 55 2504;
| | | |
Collapse
|
31
|
Missirlis D, Haraszti T, Heckmann L, Spatz JP. Substrate Resistance to Traction Forces Controls Fibroblast Polarization. Biophys J 2020; 119:2558-2572. [PMID: 33217384 DOI: 10.1016/j.bpj.2020.10.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/25/2022] Open
Abstract
The mechanics of fibronectin-rich extracellular matrix regulate cell physiology in a number of diseases, prompting efforts to elucidate cell mechanosensing mechanisms at the molecular and cellular scale. Here, the use of fibronectin-functionalized silicone elastomers that exhibit considerable frequency dependence in viscoelastic properties unveiled the presence of two cellular processes that respond discreetly to substrate mechanical properties. Weakly cross-linked elastomers supported efficient focal adhesion maturation and fibroblast spreading because of an apparent stiff surface layer. However, they did not enable cytoskeletal and fibroblast polarization; elastomers with high cross-linking and low deformability were required for polarization. Our results suggest as an underlying reason for this behavior the inability of soft elastomer substrates to resist traction forces rather than a lack of sufficient traction force generation. Accordingly, mild inhibition of actomyosin contractility rescued fibroblast polarization even on the softer elastomers. Our findings demonstrate differential dependence of substrate physical properties on distinct mechanosensitive processes and provide a premise to reconcile previously proposed local and global models of cell mechanosensing.
Collapse
Affiliation(s)
- Dimitris Missirlis
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany.
| | - Tamás Haraszti
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany; RWTH Aachen University, Institute for Technical and Macromolecular Chemistry, Aachen, Germany
| | - Lara Heckmann
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany
| | - Joachim P Spatz
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany; Heidelberg University, Department of Biophysical Chemistry, Physical Chemistry Institute, Heidelberg, Germany
| |
Collapse
|
32
|
Jo J, Abdi Nansa S, Kim DH. Molecular Regulators of Cellular Mechanoadaptation at Cell-Material Interfaces. Front Bioeng Biotechnol 2020; 8:608569. [PMID: 33364232 PMCID: PMC7753015 DOI: 10.3389/fbioe.2020.608569] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022] Open
Abstract
Diverse essential cellular behaviors are determined by extracellular physical cues that are detected by highly orchestrated subcellular interactions with the extracellular microenvironment. To maintain the reciprocity of cellular responses and mechanical properties of the extracellular matrix, cells utilize a variety of signaling pathways that transduce biophysical stimuli to biochemical reactions. Recent advances in the micromanipulation of individual cells have shown that cellular responses to distinct physical and chemical features of the material are fundamental determinants of cellular mechanosensation and mechanotransduction. In the process of outside-in signal transduction, transmembrane protein integrins facilitate the formation of focal adhesion protein clusters that are connected to the cytoskeletal architecture and anchor the cell to the substrate. The linkers of nucleoskeleton and cytoskeleton molecular complexes, collectively termed LINC, are critical signal transducers that relay biophysical signals between the extranuclear cytoplasmic region and intranuclear nucleoplasmic region. Mechanical signals that involve cytoskeletal remodeling ultimately propagate into the nuclear envelope comprising the nuclear lamina in assistance with various nuclear membrane proteins, where nuclear mechanics play a key role in the subsequent alteration of gene expression and epigenetic modification. These intracellular mechanical signaling cues adjust cellular behaviors directly associated with mechanohomeostasis. Diverse strategies to modulate cell-material interfaces, including alteration of surface rigidity, confinement of cell adhesive region, and changes in surface topology, have been proposed to identify cellular signal transduction at the cellular and subcellular levels. In this review, we will discuss how a diversity of alterations in the physical properties of materials induce distinct cellular responses such as adhesion, migration, proliferation, differentiation, and chromosomal organization. Furthermore, the pathological relevance of misregulated cellular mechanosensation and mechanotransduction in the progression of devastating human diseases, including cardiovascular diseases, cancer, and aging, will be extensively reviewed. Understanding cellular responses to various extracellular forces is expected to provide new insights into how cellular mechanoadaptation is modulated by manipulating the mechanics of extracellular matrix and the application of these materials in clinical aspects.
Collapse
Affiliation(s)
| | | | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| |
Collapse
|
33
|
Lee K, Chen Y, Yoshitomi T, Kawazoe N, Yang Y, Chen G. Osteogenic and Adipogenic Differentiation of Mesenchymal Stem Cells in Gelatin Solutions of Different Viscosities. Adv Healthc Mater 2020; 9:e2000617. [PMID: 32755043 DOI: 10.1002/adhm.202000617] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/03/2020] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates that stem cell fate can be regulated by mechanical properties of the extracellular matrix. Most studies have focused onthe influence of matrix elasticity and viscoelasticity on stem cell differentiation. However, how matrix viscosity affects stem cell differentiation has been overlooked. In this study, a biphasic gelatin solution/hydrogel system is used for 3D culture of human bone marrow-derived mesenchymal stem cells (MSCs) to investigate the influence of gelatin solution viscosity on simultaneous osteogenic and adipogenic differentiation at the same culture condition. Gelatin solution promotes cell proliferation, while its promotive effect decreases with the increase of viscosity. The influence of viscosity on osteogenic and adipogenic differentiation of MSCs shows opposite trends. A high-viscosity gelatin solution results in an increase of alkaline phosphatase (ALP) activity, calcium deposition, and expression of osteogenesis-related genes. On the other hand, in a low-viscosity gelatin solution, a lot of lipid vacuoles are formed and adipogenesis-related genes are highly expressed. The results indicate high viscosity is beneficial for osteogenic differentiation, while low viscosity is beneficial for adipogenic differentiation. These findings suggest the importance of matrix viscosity on stem cell differentiation in 3D microenvironments.
Collapse
Affiliation(s)
- Kyubae Lee
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Yazhou Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Toru Yoshitomi
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Naoki Kawazoe
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8571, Japan
| | - Guoping Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| |
Collapse
|
34
|
Brooks J, Minnick G, Mukherjee P, Jaberi A, Chang L, Espinosa HD, Yang R. High Throughput and Highly Controllable Methods for In Vitro Intracellular Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004917. [PMID: 33241661 PMCID: PMC8729875 DOI: 10.1002/smll.202004917] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/06/2020] [Indexed: 05/03/2023]
Abstract
In vitro and ex vivo intracellular delivery methods hold the key for releasing the full potential of tissue engineering, drug development, and many other applications. In recent years, there has been significant progress in the design and implementation of intracellular delivery systems capable of delivery at the same scale as viral transfection and bulk electroporation but offering fewer adverse outcomes. This review strives to examine a variety of methods for in vitro and ex vivo intracellular delivery such as flow-through microfluidics, engineered substrates, and automated probe-based systems from the perspective of throughput and control. Special attention is paid to a particularly promising method of electroporation using micro/nanochannel based porous substrates, which expose small patches of cell membrane to permeabilizing electric field. Porous substrate electroporation parameters discussed include system design, cells and cargos used, transfection efficiency and cell viability, and the electric field and its effects on molecular transport. The review concludes with discussion of potential new innovations which can arise from specific aspects of porous substrate-based electroporation platforms and high throughput, high control methods in general.
Collapse
Affiliation(s)
- Justin Brooks
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Grayson Minnick
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Arian Jaberi
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Horacio D. Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
35
|
You E, Ko P, Jeong J, Keum S, Kim JW, Seo YJ, Song WK, Rhee S. Dynein-mediated nuclear translocation of yes-associated protein through microtubule acetylation controls fibroblast activation. Cell Mol Life Sci 2020; 77:4143-4161. [PMID: 31912196 PMCID: PMC11105004 DOI: 10.1007/s00018-019-03412-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/27/2019] [Accepted: 12/04/2019] [Indexed: 12/17/2022]
Abstract
Myofibroblasts are the major cell type that is responsible for increase in the mechanical stiffness in fibrotic tissues. It has well documented that the TGF-β/Smad axis is required for myofibroblast differentiation under the rigid substrate condition. However, the mechanism driving myofibroblast differentiation in soft substrates remains unknown. In this research, we demonstrated that interaction of yes-associated protein (YAP) and acetylated microtubule via dynein, a microtubule motor protein drives nuclear localization of YAP in the soft matrix, which in turn increased TGF-β1-induced transcriptional activity of Smad for myofibroblast differentiation. Pharmacological and genetical disruption of dynein impaired the nuclear translocation of YAP and decreased the TGF-β1-induced Smad activity even though phosphorylation and nuclear localization of Smad occurred normally in α-tubulin acetyltransferase 1 (α-TAT1) knockout cell. Moreover, microtubule acetylation prominently appeared in the fibroblast-like cells nearby the blood vessel in the fibrotic liver induced by CCl4 administration, which was conversely decreased by TGF-β receptor inhibitor. As a result, quantitative inhibition of microtubule acetylation may be suggested as a new target for overcoming fibrotic diseases.
Collapse
Affiliation(s)
- Eunae You
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Panseon Ko
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jangho Jeong
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seula Keum
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Young-Jin Seo
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Woo Keun Song
- Bio Imaging and Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju, 61005, Republic of Korea.
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
36
|
Charrier EE, Pogoda K, Li R, Park CY, Fredberg JJ, Janmey PA. A novel method to make viscoelastic polyacrylamide gels for cell culture and traction force microscopy. APL Bioeng 2020; 4:036104. [PMID: 32666015 PMCID: PMC7334032 DOI: 10.1063/5.0002750] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/26/2020] [Indexed: 12/23/2022] Open
Abstract
Polyacrylamide hydrogels are commonly used in cell biology, notably to cultivate cells on soft surfaces. Polyacrylamide gels are purely elastic and well adapted to cell culture as they are inert and can be conjugated with adhesion proteins. Here, we report a method to make viscoelastic polyacrylamide gels with mechanical properties more closely resembling biological tissues and suitable for cell culture in vitro. We demonstrate that these gels can be used for traction force microscopy experiments. We also show that multiple cell types respond to the viscoelasticity of their substrate and that viscous dissipation has an influence on cell spreading, contractility, and motility. This new material provides new opportunities for investigating how normal or malignant cells sense and respond to viscous dissipation within the extra-cellular matrix.
Collapse
Affiliation(s)
| | | | - Robin Li
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Chan Young Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Jeffrey J. Fredberg
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
37
|
Uto K, Arakawa CK, DeForest CA. Next-Generation Biomaterials for Culture and Manipulation of Stem Cells. Cold Spring Harb Perspect Biol 2020; 12:a035691. [PMID: 31843993 PMCID: PMC7461762 DOI: 10.1101/cshperspect.a035691] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Stem cell fate decisions are informed by physical and chemical cues presented within and by the extracellular matrix. Despite the generally attributed importance of extracellular cues in governing self-renewal, differentiation, and collective behavior, knowledge gaps persist with regard to the individual, synergistic, and competing effects that specific physiochemical signals have on cell function. To better understand basic stem cell biology, as well as to expand opportunities in regenerative medicine and tissue engineering, a growing suite of customizable biomaterials has been developed. These next-generation cell culture materials offer user-defined biochemical and biomechanical properties, increasingly in a manner that can be controlled in time and 3D space. This review highlights recent innovations in this regard, focusing on advances to culture and maintain stemness, direct fate, and to detect stem cell function using biomaterial-based strategies.
Collapse
Affiliation(s)
- Koichiro Uto
- International Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0044, Japan
- PRIME, Japan Agency for Medical Research and Development, Tokyo 100-0044, Japan
| | - Christopher K Arakawa
- International Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0044, Japan
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, USA
| | - Cole A DeForest
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, USA
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, USA
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
38
|
Elasticity-dependent response of malignant cells to viscous dissipation. Biomech Model Mechanobiol 2020; 20:145-154. [PMID: 32785801 PMCID: PMC7892690 DOI: 10.1007/s10237-020-01374-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/31/2020] [Indexed: 12/11/2022]
Abstract
The stiffness of the cellular environment controls malignant cell phenotype and proliferation. However, the effect of viscous dissipation on these parameters has not yet been investigated, in part due to the lack of in vitro cell substrates reproducing the mechanical properties of normal tissues and tumors. In this article, we use a newly reported viscoelastic polyacrylamide gel cell substrate, and we characterize the impact of viscous dissipation on three malignant cell lines: DU145 and PC3 derived from prostate and LN229 from brain. The spreading, motility and proliferation rates of these cells were analyzed on 1 kPa and 5 kPa elastic and viscoelastic gels. Surprisingly, the effect of substrate viscous dissipation on cell behavior depended on substrate stiffness for the three cell types tested. We conclude that viscoelasticity controls the spreading, proliferation and migration of malignant cells in vitro. These results highlight the critical role of viscous dissipation in the phenotype and proliferation of malignant cells, especially in stiff tumor environments.
Collapse
|
39
|
Lacombe J, Harris AF, Zenhausern R, Karsunsky S, Zenhausern F. Plant-Based Scaffolds Modify Cellular Response to Drug and Radiation Exposure Compared to Standard Cell Culture Models. Front Bioeng Biotechnol 2020; 8:932. [PMID: 32850759 PMCID: PMC7426640 DOI: 10.3389/fbioe.2020.00932] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
Plant-based scaffolds present many advantages over a variety of biomaterials. Recent studies explored their potential to be repopulated with human cells and thus highlight a growing interest for their use in tissue engineering or for biomedical applications. However, it is still unclear if these in vitro plant-based scaffolds can modify cell phenotype or affect cellular response to external stimuli. Here, we report the characterization of the mechano-regulation of melanoma SK-MEL-28 and prostate PC3 cells seeded on decellularized spinach leaves scaffolds, compared to cells deposited on standard rigid cell culture substrate, as well as their response to drug and radiation treatment. The results showed that YAP/TAZ signaling was downregulated, cellular morphology altered and proliferation rate decreased when cells were cultured on leaf scaffold. Interestingly, cell culture on vegetal scaffold also affected cellular response to external stress. Thus, SK-MEL-28 cells phenotype is modified leading to a decrease in MITF activity and drug resistance, while PC3 cells showed altered gene expression and radiation response. These findings shed lights on the decellularization of vegetal materials to provide substrates that can be repopulated with human cells to better reproduce a soft tissue microenvironment. However, these complex scaffolds mediate changes in cell behavior and in order to exploit the capability of matching physical properties of the various plant scaffolds to diverse physiological functionalities of cells and human tissue constructs, additional studies are required to better characterize physical and biochemical cell-substrate interactions.
Collapse
Affiliation(s)
- Jerome Lacombe
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, United States
| | - Ashlee F. Harris
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, United States
| | - Ryan Zenhausern
- Department of Biomedical Engineering, College of Engineering, University of Arizona, Tucson, AZ, United States
| | - Sophia Karsunsky
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, United States
- Department of Biomedical Engineering, College of Engineering, University of Arizona, Tucson, AZ, United States
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| |
Collapse
|
40
|
Ruud KF, Hiscox WC, Yu I, Chen RK, Li W. Distinct phenotypes of cancer cells on tissue matrix gel. Breast Cancer Res 2020; 22:82. [PMID: 32736579 PMCID: PMC7395363 DOI: 10.1186/s13058-020-01321-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/23/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Breast cancer cells invading the connective tissues outside the mammary lobule or duct immerse in a reservoir of extracellular matrix (ECM) that is structurally and biochemically distinct from that of their site of origin. The ECM is a spatial network of matrix proteins, which not only provide physical support but also serve as bioactive ligands to the cells. It becomes evident that the dimensional, mechanical, structural, and biochemical properties of ECM are all essential mediators of many cellular functions. To better understand breast cancer development and cancer cell biology in native tissue environment, various tissue-mimicking culture models such as hydrogel have been developed. Collagen I (Col I) and Matrigel are the most common hydrogels used in cancer research and have opened opportunities for addressing biological questions beyond the two-dimensional (2D) cell cultures. Yet, it remains unclear whether these broadly used hydrogels can recapitulate the environmental properties of tissue ECM, and whether breast cancer cells grown on CoI I or Matrigel display similar phenotypes as they would on their native ECM. METHODS We investigated mammary epithelial cell phenotypes and metabolic profiles on animal breast ECM-derived tissue matrix gel (TMG), Col I, and Matrigel. Atomic force microscopy (AFM), fluorescence microscopy, acini formation assay, differentiation experiments, spatial migration/invasion assays, proliferation assay, and nuclear magnetic resonance (NMR) spectroscopy were used to examine biological phenotypes and metabolic changes. Student's t test was applied for statistical analyses. RESULTS Our data showed that under a similar physiological stiffness, the three types of hydrogels exhibited distinct microstructures. Breast cancer cells grown on TMG displayed quite different morphologies, surface receptor expression, differentiation status, migration and invasion, and metabolic profiles compared to those cultured on Col I and Matrigel. Depleting lactate produced by glycolytic metabolism of cancer cells abolished the cell proliferation promoted by the non-tissue-specific hydrogel. CONCLUSION The full ECM protein-based hydrogel system may serve as a biologically relevant model system to study tissue- and disease-specific pathological questions. This work provides insights into tissue matrix regulation of cancer cell biomarker expression and identification of novel therapeutic targets for the treatment of human cancers based on tissue-specific disease modeling.
Collapse
Affiliation(s)
- Kelsey F Ruud
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA
| | - William C Hiscox
- Center for NMR Spectroscopy, Washington State University, Pullman, WA, 99164, USA
| | - Ilhan Yu
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164, USA
| | - Roland K Chen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164, USA
| | - Weimin Li
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|
41
|
Selig M, Lauer JC, Hart ML, Rolauffs B. Mechanotransduction and Stiffness-Sensing: Mechanisms and Opportunities to Control Multiple Molecular Aspects of Cell Phenotype as a Design Cornerstone of Cell-Instructive Biomaterials for Articular Cartilage Repair. Int J Mol Sci 2020; 21:E5399. [PMID: 32751354 PMCID: PMC7432012 DOI: 10.3390/ijms21155399] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/23/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since material stiffness controls many cell functions, we reviewed the currently available knowledge on stiffness sensing and elucidated what is known in the context of clinical and experimental articular cartilage (AC) repair. Remarkably, no stiffness information on the various biomaterials for clinical AC repair was accessible. Using mRNA expression profiles and morphology as surrogate markers of stiffness-related effects, we deduced that the various clinically available biomaterials control chondrocyte (CH) phenotype well, but not to equal extents, and only in non-degenerative settings. Ample evidence demonstrates that multiple molecular aspects of CH and mesenchymal stromal cell (MSC) phenotype are susceptible to material stiffness, because proliferation, migration, lineage determination, shape, cytoskeletal properties, expression profiles, cell surface receptor composition, integrin subunit expression, and nuclear shape and composition of CHs and/or MSCs are stiffness-regulated. Moreover, material stiffness modulates MSC immuno-modulatory and angiogenic properties, transforming growth factor beta 1 (TGF-β1)-induced lineage determination, and CH re-differentiation/de-differentiation, collagen type II fragment production, and TGF-β1- and interleukin 1 beta (IL-1β)-induced changes in cell stiffness and traction force. We then integrated the available molecular signaling data into a stiffness-regulated CH phenotype model. Overall, we recommend using material stiffness for controlling cell phenotype, as this would be a promising design cornerstone for novel future-oriented, cell-instructive biomaterials for clinical high-quality AC repair tissue.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Jasmin C. Lauer
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Melanie L. Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| |
Collapse
|
42
|
Deng Z, Fear MW, Suk Choi Y, Wood FM, Allahham A, Mutsaers SE, Prêle CM. The extracellular matrix and mechanotransduction in pulmonary fibrosis. Int J Biochem Cell Biol 2020; 126:105802. [PMID: 32668329 DOI: 10.1016/j.biocel.2020.105802] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
Pulmonary fibrosis is characterised by excessive scarring in the lung which leads to compromised lung function, serious breathing problems and in some diseases, death. It includes several lung disorders with idiopathic pulmonary fibrosis (IPF) the most common and most severe. Pulmonary fibrosis is considered to be perpetuated by aberrant wound healing which leads to fibroblast accumulation, differentiation and activation, and deposition of excessive amounts of extracellular matrix (ECM) components, in particular, collagen. Recent studies have identified the importance of changes in the composition and structure of lung ECM during the development of pulmonary fibrosis and the interaction between ECM and lung cells. There is strong evidence that increased matrix stiffness induces changes in cell function including proliferation, migration, differentiation and activation. Understanding how changes in the ECM microenvironment influence cell behaviour during fibrogenesis, and the mechanisms regulating these changes, will provide insight for developing new treatments.
Collapse
Affiliation(s)
- Zhenjun Deng
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Institute for Respiratory Health, Nedlands, WA, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Burns Service of Western Australia, Perth Children's Hospital, Nedlands, WA, Australia; Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Amira Allahham
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Steven E Mutsaers
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Cecilia M Prêle
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
43
|
Della Sala F, Biondi M, Guarnieri D, Borzacchiello A, Ambrosio L, Mayol L. Mechanical behavior of bioactive poly(ethylene glycol) diacrylate matrices for biomedical application. J Mech Behav Biomed Mater 2020; 110:103885. [PMID: 32957192 DOI: 10.1016/j.jmbbm.2020.103885] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/12/2020] [Accepted: 05/25/2020] [Indexed: 12/20/2022]
Abstract
The biomedical applications of physically entangled polymeric hydrogels are generally limited due to their weak mechanical properties, rapid swelling and dissolution in physiologically relevant environment. Chemical crosslinking helps stabilizing hydrogel structure and enhancing mechanical properties, thereby allowing a higher stability in phisiological environment. In this context, it is known that the mechanical properties of the hydrogel are affected by both the molecular weight (MW) of the starting polymer and the concentration of the crosslinker. Here, our aim was to assess the influence of polymer MW and concentration in the precursor solution on the mechanical features of the final hydrogel and their influence on cells-material interaction. In detail, 3D synthetic matrices based on poly(ethylene glycol) diacrylate (PEGDA) at two molecular weights (PEG 700 and PEG 3400) and at three different concentrations (10, 20, 40 w/v %), which were photopolymerized using darocour as an initiator, were studied. Then, infrared and swelling analyses, along with a comprehensive mechanical characterization of the obtained hydrogels (i.e. oscillatory shear and confined compression tests), were performed. Finally, to evaluate the influence of the mechanical features on the biological behaviour, the hydrogels were characterized in terms of cell adhesion percentage and cell viability after functionalizing the substrates with RGD peptide at three different concentrations. Results have demonstrated that both the Young's modulus (E) in compression and the elastic modulus (G') in shear of the hydrogels increase with increasing polymer precursor concentration. E decreased as MW increased, and the differences are more relevant for more concentrated hydrogels. On the contrary, G' appears to increase with increasing PEGDA MW and in particular for the lowest polymer precursor concentration. The biological results have demonstrated that cells cultured for longer times seem to prefer PEG 3400 hydrogels with a larger mesh size structure that posses higher viscoelastic properties in shear.
Collapse
Affiliation(s)
- Francesca Della Sala
- Istituto per i Polimeri, Compositi e Biomateriali, Consiglio Nazionale delle Ricerche (IPCB-CNR), Viale J.F. Kennedy 54, Napoli, Italy; University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Marco Biondi
- Dipartimento di Farmacia, Università di Napoli Federico II, Via Domenico Montesano 49, Napoli, Italy; Centro di Ricerca Interdipartimentale sui Biomateriali (CRIB), Università di Napoli Federico II, Piazzale Tecchio 80, Napoli, Italy
| | - Daniela Guarnieri
- Dipartimento di Chimica e Biologia A. Zambelli, Università di Salerno, via Giovanni Paolo II 132, Fisciano, Salerno, I-84084, Italy
| | - Assunta Borzacchiello
- Istituto per i Polimeri, Compositi e Biomateriali, Consiglio Nazionale delle Ricerche (IPCB-CNR), Viale J.F. Kennedy 54, Napoli, Italy.
| | - Luigi Ambrosio
- Istituto per i Polimeri, Compositi e Biomateriali, Consiglio Nazionale delle Ricerche (IPCB-CNR), Viale J.F. Kennedy 54, Napoli, Italy
| | - Laura Mayol
- Dipartimento di Farmacia, Università di Napoli Federico II, Via Domenico Montesano 49, Napoli, Italy; Centro di Ricerca Interdipartimentale sui Biomateriali (CRIB), Università di Napoli Federico II, Piazzale Tecchio 80, Napoli, Italy
| |
Collapse
|
44
|
Cell response to substrate rigidity is regulated by active and passive cytoskeletal stress. Proc Natl Acad Sci U S A 2020; 117:12817-12825. [PMID: 32444491 DOI: 10.1073/pnas.1917555117] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Morphogenesis, tumor formation, and wound healing are regulated by tissue rigidity. Focal adhesion behavior is locally regulated by stiffness; however, how cells globally adapt, detect, and respond to rigidity remains unknown. Here, we studied the interplay between the rheological properties of the cytoskeleton and matrix rigidity. We seeded fibroblasts onto flexible microfabricated pillar arrays with varying stiffness and simultaneously measured the cytoskeleton organization, traction forces, and cell-rigidity responses at both the adhesion and cell scale. Cells adopted a rigidity-dependent phenotype whereby the actin cytoskeleton polarized on stiff substrates but not on soft. We further showed a crucial role of active and passive cross-linkers in rigidity-sensing responses. By reducing myosin II activity or knocking down α-actinin, we found that both promoted cell polarization on soft substrates, whereas α-actinin overexpression prevented polarization on stiff substrates. Atomic force microscopy indentation experiments showed that this polarization response correlated with cell stiffness, whereby cell stiffness decreased when active or passive cross-linking was reduced and softer cells polarized on softer matrices. Theoretical modeling of the actin network as an active gel suggests that adaptation to matrix rigidity is controlled by internal mechanical properties of the cytoskeleton and puts forward a universal scaling between nematic order of the actin cytoskeleton and the substrate-to-cell elastic modulus ratio. Altogether, our study demonstrates the implication of cell-scale mechanosensing through the internal stress within the actomyosin cytoskeleton and its coupling with local rigidity sensing at focal adhesions in the regulation of cell shape changes and polarity.
Collapse
|
45
|
Chen X, Deng Z, He Y, Lu F, Yuan Y. Mechanical Strain Promotes Proliferation of Adipose-Derived Stem Cells Through the Integrin β1-Mediated RhoA/Myosin Light Chain Pathway. Tissue Eng Part A 2020; 26:939-952. [PMID: 32066340 DOI: 10.1089/ten.tea.2019.0266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
External volume expansion (EVE) promotes proliferation of adipose-derived stem cells (ADSCs) during adipose tissue regeneration. However, the mechanism by which EVE is translated into biochemical signals and subsequently induces proliferation of ADSCs is poorly understood. Here, we investigated the strain in adipose tissue and mechanochemical signaling upon EVE in rats. In addition, the effect of mechanical strain on proliferation of ADSCs was assessed using a custom-built Flexcell device. The level of strain in adipose tissue upon EVE peaked at week 1 and then decreased over time, and the cell proliferation rate was similarly affected. Mechanical strain-dependent activation of integrin β1 and the RhoA/myosin light chain (MLC) pathway was involved in cell proliferation. The proliferation rate of ADSCs was higher under 12% mechanical strain than under 6% and 0% mechanical strain in vitro. Mechanical strain-dependent activation of integrin β1 promoted activation of the small GTPase RhoA and phosphorylation of MLC. Furthermore, knockdown of integrin β1 attenuated activation of the RhoA/MLC pathway and proliferation of ADSCs in response to mechanical strain. Taken together, this study provides the first evidence of mechanochemical signaling in response to EVE. These data may help elucidate the effects of different strain levels on adipose tissue regeneration. Impact statement External volume expansion (EVE) induces adipose tissue regeneration and has great therapeutic potential to correct soft tissue defects. This study showed that EVE promotes proliferation of adipose-derived stem cells by activating integrin β1 and its crucial downstream signaling molecules, namely the small GTPase RhoA and p-myosin light chain. The findings of this study may assist clinical tissue engineering applications and provide new insights into the regulation of adipose tissue regeneration in clinical practice.
Collapse
Affiliation(s)
- Xihang Chen
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Zilong Deng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,College of Stomatology, Southern Medical University, Guangzhou, People's Republic of China
| | - Yunfan He
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Feng Lu
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yi Yuan
- Department of Plastic and Reconstructive Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
46
|
Davidson CD, Jayco DKP, Matera DL, DePalma SJ, Hiraki HL, Wang WY, Baker BM. Myofibroblast activation in synthetic fibrous matrices composed of dextran vinyl sulfone. Acta Biomater 2020; 105:78-86. [PMID: 31945504 PMCID: PMC7369643 DOI: 10.1016/j.actbio.2020.01.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/18/2019] [Accepted: 01/08/2020] [Indexed: 02/07/2023]
Abstract
Mechanical interactions between fibroblasts and their surrounding extracellular matrix (ECM) guide fundamental behaviors such as spreading, migration, and proliferation that underlie disease pathogenesis. The challenges of studying ECM mechanics in vivo have motivated the development of in vitro models of the fibrous ECM in which fibroblasts reside. Natural materials such as collagen hydrogels bear structural and biochemical resemblance to stromal ECM, but mechanistic studies in these settings are often confounded by cell-mediated material degradation and the lack of structural and mechanical tunability. Here, we established a new material system composed of electrospun dextran vinyl sulfone (DexVS) polymeric fibers. These fibrous matrices exhibit mechanical tunability at both the single fiber (80-340 MPa) and bulk matrix (0.77-11.03 kPa) level, as well as long-term stability in mechanical properties over a two-week period. Cell adhesion to these matrices can be either user-defined by functionalizing synthetic fibers with thiolated adhesive peptides or methacrylated heparin to sequester cell-derived ECM proteins. We utilized DexVS fibrous matrices to investigate the role of matrix mechanics on the activation of fibroblasts into myofibroblasts, a key step of the fibrotic progression. In contrast to previous findings with non-fibrous hydrogel substrates, we find that fibroblasts in soft and deformable matrices exhibit increased spreading, focal adhesion formation, proliferation, and myofibroblast activation as compared to cells on stiffer matrices with equivalent starting architecture. STATEMENT OF SIGNIFICANCE: Cellular mechanosensing of fibrillar extracellular matrices plays a critical role in homeostasis and disease progression in stromal connective tissue. Here, we established a new material system composed of electrospun dextran vinyl sulfone polymeric fibers. These matrices exhibit architectural, mechanical, and biochemical tunability to accurately model diverse tissue microenvironments found in the body. In contrast to previous observations with non-fibrous hydrogels, we find that fibroblasts in soft and deformable fibrous matrices exhibit increased spreading and focal adhesion formation as compared to those in stiffer matrices with equivalent architecture. We also investigated the role of matrix stiffness on myofibroblast activation, a critical step in the fibrotic cascade, and find that low stiffness matrices promote increased myofibroblast activation.
Collapse
Affiliation(s)
- Christopher D Davidson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Danica Kristen P Jayco
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Daniel L Matera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Samuel J DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Harrison L Hiraki
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - William Y Wang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
47
|
Bagheri-Hosseinabadi Z, Seyedi F, Mollaei HR, Moshrefi M, Seifalian A. Combination of 5-azaytidine and hanging drop culture convert fat cell into cardiac cell. Biotechnol Appl Biochem 2020; 68:92-101. [PMID: 32028539 DOI: 10.1002/bab.1897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
One of the promising approaches for the treatment of cardiac disease is stem cell therapy. In this study, we compared the cardiomyogenic differentiation rate, from human adipose-derived stem cells (hADSCs) in a three-dimensional (3D) hanging drop (HD) spheroid culture system, versus a two-dimensional (2D) culture condition at different concentrations of 5-azacytidine (5-Aza). 5-Azaytidine (5-Aza) is a pyrimidine nucleoside analogue of cytidine that initiates cell differentiation programs through DNA demethylation. The hADSCs were isolated and cultured both in 2D and 3D HD conditions, with either 10 or 50 μM concentrations of 5-Aza. Then DNA content, gene expression, and protein content were analyzed. 3D HD culture resulted in a higher percentage of cells in G0/G1 and S phase in the cell division cycle, whereas 2D culture led to a greater percentage of cells in the G2/M phase. A significantly higher gene expression rate of HAND1, HAND2, cTnI, Cx43, βMHC, GATA4, NKX2.5, and MLC2V was observed in HD treated with 50 μM 5-Aza. This was confirmed by immunocytochemistry. These findings suggest that 50 μM concentration of 5-Aza can induce hADSCs to differentiate into cardiomyocytes. The differentiation rate was significantly higher when accompanied by the 3D HD culture system. This work provides a new culture system for cell differentiation for cardiovascular tissue engineering.
Collapse
Affiliation(s)
- Zahra Bagheri-Hosseinabadi
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Seyedi
- Department of Anatomy, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Hamid Reza Mollaei
- Department of Medical Microbiology, Afzalipour Medical Faculty, Kerman University of Medical Science, Kerman, Iran
| | - Mojgan Moshrefi
- Medical Nanotechnology & Tissue Engineering Research Centre, Yazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alexander Seifalian
- Nanotechnology and Regenerative Medicine Commercialisation Centre (NanoRegMed Ltd.), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
48
|
Nguyen AV, Trompetto B, Tan XHM, Scott MB, Hu KHH, Deeds E, Butte MJ, Chiou PY, Rowat AC. Differential Contributions of Actin and Myosin to the Physical Phenotypes and Invasion of Pancreatic Cancer Cells. Cell Mol Bioeng 2020; 13:27-44. [PMID: 32030106 PMCID: PMC6981337 DOI: 10.1007/s12195-019-00603-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Metastasis is a fundamentally physical process in which cells deform through narrow gaps and generate forces to invade surrounding tissues. While it is commonly thought that increased cell deformability is an advantage for invading cells, we previously found that more invasive pancreatic ductal adenocarcinoma (PDAC) cells are stiffer than less invasive PDAC cells. Here we investigate potential mechanisms of the simultaneous increase in PDAC cell stiffness and invasion, focusing on the contributions of myosin II, Arp2/3, and formins. METHOD We measure cell invasion using a 3D scratch wound invasion assay and cell stiffness using atomic force microscopy (AFM). To determine the effects of actin- and myosin-mediated force generation on cell stiffness and invasion, we treat cells with pharmacologic inhibitors of myosin II (blebbistatin), Arp2/3 (CK-666), and formins (SMIFH2). RESULTS We find that the activity of myosin II, Arp2/3, and formins all contribute to the stiffness of PDAC cells. Interestingly, we find that the invasion of PDAC cell lines is differentially affected when the activity of myosin II, Arp2/3, or formins is inhibited, suggesting that despite having similar tissue origins, different PDAC cell lines may rely on different mechanisms for invasion. CONCLUSIONS These findings deepen our knowledge of the factors that regulate cancer cell mechanotype and invasion, and incite further studies to develop therapeutics that target multiple mechanisms of invasion for improved clinical benefit.
Collapse
Affiliation(s)
- Angelyn V. Nguyen
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
| | - Brittany Trompetto
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
| | | | - Michael B. Scott
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, USA
- Present Address: Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, USA
- Department of Biomedical Engineering, Northwestern McCormick School of Engineering, Evanston, USA
| | | | - Eric Deeds
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, USA
| | - Manish J. Butte
- Department of Pediatrics, University of California, Los Angeles, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, USA
| | - Pei Yu Chiou
- Department of Bioengineering, University of California, Los Angeles, USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, USA
| | - Amy C. Rowat
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, USA
| |
Collapse
|
49
|
Kim MH, Kino-Oka M. Bioengineering Considerations for a Nurturing Way to Enhance Scalable Expansion of Human Pluripotent Stem Cells. Biotechnol J 2020; 15:e1900314. [PMID: 31904180 DOI: 10.1002/biot.201900314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Understanding how defects in mechanotransduction affect cell-to-cell variability will add to the fundamental knowledge of human pluripotent stem cell (hPSC) culture, and may suggest new approaches for achieving a robust, reproducible, and scalable process that result in consistent product quality and yields. Here, the current state of the understanding of the fundamental mechanisms that govern the growth kinetics of hPSCs between static and dynamic cultures is reviewed, the factors causing fluctuations are identified, and culture strategies that might eliminate or minimize the occurrence of cell-to-cell variability arising from these fluctuations are discussed. The existing challenges in the development of hPSC expansion methods for enabling the transition from process development to large-scale production are addressed, a mandatory step for industrial and clinical applications of hPSCs.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
50
|
Zhong J, Yang Y, Liao L, Zhang C. Matrix stiffness-regulated cellular functions under different dimensionalities. Biomater Sci 2020; 8:2734-2755. [DOI: 10.1039/c9bm01809c] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The microenvironments that cells encounter with in vitro.
Collapse
Affiliation(s)
- Jiajun Zhong
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Yuexiong Yang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Liqiong Liao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Biomaterials Research Center
- School of Biomedical Engineering
- Southern Medical University
- Guangzhou
| | - Chao Zhang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| |
Collapse
|