1
|
Lipari S, Sacco P, Cok M, Scognamiglio F, Romano M, Brun F, Giulianini PG, Marsich E, Aachmann FL, Donati I. Hydrogel Elastic Energy: A Stressor Triggering an Adaptive Stress-Mediated Cell Response. Adv Healthc Mater 2025; 14:e2402400. [PMID: 39535422 PMCID: PMC11730662 DOI: 10.1002/adhm.202402400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The crosstalk between the cells and the extracellular matrix (ECM) is bidirectional and consists of a pushing/pulling stretch exerted by the cells and a mechanical resistance counteracted by the surrounding microenvironment. It is widely recognized that the stiffness of the ECM, its viscoelasticity, and its overall deformation are the most important traits influencing the response of the cells. Here these three parameters are combined into a concept of elastic energy, which in biological terms represents the mechanical feedback that cells perceive when the ECM is deformed. It is shown that elastic energy is a stress factor that influences the response of cells in three-dimensional (3D) cultures. Strikingly, the higher the elastic energy of the matrix and thus the mechanical feedback, the higher the stress state of the cells, which correlates with the formation of G3BP-mediated stress granules. This condition is associated with an increase in alkaline phosphatase (ALP) activity but a decrease in gene expression and is mediated by the nuclear translocation of Yes-associated protein (YAP). This work supports the importance of considering the elastic energy as mechano-controller in regulating cellular stress state in 3D cultures.
Collapse
Affiliation(s)
- Sara Lipari
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Pasquale Sacco
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Michela Cok
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | | | - Maurizio Romano
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Francesco Brun
- Department of Engineering and ArchitectureUniversity of TriesteVia A. Valerio 6/1Trieste34127Italy
| | | | - Eleonora Marsich
- Department of MedicineSurgery and Health SciencesUniversity of TriestePiazza dell'Ospitale 1Trieste34129Italy
| | - Finn L. Aachmann
- Department of Biotechnology and Food ScienceNorwegian Biopolymer Laboratory (NOBIPOL)NTNU Norwegian University of Science and TechnologySem Sælands vei 6/8Trondheim7491Norway
| | - Ivan Donati
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| |
Collapse
|
2
|
Wang D, Feng S, Yang M. Multi-Gradient Bone-Like Nanocomposites Induced by Strain Distribution. ACS NANO 2024; 18:29636-29647. [PMID: 39425938 DOI: 10.1021/acsnano.4c08442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
The heterogeneity of bones is elegantly adapted to the local strain environment, which is critical for maintaining mechanical functions. Such an adaptation enables the strong correlation between strain distributions and multiple gradients, underlying a promising pathway for creating complex gradient structures. However, this potential remains largely unexplored for the synthesis of functional gradient materials. In this work, heterogeneous bone-like nanocomposites with complex structural and compositional gradients comparable to bones are synthesized by inducing strain distributions within the polymer matrix containing amorphous calcium phosphate (ACP). Uniaxial stretching of composite films exerts the highest strain in the center, which ceases gradually toward the sides, resulting in the gradual decrease of polymer alignment and crystallinity. Simultaneously, the center with high orientation traps most ACP during stretching due to the nanoconfinement effect, which in turn promotes the formation of aligned nanofibrous structures. The sides experiencing the least strain have the smallest amounts of ACP, characteristic of porous architectures. Further crystallization of ACP produces oriented apatite nanorods in the center with a larger crystalline/amorphous ratio than the sides because of template-induced crystallization. The combination of structural and compositional gradients leads to gradient mechanical properties, and the gradient span and magnitude correlate nicely with strain distributions. Accompanying bone-like mechanical gradients, the center is less adhesive and self-healable than the sides, which allows a better recovery after a complete cutting. Our work may represent a general strategy for the synthesis of biomimetic materials with complex gradients thanks to the ubiquitous presence of strain distributions in load-bearing structures.
Collapse
Affiliation(s)
- Di Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, China
| | - Shouhua Feng
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, China
| | - Ming Yang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
3
|
Stephens CJ, Kobayashi R, Berry DC, Butcher JT. The Role of Matrix Stiffness And Viscosity on Lipid Phenotype And Fat Lineage Potential. Tissue Eng Part A 2024. [PMID: 39165245 DOI: 10.1089/ten.tea.2024.0149] [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] [Indexed: 08/22/2024] Open
Abstract
Autologous fat transfer is a common procedure that patients undergo to rejuvenate large soft tissue defects. However, these surgeries are complicated by limited tissue sources, donor-site morbidity, and necrosis. While the biofabrication of fat tissue can serve as a clinical option for reconstructive surgery, the influence of matrix mechanics, specifically stiffness and viscosity, on adipogenesis requires further elucidation. Additionally, the effects of these mechanical parameters on metabolic and thermogenic fat potential have yet to be investigated. In this study, gelatin methacryloyl (GelMA) polymers with varying degrees of methacrylation (DoM) were fabricated to create matrices with different stiffnesses and viscosities. Human adipose-derived mesenchymal stem cells were then encapsulated in mechanically tunable GelMA and underwent adipogenesis to investigate the effects of matrix mechanics on lipid phenotype and fat potential. Mechanical testing confirmed that GelMA stiffness was regulated by DoM and weight composition, whereas viscosity was determined by the latter. Further work revealed that while lipid phenotype became more enriched as matrix stiffness and viscosity declined, the potential toward metabolic and thermogenic fat appeared to be more viscous dependent rather than stiffness dependent. In addition, fatty acid binding protein 4 and uncoupling protein 1 gene expression exhibited viscous-dependent behavior despite comparable levels of peroxisome proliferator-activated receptor gamma. However, despite the superior role of viscosity, lipid quantity and mitochondrial abundance demonstrated stiffness-dependent behavior. Overall, this work revealed that matrix viscosity played a more superior role than stiffness in driving adipogenesis and distinguishing between metabolic and thermogenic fat potential. Ultimately, this differentiation in fat production is important for engineering ideal adipose tissue for large soft tissue defects.
Collapse
Affiliation(s)
- Chelsea J Stephens
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| | - Reina Kobayashi
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| | - Daniel C Berry
- College of Human Ecology, Division of Nutrition Science, Cornell University, Ithaca, New York, USA
| | - Jonathan T Butcher
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| |
Collapse
|
4
|
Mowla A, Hepburn MS, Li J, Vahala D, Amos SE, Hirvonen LM, Sanderson RW, Wijesinghe P, Maher S, Choi YS, Kennedy BF. Multimodal mechano-microscopy reveals mechanical phenotypes of breast cancer spheroids in three dimensions. APL Bioeng 2024; 8:036113. [PMID: 39257700 PMCID: PMC11387014 DOI: 10.1063/5.0213077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/01/2024] [Indexed: 09/12/2024] Open
Abstract
Cancer cell invasion relies on an equilibrium between cell deformability and the biophysical constraints imposed by the extracellular matrix (ECM). However, there is little consensus on the nature of the local biomechanical alterations in cancer cell dissemination in the context of three-dimensional (3D) tumor microenvironments (TMEs). While the shortcomings of two-dimensional (2D) models in replicating in situ cell behavior are well known, 3D TME models remain underutilized because contemporary mechanical quantification tools are limited to surface measurements. Here, we overcome this major challenge by quantifying local mechanics of cancer cell spheroids in 3D TMEs. We achieve this using multimodal mechano-microscopy, integrating optical coherence microscopy-based elasticity imaging with confocal fluorescence microscopy. We observe that non-metastatic cancer spheroids show no invasion while showing increased peripheral cell elasticity in both stiff and soft environments. Metastatic cancer spheroids, however, show ECM-mediated softening in a stiff microenvironment and, in a soft environment, initiate cell invasion with peripheral softening associated with early metastatic dissemination. This exemplar of live-cell 3D mechanotyping supports that invasion increases cell deformability in a 3D context, illustrating the power of multimodal mechano-microscopy for quantitative mechanobiology in situ.
Collapse
Affiliation(s)
| | | | | | - Danielle Vahala
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Sebastian E Amos
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Liisa M Hirvonen
- Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, Perth, WA 6009, Australia
| | | | - Philip Wijesinghe
- Centre of Biophotonics, SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Samuel Maher
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | | |
Collapse
|
5
|
Hu X, Bao M. Advances in micropatterning technology for mechanotransduction research. MECHANOBIOLOGY IN MEDICINE 2024; 2:100066. [PMID: 40395493 PMCID: PMC12082312 DOI: 10.1016/j.mbm.2024.100066] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/19/2024] [Accepted: 03/24/2024] [Indexed: 05/22/2025]
Abstract
Micropatterning is a sophisticated technique that precisely manipulates the spatial distribution of cell adhesion proteins on various substrates across multiple scales. This precise control over adhesive regions facilitates the manipulation of architectures and physical constraints for single or multiple cells. Furthermore, it allows for an in-depth analysis of how chemical and physical properties influence cellular functionality. In this comprehensive review, we explore the current understanding of the impact of geometrical confinement on cellular functions across various dimensions, emphasizing the benefits of micropatterning in addressing fundamental biological queries. We advocate that utilizing directed self-organization via physical confinement and morphogen gradients on micropatterned surfaces represents an innovative approach to generating functional tissue and controlling morphogenesis in vitro. Integrating this technique with cutting-edge technologies, micropatterning presents a significant potential to bridge a crucial knowledge gap in understanding core biological processes.
Collapse
Affiliation(s)
- Xinyu Hu
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, the Netherlands
| | - Min Bao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325001, Zhejiang, China
| |
Collapse
|
6
|
Soliman BG, Chin IL, Li Y, Ishii M, Ho MH, Doan VK, Cox TR, Wang PY, Lindberg GCJ, Zhang YS, Woodfield TBF, Choi YS, Lim KS. Droplet-based microfluidics for engineering shape-controlled hydrogels with stiffness gradient. Biofabrication 2024; 16:045026. [PMID: 39121873 DOI: 10.1088/1758-5090/ad6d8e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Current biofabrication strategies are limited in their ability to replicate native shape-to-function relationships, that are dependent on adequate biomimicry of macroscale shape as well as size and microscale spatial heterogeneity, within cell-laden hydrogels. In this study, a novel diffusion-based microfluidics platform is presented that meets these needs in a two-step process. In the first step, a hydrogel-precursor solution is dispersed into a continuous oil phase within the microfluidics tubing. By adjusting the dispersed and oil phase flow rates, the physical architecture of hydrogel-precursor phases can be adjusted to generate spherical and plug-like structures, as well as continuous meter-long hydrogel-precursor phases (up to 1.75 m). The second step involves the controlled introduction a small molecule-containing aqueous phase through a T-shaped tube connector to enable controlled small molecule diffusion across the interface of the aqueous phase and hydrogel-precursor. Application of this system is demonstrated by diffusing co-initiator sodium persulfate (SPS) into hydrogel-precursor solutions, where the controlled SPS diffusion into the hydrogel-precursor and subsequent photo-polymerization allows for the formation of unique radial stiffness patterns across the shape- and size-controlled hydrogels, as well as allowing the formation of hollow hydrogels with controllable internal architectures. Mesenchymal stromal cells are successfully encapsulated within hollow hydrogels and hydrogels containing radial stiffness gradient and found to respond to the heterogeneity in stiffness through the yes-associated protein mechano-regulator. Finally, breast cancer cells are found to phenotypically switch in response to stiffness gradients, causing a shift in their ability to aggregate, which may have implications for metastasis. The diffusion-based microfluidics thus finds application mimicking native shape-to-function relationship in the context of tissue engineering and provides a platform to further study the roles of micro- and macroscale architectural features that exist within native tissues.
Collapse
Affiliation(s)
- Bram G Soliman
- Light Activated Biomaterials (LAB) Group, University of Otago, Christchurch 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
- School of Material Science and Engineering, University of New South Wales, Sydney 2052, Australia
| | - Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth 6009, Australia
| | - Yiwei Li
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| | - Melissa Ishii
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
| | - Minh Hieu Ho
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| | - Vinh Khanh Doan
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| | - Thomas R Cox
- The Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Peng Yuan Wang
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 32500, People's Republic of China
| | - Gabriella C J Lindberg
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, United States of America
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth 6009, Australia
| | - Khoon S Lim
- Light Activated Biomaterials (LAB) Group, University of Otago, Christchurch 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
7
|
Kan T, Li H, Hou L, Cui J, Wang Y, Sun L, Wang L, Yan M, Yu Z. Matrix stiffness aggravates osteoarthritis progression through H3K27me3 demethylation induced by mitochondrial damage. iScience 2024; 27:110507. [PMID: 39156637 PMCID: PMC11328034 DOI: 10.1016/j.isci.2024.110507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/20/2024] [Accepted: 07/11/2024] [Indexed: 08/20/2024] Open
Abstract
Abnormal epigenetics is the initial factor of the occurrence and development of osteoarthritis (OA), and abnormal mechanical load is a key pathogenic factor of OA. However, how abnormal mechanical load affects chondrocyte epigenetics is unclear. Chondrocytes reportedly respond to mechanics through the extracellular matrix (ECM), which has a role in regulating epigenetics in various diseases, and mitochondria are potential mediators of communication between mechanics and epigenetics. Therefore, it is hypothesized that the matrix mechanics of cartilage regulates their epigenetics through mitochondria and leads to OA. The matrix stiffness of OA cartilage on the stress-concentrated side increases, mitochondrial damage of chondrocyte is severe, and the chondrocyte H3K27me3 is demethylated. Moreover, mitochondrial permeability transition pore (mPTP) opens to increase the translocation of plant homeodomain finger protein 8 (Phf8) into the nucleus to catalyze H3K27me3 demethylation. This provides a new perspective for us to understand the mechanism of OA based on mechanobiology.
Collapse
Affiliation(s)
- Tianyou Kan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Hanjun Li
- Renji-Med X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lingli Hou
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Junqi Cui
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yao Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Lin Sun
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Liao Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mengning Yan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
8
|
Zhao Y, Dong X, Li Y, Cui J, Shi Q, Huang HW, Huang Q, Wang H. Integrated Cross-Scale Manipulation and Modulable Encapsulation of Cell-Laden Hydrogel for Constructing Tissue-Mimicking Microstructures. RESEARCH (WASHINGTON, D.C.) 2024; 7:0414. [PMID: 39050820 PMCID: PMC11266663 DOI: 10.34133/research.0414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/27/2024] [Indexed: 07/27/2024]
Abstract
Engineered microstructures that mimic in vivo tissues have demonstrated great potential for applications in regenerative medicine, drug screening, and cell behavior exploration. However, current methods for engineering microstructures that mimic the multi-extracellular matrix and multicellular features of natural tissues to realize tissue-mimicking microstructures in vitro remain insufficient. Here, we propose a versatile method for constructing tissue-mimicking heterogeneous microstructures by orderly integration of macroscopic hydrogel exchange, microscopic cell manipulation, and encapsulation modulation. First, various cell-laden hydrogel droplets are manipulated at the millimeter scale using electrowetting on dielectric to achieve efficient hydrogel exchange. Second, the cells are manipulated at the micrometer scale using dielectrophoresis to adjust their density and arrangement within the hydrogel droplets. Third, the photopolymerization of these hydrogel droplets is triggered in designated regions by dynamically modulating the shape and position of the excitation ultraviolet beam. Thus, heterogeneous microstructures with different extracellular matrix geometries and components were constructed, including specific cell densities and patterns. The resulting heterogeneous microstructure supported long-term culture of hepatocytes and fibroblasts with high cell viability (over 90%). Moreover, the density and distribution of the 2 cell types had significant effects on the cell proliferation and urea secretion. We propose that our method can lead to the construction of additional biomimetic heterogeneous microstructures with unprecedented potential for use in future tissue engineering applications.
Collapse
Affiliation(s)
- Yanfeng Zhao
- Intelligent Robotics Institute, School of Mechatronical Engineering,
Beijing Institute of Technology, Beijing 100081, China
| | - Xinyi Dong
- Intelligent Robotics Institute, School of Mechatronical Engineering,
Beijing Institute of Technology, Beijing 100081, China
| | - Yang Li
- Peking University First Hospital, Xicheng District, Beijing 100034, China
| | - Juan Cui
- Key Laboratory of Instrumentation Science and Dynamic Measurement, Ministry of Education,
North University of China, Taiyuan 030051, China
| | - Qing Shi
- Beijing Advanced Innovation Center for Intelligent Robots and Systems,
Beijing Institute of Technology, Beijing 100081, China
| | - Hen-Wei Huang
- Laboratory for Translational Engineering,
Harvard Medical School, Cambridge, MA 02139, USA
| | - Qiang Huang
- Beijing Advanced Innovation Center for Intelligent Robots and Systems,
Beijing Institute of Technology, Beijing 100081, China
| | - Huaping Wang
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| |
Collapse
|
9
|
Xie J, Huck WTS, Bao M. Unveiling the Intricate Connection: Cell Volume as a Key Regulator of Mechanotransduction. Annu Rev Biophys 2024; 53:299-317. [PMID: 38424091 DOI: 10.1146/annurev-biophys-030822-035656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The volumes of living cells undergo dynamic changes to maintain the cells' structural and functional integrity in many physiological processes. Minor fluctuations in cell volume can serve as intrinsic signals that play a crucial role in cell fate determination during mechanotransduction. In this review, we discuss the variability of cell volume and its role in vivo, along with an overview of the mechanisms governing cell volume regulation. Additionally, we provide insights into the current approaches used to control cell volume in vitro. Furthermore, we summarize the biological implications of cell volume regulation and discuss recent advances in understanding the fundamental relationship between cell volume and mechanotransduction. Finally, we delve into the potential underlying mechanisms, including intracellular macromolecular crowding and cellular mechanics, that govern the global regulation of cell fate in response to changes in cell volume. By exploring the intricate interplay between cell volume and mechanotransduction, we underscore the importance of considering cell volume as a fundamental signaling cue to unravel the basic principles of mechanotransduction. Additionally, we propose future research directions that can extend our current understanding of cell volume in mechanotransduction. Overall, this review highlights the significance of considering cell volume as a fundamental signal in understanding the basic principles in mechanotransduction and points out the possibility of controlling cell volume to control cell fate, mitigate disease-related damage, and facilitate the healing of damaged tissues.
Collapse
Affiliation(s)
- Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands;
| | - Min Bao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China;
| |
Collapse
|
10
|
Tan YH, Wang KCW, Chin IL, Sanderson RW, Li J, Kennedy BF, Noble PB, Choi YS. Stiffness Mediated-Mechanosensation of Airway Smooth Muscle Cells on Linear Stiffness Gradient Hydrogels. Adv Healthc Mater 2024; 13:e2304254. [PMID: 38593989 DOI: 10.1002/adhm.202304254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/28/2024] [Indexed: 04/11/2024]
Abstract
In obstructive airway diseases such as asthma and chronic obstructive pulmonary disease (COPD), the extracellular matrix (ECM) protein amount and composition of the airway smooth muscle (ASM) is often remodelled, likely altering tissue stiffness. The underlying mechanism of how human ASM cell (hASMC) mechanosenses the aberrant microenvironment is not well understood. Physiological stiffnesses of the ASM were measured by uniaxial compression tester using porcine ASM layers under 0, 5 and 10% longitudinal stretch above in situ length. Linear stiffness gradient hydrogels (230 kPa range) were fabricated and functionalized with ECM proteins, collagen I (ColI), fibronectin (Fn) and laminin (Ln), to recapitulate the above-measured range of stiffnesses. Overall, hASMC mechanosensation exhibited a clear correlation with the underlying hydrogel stiffness. Cell size, nuclear size and contractile marker alpha-smooth muscle actin (αSMA) expression showed a strong correlation to substrate stiffness. Mechanosensation, assessed by Lamin-A intensity and nuc/cyto YAP, exhibited stiffness-mediated behaviour only on ColI and Fn-coated hydrogels. Inhibition studies using blebbistatin or Y27632 attenuated most mechanotransduction-derived cell morphological responses, αSMA and Lamin-A expression and nuc/cyto YAP (blebbistatin only). This study highlights the interplay and complexities between stiffness and ECM protein type on hASMC mechanosensation, relevant to airway remodelling in obstructive airway diseases.
Collapse
Affiliation(s)
- Yong Hwee Tan
- School of Human Sciences, The University of Western Australia, Perth, WA, 6009, Australia
| | - Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Perth, WA, 6009, Australia
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth, WA, 6009, Australia
| | - Rowan W Sanderson
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA, 6009, Australia
| | - Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA, 6009, Australia
| | - Brendan F Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA, 6009, Australia
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziadzka 5, Torun, 87-100, Poland
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Perth, WA, 6009, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA, 6009, Australia
| |
Collapse
|
11
|
Navaeipour F, Hepburn MS, Li J, Metzner KL, Amos SE, Vahala D, Maher S, Choi YS, Kennedy BF. In situ stress estimation in quantitative micro-elastography. BIOMEDICAL OPTICS EXPRESS 2024; 15:3609-3626. [PMID: 38867802 PMCID: PMC11166433 DOI: 10.1364/boe.522002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 06/14/2024]
Abstract
In quantitative micro-elastography (QME), a pre-characterized compliant layer with a known stress-strain curve is utilized to map stress at the sample surface. However, differences in the boundary conditions of the compliant layer when it is mechanically characterized and when it is used in QME experiments lead to inconsistent stress estimation and consequently, inaccurate elasticity measurements. Here, we propose a novel in situ stress estimation method using an optical coherence tomography (OCT)-based uniaxial compression testing system integrated with the QME experimental setup. By combining OCT-measured axial strain with axial stress determined using a load cell in the QME experiments, we can estimate in situ stress for the compliant layer, more accurately considering its boundary conditions. Our proposed method shows improved accuracy, with an error below 10%, compared to 85% using the existing QME technique with no lubrication. Furthermore, demonstrations on hydrogels and cells indicate the potential of this approach for improving the characterization of the micro-scale mechanical properties of cells and their interactions with the surrounding biomaterial, which has potential for application in cell mechanobiology.
Collapse
Affiliation(s)
- Farzaneh Navaeipour
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Western Australia 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia 6009, Australia
| | - Matt S. Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Western Australia 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia 6009, Australia
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziadzka 5, 87-100 Torun, Poland
| | - Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Western Australia 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Kai L. Metzner
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Western Australia 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia 6009, Australia
| | - Sebastian E. Amos
- School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia 6009, Australia
| | - Danielle Vahala
- School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia 6009, Australia
| | - Samuel Maher
- School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia 6009, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia 6009, Australia
| | - Brendan F. Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Western Australia 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia 6009, Australia
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziadzka 5, 87-100 Torun, Poland
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| |
Collapse
|
12
|
Rezaei Z, Navarro Torres A, Ge D, Wang T, Méndez Terán EC, García Vera SE, Bassous NJ, Soria OYP, Ávila Ramírez AE, Flores Campos LM, Azuela Rosas DA, Hassan S, Khorsandi D, Jucaud V, Hussain MA, Khateeb A, Zhang YS, Lee H, Kim DH, Khademhosseini A, Dokmeci MR, Shin SR. Noninvasive and Continuous Monitoring of On-Chip Stem Cell Osteogenesis Using a Reusable Electrochemical Immunobiosensor. ACS Sens 2024; 9:2334-2345. [PMID: 38639453 DOI: 10.1021/acssensors.3c02165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Noninvasive monitoring of biofabricated tissues during the biomanufacturing process is needed to obtain reproducible, healthy, and functional tissues. Measuring the levels of biomarkers secreted from tissues is a promising strategy to understand the status of tissues during biofabrication. Continuous and real-time information from cultivated tissues enables users to achieve scalable manufacturing. Label-free biosensors are promising candidates for detecting cell secretomes since they can be noninvasive and do not require labor-intensive processes such as cell lysing. Moreover, most conventional monitoring techniques are single-use, conducted at the end of the fabrication process, and, challengingly, are not permissive to in-line and continual detection. To address these challenges, we developed a noninvasive and continual monitoring platform to evaluate the status of cells during the biofabrication process, with a particular focus on monitoring the transient processes that stem cells go through during in vitro differentiation over extended periods. We designed and evaluated a reusable electrochemical immunosensor with the capacity for detecting trace amounts of secreted osteogenic markers, such as osteopontin (OPN). The sensor has a low limit of detection (LOD), high sensitivity, and outstanding selectivity in complex biological media. We used this OPN immunosensor to continuously monitor on-chip osteogenesis of human mesenchymal stem cells (hMSCs) cultured 2D and 3D hydrogel constructs inside a microfluidic bioreactor for more than a month and were able to observe changing levels of OPN secretion during culture. The proposed platform can potentially be adopted for monitoring a variety of biological applications and further developed into a fully automated system for applications in advanced cellular biomanufacturing.
Collapse
Affiliation(s)
- Zahra Rezaei
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Sharif University of Technology, Azadi Avenue, Tehran 11365-11155, Iran
| | - Andrea Navarro Torres
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - David Ge
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - Ting Wang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Eloísa Carolina Méndez Terán
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Stefany Elizabeth García Vera
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Nicole Joy Bassous
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - Oscar Yael Perez Soria
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Alan Eduardo Ávila Ramírez
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
- Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Luis Mario Flores Campos
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Diego Arnoldo Azuela Rosas
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- School of Science and Engineering, Tecnologico de Monterrey, Avenida Eugenio Garza Sada 2501 Sur, Monterrey 64849, Mexico
| | - Shabir Hassan
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi 127788, United Arab Emirates
- Functional Biomaterials Group, Khalifa University, SAN Campus, Abu Dhabi 127788, United Arab Emirates
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, California 90024, United States
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, California 90024, United States
| | - Mohammad Asif Hussain
- Electrical and Computer Engineering Department, Faculty of Engineering, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulhameed Khateeb
- Electrical and Computer Engineering Department, Faculty of Engineering, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - HeaYeon Lee
- Mara Nanotech Inc., Hanmir Hall, Yongdang Campus, Pukyong National University, 365 Sinseon-ro, Nam-gu 48548, Republic of Korea
- MARA Nanotech New York INC., NY Designs, 29-10 Thomson Ave, Rm. C760, L.I.C., New York 11101, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, California 90024, United States
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, California 90024, United States
| | - Su Ryon Shin
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
13
|
Nakamura F. The Role of Mechanotransduction in Contact Inhibition of Locomotion and Proliferation. Int J Mol Sci 2024; 25:2135. [PMID: 38396812 PMCID: PMC10889191 DOI: 10.3390/ijms25042135] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Contact inhibition (CI) represents a crucial tumor-suppressive mechanism responsible for controlling the unbridled growth of cells, thus preventing the formation of cancerous tissues. CI can be further categorized into two distinct yet interrelated components: CI of locomotion (CIL) and CI of proliferation (CIP). These two components of CI have historically been viewed as separate processes, but emerging research suggests that they may be regulated by both distinct and shared pathways. Specifically, recent studies have indicated that both CIP and CIL utilize mechanotransduction pathways, a process that involves cells sensing and responding to mechanical forces. This review article describes the role of mechanotransduction in CI, shedding light on how mechanical forces regulate CIL and CIP. Emphasis is placed on filamin A (FLNA)-mediated mechanotransduction, elucidating how FLNA senses mechanical forces and translates them into crucial biochemical signals that regulate cell locomotion and proliferation. In addition to FLNA, trans-acting factors (TAFs), which are proteins or regulatory RNAs capable of directly or indirectly binding to specific DNA sequences in distant genes to regulate gene expression, emerge as sensitive players in both the mechanotransduction and signaling pathways of CI. This article presents methods for identifying these TAF proteins and profiling the associated changes in chromatin structure, offering valuable insights into CI and other biological functions mediated by mechanotransduction. Finally, it addresses unanswered research questions in these fields and delineates their possible future directions.
Collapse
Affiliation(s)
- Fumihiko Nakamura
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| |
Collapse
|
14
|
Ouderkirk S, Sedley A, Ong M, Shifflet MR, Harkrider QC, Wright NT, Miller CJ. A Perspective on Developing Modeling and Image Analysis Tools to Investigate Mechanosensing Proteins. Integr Comp Biol 2023; 63:1532-1542. [PMID: 37558388 PMCID: PMC10755202 DOI: 10.1093/icb/icad107] [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: 05/30/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 08/11/2023] Open
Abstract
The shift of funding organizations to prioritize interdisciplinary work points to the need for workflow models that better accommodate interdisciplinary studies. Most scientists are trained in a specific field and are often unaware of the kind of insights that other disciplines could contribute to solving various problems. In this paper, we present a perspective on how we developed an experimental pipeline between a microscopy and image analysis/bioengineering lab. Specifically, we connected microscopy observations about a putative mechanosensing protein, obscurin, to image analysis techniques that quantify cell changes. While the individual methods used are well established (fluorescence microscopy; ImageJ WEKA and mTrack2 programs; MATLAB), there are no existing best practices for how to integrate these techniques into a cohesive, interdisciplinary narrative. Here, we describe a broadly applicable workflow of how microscopists can more easily quantify cell properties (e.g., perimeter, velocity) from microscopy videos of eukaryotic (MDCK) adherent cells. Additionally, we give examples of how these foundational measurements can create more complex, customizable cell mechanics tools and models.
Collapse
Affiliation(s)
- Stephanie Ouderkirk
- Department of Chemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Alex Sedley
- Department of Engineering, James Madison University, Harrisonburg, VA 22807, USA
| | - Mason Ong
- Department of Engineering, James Madison University, Harrisonburg, VA 22807, USA
| | - Mary Ruth Shifflet
- Department of Chemistry, Bridgewater College, Bridgewater, VA 22812, USA
| | - Quinn C Harkrider
- Department of Chemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Nathan T Wright
- Department of Chemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Callie J Miller
- Department of Engineering, James Madison University, Harrisonburg, VA 22807, USA
| |
Collapse
|
15
|
Cheng L, Zhou Z, Li Q, Li W, Li X, Li G, Fan J, Yu L, Yin G. Dendronized chitosan hydrogel with GIT1 to accelerate bone defect repair through increasing local neovascular amount. Bone Rep 2023; 19:101712. [PMID: 37744736 PMCID: PMC10511783 DOI: 10.1016/j.bonr.2023.101712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/15/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Bone defects have long been a major healthcare issue because of the difficulties in regenerating bone mass volume and the high cost of treatment. G protein-coupled receptor kinase 2 interacting protein 1 (GIT1) has been proven to play an important role both in vascular development and in bone fracture healing. In this study, a type of thermoresponsive injectable hydrogel from oligoethylene glycol-based dendronized chitosan (G1-CS) was loaded with GIT1-plasmids (G1-CS/GIT1), and used to fill unicortical bone defects. RT-PCR analysis confirmed that G1-CS/GIT1 enhanced DNA transfection in MSCs both in vitro and in vivo. From the results of micro-CT, RT-PCR and histological analysis, it can be concluded that G1-CS/GIT1 accelerated the bone healing rate and increased the amount of neovascularization around the bone defects. In addition, an adeno-associated virus (AAV)-GIT1 was constructed to transfect mesenchymal stem cells. The results of capillary tube formation assay, immunofluorescence staining and western blot analysis proved that high expression of GIT1 induces mesenchymal stem cells to differentiate into endothelial cells. RT-PCR analysis and capillary tube formation assay confirmed that the Notch signaling pathway was activated in the differentiation process. Overall, we developed an efficient strategy through combination of injectable hydrogel and G1T1 for bone tissue engineering.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, Jiangsu Province 221000, China
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Zhimin Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Qingqing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Wen Li
- School of Materials Science and Engineering, Shanghai University, Nanchen Street 333, Shanghai 200444, China
| | - Xin Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, Jiangsu Province 221000, China
| | - Gen Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, Jiangsu Province 221000, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Lipeng Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| |
Collapse
|
16
|
Vahala D, Amos SE, Sacchi M, Soliman BG, Hepburn MS, Mowla A, Li J, Jeong JH, Astell C, Hwang Y, Kennedy BF, Lim KS, Choi YS. 3D Volumetric Mechanosensation of MCF7 Breast Cancer Spheroids in a Linear Stiffness Gradient GelAGE. Adv Healthc Mater 2023; 12:e2301506. [PMID: 37670531 PMCID: PMC11481087 DOI: 10.1002/adhm.202301506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/26/2023] [Indexed: 09/07/2023]
Abstract
The tumor microenvironment presents spatiotemporal shifts in biomechanical properties with cancer progression. Hydrogel biomaterials like GelAGE offer the stiffness tuneability to recapitulate dynamic changes in tumor tissues by altering photo-energy exposures. Here, a tuneable hydrogel with spatiotemporal control of stiffness and mesh-network is developed. The volume of MCF7 spheroids encapsulated in a linear stiffness gradient demonstrates an inverse relationship with stiffness (p < 0.0001). As spheroids are exposed to increased crosslinking (stiffer) and greater mechanical confinement, spheroid stiffness increases. Protein expression (TRPV4, β1 integrin, E-cadherin, and F-actin) decreases with increasing stiffness while showing strong correlations to spheroid volume (r2 > 0.9). To further investigate the role of volume, MCF7 spheroids are grown in a soft matrix for 5 days prior to a second polymerisation which presents a stiffness gradient to equally expanded spheroids. Despite being exposed to variable stiffness, these spheroids show even protein expression, confirming volume as a key regulator. Overall, this work showcases the versatility of GelAGE and demonstrates volume expansion as a key regulator of 3D mechanosensation in MCF7 breast cancer spheroids. This platform has the potential to further investigation into the role of stiffness and dimensionality in 3D spheroid culture for other types of cancers and diseases.
Collapse
Affiliation(s)
- Danielle Vahala
- School of Human SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Sebastian E. Amos
- School of Human SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Marta Sacchi
- School of Human SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Bram G. Soliman
- Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of Otago ChristchurchChristchurch8140New Zealand
| | - Matt S. Hepburn
- Department of ElectricalElectronic & Computer EngineeringSchool of EngineeringThe University of Western AustraliaPerthWA6009Australia
- BRITElabHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsand Centre for Medical ResearchThe University of Western AustraliaPerthWA6009Australia
| | - Alireza Mowla
- Department of ElectricalElectronic & Computer EngineeringSchool of EngineeringThe University of Western AustraliaPerthWA6009Australia
- BRITElabHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsand Centre for Medical ResearchThe University of Western AustraliaPerthWA6009Australia
| | - Jiayue Li
- Department of ElectricalElectronic & Computer EngineeringSchool of EngineeringThe University of Western AustraliaPerthWA6009Australia
- BRITElabHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsand Centre for Medical ResearchThe University of Western AustraliaPerthWA6009Australia
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi‐Bio ScienceSoonchunhyang UniversityCheonan‐siChungcheongnam‐do31151South Korea
| | - Chrissie Astell
- School of Human SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi‐Bio ScienceSoonchunhyang UniversityCheonan‐siChungcheongnam‐do31151South Korea
| | - Brendan F. Kennedy
- Department of ElectricalElectronic & Computer EngineeringSchool of EngineeringThe University of Western AustraliaPerthWA6009Australia
- BRITElabHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsand Centre for Medical ResearchThe University of Western AustraliaPerthWA6009Australia
| | - Khoon S. Lim
- Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of Otago ChristchurchChristchurch8140New Zealand
- School of Medical SciencesUniversity of SydneySydneyNSW2006Australia
| | - Yu Suk Choi
- School of Human SciencesThe University of Western AustraliaPerthWA6009Australia
| |
Collapse
|
17
|
Li X, Liu S, Han S, Sun Q, Yang J, Zhang Y, Jiang Y, Wang X, Li Q, Wang J. Dynamic Stiffening Hydrogel with Instructive Stiffening Timing Modulates Stem Cell Fate In Vitro and Enhances Bone Remodeling In Vivo. Adv Healthc Mater 2023; 12:e2300326. [PMID: 37643370 DOI: 10.1002/adhm.202300326] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/24/2023] [Indexed: 08/31/2023]
Abstract
Biomechanical stimuli derived from the extracellular matrix (ECM) extremely tune stem cell fate through 3D and spatiotemporal changes in vivo. The matrix stiffness is a crucial factor during bone tissue development. However, most in vitro models to study the osteogenesis of mesenchymal stem cells (MSCs) are static or stiffening in a 2D environment. Here, a dynamic and controllable stiffening 3D biomimetic model is created to regulate the osteogenic differentiation of MSCs with a dual-functional gelatin macromer that can generate a double-network hydrogel by sequential enzymatic and light-triggered crosslinking reactions. The findings show that these dynamic hydrogels allowed cells to spread and expand prior to the secondary crosslinking and to sense high stiffness after stiffening. The MSCs in the dynamic hydrogels, especially the hydrogel stiffened at the late period, present significantly elevated osteogenic ECM secretion, gene expression, and nuclear localization of Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). In vivo evaluation of animal experiments further indicates that the enhancement of dynamic stiffening on osteogenesis of MSCs substantially promotes bone remodeling. Consequently, this work reveals that the 3D dynamic stiffening microenvironment as a critical biophysical cue not only mediates the stem cell fate in vitro, but also augments bone restoration in vivo.
Collapse
Affiliation(s)
- Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, China
- National Center for International Joint Research of Micro-Nano Moulding Technology, Zhengzhou University, Zhengzhou, 450001, China
| | - Shuaibing Liu
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Shanshan Han
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, China
- National Center for International Joint Research of Micro-Nano Moulding Technology, Zhengzhou University, Zhengzhou, 450001, China
| | - Qingqing Sun
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Yuhang Zhang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, China
- National Center for International Joint Research of Micro-Nano Moulding Technology, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongchao Jiang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, China
- National Center for International Joint Research of Micro-Nano Moulding Technology, Zhengzhou University, Zhengzhou, 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, China
- National Center for International Joint Research of Micro-Nano Moulding Technology, Zhengzhou University, Zhengzhou, 450001, China
| | - Jianglin Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
18
|
Tan K, Yang Q, Han Y, Zhuang Z, Zhao Y, Guo K, Tan A, Zheng Y, Li W. Elastic modulus of hydrogel regulates osteogenic differentiation via liquid-liquid phase separation of YAP. J Biomed Mater Res A 2023; 111:1781-1797. [PMID: 37494632 DOI: 10.1002/jbm.a.37590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 06/26/2023] [Accepted: 07/08/2023] [Indexed: 07/28/2023]
Abstract
Craniofacial bone defects induced by congenital malformations, trauma, or diseases frequently challenge the orthodontic or restorative treatment. Stem cell-based bone regenerative approaches emerged as a promising method to resolve bone defects. Microenvironment physical cues, such as the matrix elastic modulus or matrix topography, regulate stem cell differentiation via multiple genes. We constructed gelatin methacryloyl (GelMA), a well-known scaffold, to investigate the impact of elastic modulus on osteogenic differentiation in a three-dimensional environment. Confocal microscope was used to observe and assess the condensates fission and fusion. New bone formation was evaluated by micro-computed tomography at 6 weeks in calvarial defect rat. We found that the light curing increased elastic modulus of GelMA, and the pore size of GelMA decreased. The expression of osteogenic markers was inhibited in hBMSCs cultured in the low-elastic-modulus GelMA. In contrast, the expression of YAP, TAZ and TEAD was increased in the hBMSCs in the low-elastic-modulus GelMA. Furthermore, YAP assembled via liquid-liquid phase separation (LLPS) into condensates that were sensitive to 1'6-hexanediol. YAP recruit TAZ and TEAD4, but not RUNX2 into the condensates. In vivo, we also found that hBMSCs in high-elastic-modulus GelMA was more apt to form new bone. This study provides new insight into the mechanism of osteogenic differentiation. Reagents that can regulate the elastic modulus of substrate or LLPS may be applied to promote bone regeneration.
Collapse
Affiliation(s)
- Kuang Tan
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - Qiaolin Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - Yineng Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - Ziyao Zhuang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - Yi Zhao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - KunYao Guo
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - Anqi Tan
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - Yunfei Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| | - Weiran Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China
| |
Collapse
|
19
|
Kofler M, Kapus A. Nuclear Import and Export of YAP and TAZ. Cancers (Basel) 2023; 15:4956. [PMID: 37894323 PMCID: PMC10605228 DOI: 10.3390/cancers15204956] [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: 09/04/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Yes-associated Protein (YAP) and its paralog Transcriptional Coactivator with PDZ-binding Motif (TAZ) are major regulators of gene transcription/expression, primarily controlled by the Hippo pathway and the cytoskeleton. Integrating an array of chemical and mechanical signals, they impact growth, differentiation, and regeneration. Accordingly, they also play key roles in tumorigenesis and metastasis formation. Their activity is primarily regulated by their localization, that is, Hippo pathway- and/or cytoskeleton-controlled cytosolic or nuclear sequestration. While many details of such prevailing retention models have been elucidated, much less is known about their actual nuclear traffic: import and export. Although their size is not far from the cutoff for passive diffusion through the nuclear pore complex (NPC), and they do not contain any classic nuclear localization (NLS) or nuclear export signal (NES), evidence has been accumulating that their shuttling involves mediated and thus regulatable/targetable processes. The aim of this review is to summarize emerging information/concepts about their nucleocytoplasmic shuttling, encompassing the relevant structural requirements (NLS, NES), nuclear transport receptors (NTRs, karyophererins), and NPC components, along with the potential transport mechanisms and their regulation. While dissecting retention vs. transport is often challenging, the emerging picture suggests that YAP/TAZ shuttles across the NPC via multiple, non-exclusive, mediated mechanisms, constituting a novel and intriguing facet of YAP/TAZ biology.
Collapse
Affiliation(s)
- Michael Kofler
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
20
|
Zhu Y, Zhang M, Sun Q, Wang X, Li X, Li Q. Advanced Mechanical Testing Technologies at the Cellular Level: The Mechanisms and Application in Tissue Engineering. Polymers (Basel) 2023; 15:3255. [PMID: 37571149 PMCID: PMC10422338 DOI: 10.3390/polym15153255] [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: 07/11/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Mechanics, as a key physical factor which affects cell function and tissue regeneration, is attracting the attention of researchers in the fields of biomaterials, biomechanics, and tissue engineering. The macroscopic mechanical properties of tissue engineering scaffolds have been studied and optimized based on different applications. However, the mechanical properties of the overall scaffold materials are not enough to reveal the mechanical mechanism of the cell-matrix interaction. Hence, the mechanical detection of cell mechanics and cellular-scale microenvironments has become crucial for unraveling the mechanisms which underly cell activities and which are affected by physical factors. This review mainly focuses on the advanced technologies and applications of cell-scale mechanical detection. It summarizes the techniques used in micromechanical performance analysis, including atomic force microscope (AFM), optical tweezer (OT), magnetic tweezer (MT), and traction force microscope (TFM), and analyzes their testing mechanisms. In addition, the application of mechanical testing techniques to cell mechanics and tissue engineering scaffolds, such as hydrogels and porous scaffolds, is summarized and discussed. Finally, it highlights the challenges and prospects of this field. This review is believed to provide valuable insights into micromechanics in tissue engineering.
Collapse
Affiliation(s)
- Yingxuan Zhu
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Mengqi Zhang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qingqing Sun
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
21
|
Chin IL, Amos SE, Jeong JH, Hool L, Hwang Y, Choi YS. Volume adaptation of neonatal cardiomyocyte spheroids in 3D stiffness gradient GelMA. J Biomed Mater Res A 2023; 111:801-813. [PMID: 36239543 PMCID: PMC10952714 DOI: 10.1002/jbm.a.37456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/25/2022] [Accepted: 09/28/2022] [Indexed: 11/11/2022]
Abstract
Present understandings of cardiomyocyte mechanobiology have primarily been developed using 2-dimensional, monocellular cell cultures, however the emergence of 3-dimensional (3D) multicellular cardiac constructs has enabled us to develop more sophisticated recapitulations of the cardiac microenvironment. Several of these strategies have illustrated that incorporating elements of the extracellular matrix (ECM) can promote greater maturation and enhance desirable cardiac functions, such as contractility, but the responses of these cardiac constructs to biophysically aberrant conditions, such as in the post-infarct heart, has remained relatively unexplored. In our study, we employ a stiffness gradient gelatin methacryloyl (GelMA) hydrogel platform to unpack the mechanobiology of cardiac spheroids. We encapsulated neonatal rat cardiac cell spheroids in a 4.4-18.7 kPa linear stiffness gradient up to 120 h. We found the proportion of viable cells within the spheroids increased over time, but the cell number per spheroid decreased. Spheroids expand more in softer matrices while stiffer matrices promote larger nuclei without changing nuclei shape. Volume expansion came primarily from cells expressing vimentin. We did not observe any correlations between stiffness and mechanomarker expression, however we found that after 120 h post-encapsulation, the localization of YAP, the localization of MRTF-A and the expression of Lamin-A was correlated with spheroid morphology. The same trends were not observed 24 h post-encapsulation, indicating that volume adaptation can take a relatively long time. Our data demonstrates that cardiac spheroids are mechanosensitive and that their capacity to respond to ECM-based cues depends on their capacity to adapt their volume with a 3D microenvironment.
Collapse
Affiliation(s)
- Ian L. Chin
- School of Human SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Sebastian E. Amos
- School of Human SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi‐bio Science (SIMS)Soonchunhyang UniversityCheonan‐siChungnam‐doRepublic of Korea
- Department of Integrated Biomedical ScienceSoonchunhyang UniversityAsan‐siChungnam‐doRepublic of Korea
| | - Livia Hool
- School of Human SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
- Victor Chang Cardiac Research InstituteSydneyNew South WalesAustralia
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi‐bio Science (SIMS)Soonchunhyang UniversityCheonan‐siChungnam‐doRepublic of Korea
- Department of Integrated Biomedical ScienceSoonchunhyang UniversityAsan‐siChungnam‐doRepublic of Korea
| | - Yu Suk Choi
- School of Human SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| |
Collapse
|
22
|
Oliver-Cervelló L, Martin-Gómez H, Gonzalez-Garcia C, Salmeron-Sanchez M, Ginebra MP, Mas-Moruno C. Protease-degradable hydrogels with multifunctional biomimetic peptides for bone tissue engineering. Front Bioeng Biotechnol 2023; 11:1192436. [PMID: 37324414 PMCID: PMC10267393 DOI: 10.3389/fbioe.2023.1192436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Mimicking bone extracellular matrix (ECM) is paramount to develop novel biomaterials for bone tissue engineering. In this regard, the combination of integrin-binding ligands together with osteogenic peptides represents a powerful approach to recapitulate the healing microenvironment of bone. In the present work, we designed polyethylene glycol (PEG)-based hydrogels functionalized with cell instructive multifunctional biomimetic peptides (either with cyclic RGD-DWIVA or cyclic RGD-cyclic DWIVA) and cross-linked with matrix metalloproteinases (MMPs)-degradable sequences to enable dynamic enzymatic biodegradation and cell spreading and differentiation. The analysis of the intrinsic properties of the hydrogel revealed relevant mechanical properties, porosity, swelling and degradability to engineer hydrogels for bone tissue engineering. Moreover, the engineered hydrogels were able to promote human mesenchymal stem cells (MSCs) spreading and significantly improve their osteogenic differentiation. Thus, these novel hydrogels could be a promising candidate for applications in bone tissue engineering, such as acellular systems to be implanted and regenerate bone or in stem cells therapy.
Collapse
Affiliation(s)
- Lluís Oliver-Cervelló
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| | - Helena Martin-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| | - Cristina Gonzalez-Garcia
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, Spain
| |
Collapse
|
23
|
Pragya A, Ghosh TK. Soft Functionally Gradient Materials and Structures - Natural and Manmade: A Review. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2300912. [PMID: 37031358 DOI: 10.1002/adma.202300912] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/31/2023] [Indexed: 06/19/2023]
Abstract
Functionally gradient materials (FGM) have gradual variations in their properties along one or more dimensions due to local compositional or structural distinctions by design. Traditionally, hard materials (e.g., metals, ceramics) are used to design and fabricate FGMs; however, there is increasing interest in polymer-based soft and compliant FGMs mainly because of their potential application in the human environment. Soft FGMs are ideally suitable to manage interfacial problems in dissimilar materials used in many emerging devices and systems for human interaction, such as soft robotics and electronic textiles and beyond. Soft systems are ubiquitous in everyday lives; they are resilient and can easily deform, absorb energy, and adapt to changing environments. Here, the basic design and functional principles of biological FGMs and their manmade counterparts are discussed using representative examples. The remarkable multifunctional properties of natural FGMs resulting from their sophisticated hierarchical structures, built from a relatively limited choice of materials, offer a rich source of new design paradigms and manufacturing strategies for manmade materials and systems for emerging technological needs. Finally, the challenges and potential pathways are highlighted to leverage soft materials' facile processability and unique properties toward functional FGMs.
Collapse
Affiliation(s)
- Akanksha Pragya
- Department of Textile Engineering Chemistry and Science, Fiber, and Polymer Science Program, Wilson College of Textiles, North Carolina State University, North Carolina State University, 1020 Main Campus Drive, Raleigh, NC, 27606, USA
| | - Tushar K Ghosh
- Department of Textile Engineering Chemistry and Science, Fiber, and Polymer Science Program, Wilson College of Textiles, North Carolina State University, North Carolina State University, 1020 Main Campus Drive, Raleigh, NC, 27606, USA
| |
Collapse
|
24
|
A maladaptive feedback mechanism between the extracellular matrix and cytoskeleton contributes to hypertrophic cardiomyopathy pathophysiology. Commun Biol 2023; 6:4. [PMID: 36596888 PMCID: PMC9810744 DOI: 10.1038/s42003-022-04278-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 11/17/2022] [Indexed: 01/04/2023] Open
Abstract
Hypertrophic cardiomyopathy is an inherited disorder due to mutations in contractile proteins that results in a stiff, hypercontractile myocardium. To understand the role of cardiac stiffness in disease progression, here we create an in vitro model of hypertrophic cardiomyopathy utilizing hydrogel technology. Culturing wild-type cardiac myocytes on hydrogels with a Young's Moduli (stiffness) mimicking hypertrophic cardiomyopathy myocardium is sufficient to induce a hypermetabolic mitochondrial state versus myocytes plated on hydrogels simulating healthy myocardium. Significantly, these data mirror that of myocytes isolated from a murine model of human hypertrophic cardiomyopathy (cTnI-G203S). Conversely, cTnI-G203S myocyte mitochondrial function is completely restored when plated on hydrogels mimicking healthy myocardium. We identify a mechanosensing feedback mechanism between the extracellular matrix and cytoskeletal network that regulates mitochondrial function under healthy conditions, but participates in the progression of hypertrophic cardiomyopathy pathophysiology resulting from sarcomeric gene mutations. Importantly, we pinpoint key 'linker' sites in this schema that may represent potential therapeutic targets.
Collapse
|
25
|
Zhang H, Ma Y, Wang Y, Niu L, Zou R, Zhang M, Liu H, Genin GM, Li A, Xu F. Rational Design of Soft-Hard Interfaces through Bioinspired Engineering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204498. [PMID: 36228093 DOI: 10.1002/smll.202204498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Soft-hard tissue interfaces in nature present a diversity of hierarchical transitions in composition and structure to address the challenge of stress concentrations that would otherwise arise at their interface. The translation of these into engineered materials holds promise for improved function of biomedical interfaces. Here, soft-hard tissue interfaces found in the body in health and disease, and the application of the diverse, functionally graded, and hierarchical structures that they present to bioinspired engineering materials are reviewed. A range of such bioinspired engineering materials and associated manufacturing technologies that are on the horizon in interfacial tissue engineering, hydrogel bioadhesion at the interfaces, and healthcare and medical devices are described.
Collapse
Affiliation(s)
- Hui Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Min Zhang
- State Key Laboratory of Military Stomatology, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Hao Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, 63130, USA
- NSF Science and Technology Center for Engineering MechanoBiology, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
26
|
Ghuloum FI, Johnson CA, Riobo-Del Galdo NA, Amer MH. From mesenchymal niches to engineered in vitro model systems: Exploring and exploiting biomechanical regulation of vertebrate hedgehog signalling. Mater Today Bio 2022; 17:100502. [PMID: 36457847 PMCID: PMC9707069 DOI: 10.1016/j.mtbio.2022.100502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
Tissue patterning is the result of complex interactions between transcriptional programs and various mechanical cues that modulate cell behaviour and drive morphogenesis. Vertebrate Hedgehog signalling plays key roles in embryogenesis and adult tissue homeostasis, and is central to skeletal development and the osteogenic differentiation of mesenchymal stem cells. The expression of several components of the Hedgehog signalling pathway have been reported to be mechanically regulated in mesodermal tissue patterning and osteogenic differentiation in response to external stimulation. Since a number of bone developmental defects and skeletal diseases, such as osteoporosis, are directly linked to aberrant Hedgehog signalling, a better knowledge of the regulation of Hedgehog signalling in the mechanosensitive bone marrow-residing mesenchymal stromal cells will present novel avenues for modelling these diseases and uncover novel opportunities for extracellular matrix-targeted therapies. In this review, we present a brief overview of the key molecular players involved in Hedgehog signalling and the basic concepts of mechanobiology, with a focus on bone development and regeneration. We also highlight the correlation between the activation of the Hedgehog signalling pathway in response to mechanical cues and osteogenesis in bone marrow-derived mesenchymal stromal cells. Finally, we propose different tissue engineering strategies to apply the expanding knowledge of 3D material-cell interactions in the modulation of Hedgehog signalling in vitro for fundamental and translational research applications.
Collapse
Affiliation(s)
- Fatmah I. Ghuloum
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Colin A. Johnson
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natalia A. Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, UK
| | - Mahetab H. Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
27
|
Chin IL, Amos SE, Jeong JH, Hool L, Hwang Y, Choi YS. Mechanosensation mediates volume adaptation of cardiac cells and spheroids in 3D. Mater Today Bio 2022; 16:100391. [PMID: 36042852 PMCID: PMC9420370 DOI: 10.1016/j.mtbio.2022.100391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022]
Abstract
With the adoption of 3-dimensional (3D) cell culture for in vitro modelling of cardiac function and regenerative medicine applications, there is an increased need to understand cardiomyocyte mechanosensation in 3D. With existing studies of cardiomyocyte mechanosensation primarily focussed on the behaviour of individual cells in a 2-Dimensional context, it is unclear whether mechanosensation is the same in a 3D, multicellular context. In this study, H9C2 cardiac-derived myoblasts were encapsulated as individual cells and as cell spheroids within stiffness gradient gelatin methacryloyl (GelMA) hydrogels to investigate individual and collective cardiac cell mechanosensation in 3D. Over a 3.68–17.52 kPa stiffness range, it was found that H9C2 cells have a limited capacity to adapt their volume to increasing substrate stiffness, demonstrated by the lack of changes in cell volume and shape across the stiffness gradient. Morphological trends were reflected by the expression of the mechanomarkers YAP, MRTF-A and Lamin-A, which were better correlated with cell and nuclear volume than with substrate stiffness. The localisation of YAP and MRTF-A were dependent on the relative volumes of the cytoplasm and nucleus while Lamin-A expression was elevated with increasing cytoplasmic and nuclear volumes. When cultured as spheroids rather than as individual cells, H9C2 cells adopted a distinct morphology with comparably smaller nuclei than individually cultured cells, while retaining the same overall cell volume. As spheroids, H9C2 cells were sensitive to stiffness cues, shown by decreasing YAP and MRTF-A nuclear localisation, increasing Lamin-A expression, and increasing vinculin expression with increasing substrate stiffness. Like the individually cultured H9C2 cells, mechanomarker expression was correlated to volume adaptation. With increasing cytoplasmic volume, YAP and MRTF-A became less nuclear localised, vinculin expression was increased, and with increasing nuclear volume, the Lamin-A expression fincreased. Together, these data suggest that cardiac cell volume adaptation may be enhanced by cell-cell interactions.
Collapse
Affiliation(s)
- Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Sebastian E Amos
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungnam-do, 31151, Republic of Korea.,Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si, Chungnam-do, 31538, Republic of Korea
| | - Livia Hool
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.,Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Chungnam-do, 31151, Republic of Korea.,Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si, Chungnam-do, 31538, Republic of Korea
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
28
|
Lloyd EM, Hepburn MS, Li J, Mowla A, Hwang Y, Choi YS, Grounds MD, Kennedy BF. Three-dimensional mechanical characterization of murine skeletal muscle using quantitative micro-elastography. BIOMEDICAL OPTICS EXPRESS 2022; 13:5879-5899. [PMID: 36733728 PMCID: PMC9872891 DOI: 10.1364/boe.471062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 06/18/2023]
Abstract
Skeletal muscle function is governed by both the mechanical and structural properties of its constituent tissues, which are both modified by disease. Characterizing the mechanical properties of skeletal muscle tissue at an intermediate scale, i.e., between that of cells and organs, can provide insight into diseases such as muscular dystrophies. In this study, we use quantitative micro-elastography (QME) to characterize the micro-scale elasticity of ex vivo murine skeletal muscle in three-dimensions in whole muscles. To address the challenge of achieving high QME image quality with samples featuring uneven surfaces and geometry, we encapsulate the muscles in transparent hydrogels with flat surfaces. Using this method, we study aging and disease in quadriceps tissue by comparing normal wild-type (C57BL/6J) mice with dysferlin-deficient BLAJ mice, a model for the muscular dystrophy dysferlinopathy, at 3, 10, and 24 months of age (sample size of three per group). We observe a 77% decrease in elasticity at 24 months in dysferlin-deficient quadriceps compared to wild-type quadriceps.
Collapse
Affiliation(s)
- Erin M. Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia
- These authors contributed equally to this work
| | - Matt S. Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
- These authors contributed equally to this work
| | - Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Alireza Mowla
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, 31151, Republic of Korea
| | - Yu Suk Choi
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia
| | - Miranda D. Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia
| | - Brendan F. Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| |
Collapse
|
29
|
Wang Q, Wang Z, Zhang D, Gu J, Ma Y, Zhang Y, Chen J. Circular Patterns of Dynamic Covalent Hydrogels with Gradient Stiffness for Screening of the Stem Cell Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:47461-47471. [PMID: 36240467 DOI: 10.1021/acsami.2c14924] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
As extracellular matrix (ECM) mimetic materials, hydrogels have been widely used for broad biomedical applications. However, with so many physical or chemical cues in the matrix that regulate cell behaviors or functions, it remains challenging to design a customizable hydrogel with the desired properties on demand. In the current study, we aim to establish a circular-patterned hydrogel model with gradient stiffness for screening the most favorable ECM environment for specific cells or certain application purposes. First, six types of hydrogels with a wide stiffness range of 1.2-28.9 kPa were prepared by dynamic covalent cross-linking between gelatin derivatives and oxidized hyaluronic acid. Taking advantage of their instantaneous self-healing property from dynamic chemistry, the hydrogels were further spliced into one whole piece of circular-patterned hydrogel. When rabbit bone marrow mesenchymal stem cells were seeded in the center, the influences of matrix stiffness on the regulation of stem cell adhesion, migration, and differentiation were directly observed and compared under one visual field. In addition, these hydrogels all possessed good biocompatibility, degradability, and injectability, showing great potential for tissue-engineering-related applications.
Collapse
Affiliation(s)
- Qimeng Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Ziyan Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Difei Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Jieyu Gu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Yongxin Ma
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Yan Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Jinghua Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| |
Collapse
|
30
|
Chalard AE, Dixon AW, Taberner AJ, Malmström J. Visible-Light Stiffness Patterning of GelMA Hydrogels Towards In Vitro Scar Tissue Models. Front Cell Dev Biol 2022; 10:946754. [PMID: 35865624 PMCID: PMC9294371 DOI: 10.3389/fcell.2022.946754] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022] Open
Abstract
Variations in mechanical properties of the extracellular matrix occurs in various processes, such as tissue fibrosis. The impact of changes in tissue stiffness on cell behaviour are studied in vitro using various types of biomaterials and methods. Stiffness patterning of hydrogel scaffolds, through the use of stiffness gradients for instance, allows the modelling and studying of cellular responses to fibrotic mechanisms. Gelatine methacryloyl (GelMA) has been used extensively in tissue engineering for its inherent biocompatibility and the ability to precisely tune its mechanical properties. Visible light is now increasingly employed for crosslinking GelMA hydrogels as it enables improved cell survival when performing cell encapsulation. We report here, the photopatterning of mechanical properties of GelMA hydrogels with visible light and eosin Y as the photoinitiator using physical photomasks and projection with a digital micromirror device. Using both methods, binary hydrogels with areas of different stiffnesses and hydrogels with stiffness gradients were fabricated. Their mechanical properties were characterised using force indentation with atomic force microscopy, which showed the efficiency of both methods to spatially pattern the elastic modulus of GelMA according to the photomask or the projected pattern. Crosslinking through projection was also used to build constructs with complex shapes. Overall, this work shows the feasibility of patterning the stiffness of GelMA scaffolds, in the range from healthy to pathological stiffness, with visible light. Consequently, this method could be used to build in vitro models of healthy and fibrotic tissue and study the cellular behaviours involved at the interface between the two.
Collapse
Affiliation(s)
- Anaïs E. Chalard
- Department of Chemical and Materials Engineering, Faculty of Engineering, The University of Auckland, Auckland, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand
- *Correspondence: Anaïs E. Chalard, ; Jenny Malmström,
| | - Alexander W. Dixon
- The Auckland Bioengineering Institute (ABI), The University of Auckland, Auckland, New Zealand
| | - Andrew J. Taberner
- The Auckland Bioengineering Institute (ABI), The University of Auckland, Auckland, New Zealand
- Department of Engineering Science, Faculty of Engineering, The University of Auckland, Auckland, New Zealand
| | - Jenny Malmström
- Department of Chemical and Materials Engineering, Faculty of Engineering, The University of Auckland, Auckland, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand
- *Correspondence: Anaïs E. Chalard, ; Jenny Malmström,
| |
Collapse
|
31
|
Vahala D, Choi YS. Modelling the Tumor Microenvironment: Recapitulating Nano- and Micro-Scale Properties that Regulate Tumor Progression. Front Cell Dev Biol 2022; 10:908799. [PMID: 35800896 PMCID: PMC9254080 DOI: 10.3389/fcell.2022.908799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 12/02/2022] Open
Abstract
Breast cancer remains a significant burden with 1 in 8 women affected and metastasis posing a significant challenge for patient survival. Disease progression involves remodeling of the extracellular matrix (ECM). In breast cancer, tissue stiffness increases owing to an increase in collagen production by recruited cancer-associated fibroblasts (CAFs). These stromal modifications are notable during primary tumor growth and have a dualistic action by creating a hard capsule to prevent penetration of anti-cancer therapies and forming a favorable environment for tumor progression. Remodeling of the tumor microenvironment immediately presented to cells can include changes in protein composition, concentration and structural arrangement and provides the first mechanical stimuli in the metastatic cascade. Not surprisingly, metastatic cancer cells possess the ability to mechanically adapt, and their adaptability ensures not only survival but successful invasion within altered environments. In the past decade, the importance of the microenvironment and its regulatory role in diseases have gained traction and this is evident in the shift from plastic culture to the development of novel biomaterials that mimic in vivo tissue. With these advances, elucidations can be made into how ECM remodeling and more specifically, altered cell-ECM adhesions, regulate tumor growth and cancer cell plasticity. Such enabling tools in mechanobiology will identify fundamental mechanisms in cancer progression that eventually help develop preventative and therapeutic treatment from a clinical perspective. This review will focus on current platforms engineered to mimic the micro and nano-properties of the tumor microenvironment and subsequent understanding of mechanically regulated pathways in cancer.
Collapse
|
32
|
Li J, Pijewska E, Fang Q, Szkulmowski M, Kennedy BF. Analysis of strain estimation methods in phase-sensitive compression optical coherence elastography. BIOMEDICAL OPTICS EXPRESS 2022; 13:2224-2246. [PMID: 35519281 PMCID: PMC9045929 DOI: 10.1364/boe.447340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 05/11/2023]
Abstract
In compression optical coherence elastography (OCE), deformation is quantified as the local strain at each pixel in the OCT field-of-view. A range of strain estimation methods have been demonstrated, yet it is unclear which method provides the best performance. Here, we analyze the two most prevalent strain estimation methods used in phase-sensitive compression OCE, i.e., weighted least squares (WLS) and the vector method. We introduce a framework to compare strain imaging metrics, incorporating strain sensitivity, strain signal-to-noise ratio (SNR), strain resolution, and strain accuracy. In addition, we propose a new phase unwrapping algorithm in OCE, fast phase unwrapping (FPU), and combine it with WLS, termed WLSFPU. Using the framework, we compare this new strain estimation method with both a current implementation of WLS that incorporates weighted phase unwrapping (WPU), termed WLSWPU, and the vector method. Our analysis reveals that the three methods provide similar strain sensitivity, strain SNR, and strain resolution, but that WLSFPU extends the dynamic range of accurate, measurable local strain, e.g., measuring a strain of 2.5 mɛ with ∼4% error, that is ×11 and ×15 smaller than the error measured using WLSWPU and the vector method, respectively. We also demonstrate, for the first time, the capability to detect sub-resolution contrast in compression OCE, i.e., changes in strain occurring within the strain axial resolution, and how this contrast varies between the different strain estimation methods. Lastly, we compare the performance of the three strain estimation methods on mouse skeletal muscle and human breast tissue and demonstrate that WLSFPU avoids strain imaging artifacts resulting from phase unwrapping errors in WLSWPU and provides improved contrast over the other two methods.
Collapse
Affiliation(s)
- Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Crawley 6009, Australia
- Australian Research Council Centre for Personalized Therapeutics Technologies, Australia
- These authors contributed equally to this work
| | - Ewelina Pijewska
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziądzka 5, 87-100 Torun, Poland
- These authors contributed equally to this work
| | - Qi Fang
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Crawley 6009, Australia
| | - Maciej Szkulmowski
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziądzka 5, 87-100 Torun, Poland
| | - Brendan F. Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Crawley 6009, Australia
- Australian Research Council Centre for Personalized Therapeutics Technologies, Australia
| |
Collapse
|
33
|
Luciano M, Versaevel M, Vercruysse E, Procès A, Kalukula Y, Remson A, Deridoux A, Gabriele S. Appreciating the role of cell shape changes in the mechanobiology of epithelial tissues. BIOPHYSICS REVIEWS 2022; 3:011305. [PMID: 38505223 PMCID: PMC10903419 DOI: 10.1063/5.0074317] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/23/2022] [Indexed: 03/21/2024]
Abstract
The wide range of epithelial cell shapes reveals the complexity and diversity of the intracellular mechanisms that serve to construct their morphology and regulate their functions. Using mechanosensitive steps, epithelial cells can sense a variety of different mechanochemical stimuli and adapt their behavior by reshaping their morphology. These changes of cell shape rely on a structural reorganization in space and time that generates modifications of the tensional state and activates biochemical cascades. Recent studies have started to unveil how the cell shape maintenance is involved in mechanical homeostatic tasks to sustain epithelial tissue folding, identity, and self-renewal. Here, we review relevant works that integrated mechanobiology to elucidate some of the core principles of how cell shape may be conveyed into spatial information to guide collective processes such as epithelial morphogenesis. Among many other parameters, we show that the regulation of the cell shape can be understood as the result of the interplay between two counteracting mechanisms: actomyosin contractility and intercellular adhesions, and that both do not act independently but are functionally integrated to operate on molecular, cellular, and tissue scales. We highlight the role of cadherin-based adhesions in force-sensing and mechanotransduction, and we report recent developments that exploit physics of liquid crystals to connect cell shape changes to orientational order in cell aggregates. Finally, we emphasize that the further intermingling of different disciplines to develop new mechanobiology assays will lead the way toward a unified picture of the contribution of cell shape to the pathophysiological behavior of epithelial tissues.
Collapse
Affiliation(s)
- Marine Luciano
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Marie Versaevel
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Eléonore Vercruysse
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Anthony Procès
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Yohalie Kalukula
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Alexandre Remson
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Amandine Deridoux
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Sylvain Gabriele
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| |
Collapse
|
34
|
Zarka M, Haÿ E, Cohen-Solal M. YAP/TAZ in Bone and Cartilage Biology. Front Cell Dev Biol 2022; 9:788773. [PMID: 35059398 PMCID: PMC8764375 DOI: 10.3389/fcell.2021.788773] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022] Open
Abstract
YAP and TAZ were initially described as the main regulators of organ growth during development and more recently implicated in bone biology. YAP and TAZ are regulated by mechanical and cytoskeletal cues that lead to the control of cell fate in response to the cellular microenvironment. The mechanical component represents a major signal for bone tissue adaptation and remodelling, so YAP/TAZ contributes significantly in bone and cartilage homeostasis. Recently, mice and cellular models have been developed to investigate the precise roles of YAP/TAZ in bone and cartilage cells, and which appear to be crucial. This review provides an overview of YAP/TAZ regulation and function, notably providing new insights into the role of YAP/TAZ in bone biology.
Collapse
Affiliation(s)
- Mylène Zarka
- INSERM UMR 1132 BIOSCAR, Hôpital Lariboisière, Paris, France.,Faculté de Santé, Université de Paris, Paris, France
| | - Eric Haÿ
- INSERM UMR 1132 BIOSCAR, Hôpital Lariboisière, Paris, France.,Faculté de Santé, Université de Paris, Paris, France
| | - Martine Cohen-Solal
- INSERM UMR 1132 BIOSCAR, Hôpital Lariboisière, Paris, France.,Faculté de Santé, Université de Paris, Paris, France
| |
Collapse
|
35
|
Afzali Z, Matsushita T, Kogure A, Masuda T, Azuma T, Kushiro K, Kasama T, Miyake R, Takai M. Cell Adhesion and Migration on Thickness Gradient Bilayer Polymer Brush Surfaces: Effects of Properties of Polymeric Materials of the Underlayer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:2605-2617. [PMID: 35001615 DOI: 10.1021/acsami.1c21453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In the field of tissue engineering and biomaterials, controlling the surface properties and mechanical properties of scaffold materials is crucial and has attracted much attention. Here, two types of bilayer polymer brushes composed of a hydrophilic underlying layer and a cationic surface layer [made of poly(2-aminoethyl methacrylate)] with a thickness gradient were prepared by surface-initiated atom-transfer radical polymerization. To investigate the influence of the stiffness as a mechanical property of the polymer brush on cell behavior, the underlayer was prepared from either 2-methacryloyloxyethyl phosphorylcholine or oligo(ethylene glycol) methyl ether methacrylate, with the bilayers designated as gradient poly(2-methacryloyloxyethyl phosphorylcholine)-block-poly(2-aminoethyl methacrylate) [grad-pMbA] and gradient poly(oligo[ethylene glycol] methyl ether methacrylate)-block-poly(2-aminoethyl methacrylate) [grad-pEGbA], respectively. Characterization of these surfaces was performed by spectroscopic ellipsometry, X-ray reflectivity, and determination of the zeta potential, static contact angle, and force curve. These diblock copolymer brushes with a thickness gradient helped to distinguish the effects of the mechanical and surface properties of the brushes on cell behavior. The attachment and motility of L929 fibroblasts and epithelial MCF 10A cells on the fabricated brushes were then assessed. L929 cells had a round shape on the thin surface layer of grad-pMbA and spread well on thicker areas. In contrast, MCF 10A cells spread well in areas of any thickness of either grad-pMbA or grad-pEGbA. Single MCF 10A cells migrated randomly on grad-pMbA, whereas grouped cells started to climb up along the thickness gradient of grad-pMbA. In contrast, both single and grouped MCF 10A cells migrated randomly on grad-pEGbA. These thickness gradient diblock copolymer brushes are simple, reproducible, and reasonable platforms that can facilitate practical applications of biomaterials, for example, in tissue engineering and biomaterials.
Collapse
Affiliation(s)
- Zahra Afzali
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Taishi Matsushita
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Akinori Kogure
- Shimadzu Techno-Research, Inc., 380-1 Horiyamashita, Hatano-city, Kanagawa 259-1304, Japan
| | - Tsukuru Masuda
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tomoyuki Azuma
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Keiichiro Kushiro
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Toshihiro Kasama
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Ryo Miyake
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Madoka Takai
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
36
|
Han P, Gomez GA, Duda GN, Ivanovski S, Poh PS. Scaffold geometry modulation of mechanotransduction and its influence on epigenetics. Acta Biomater 2022; 163:259-274. [PMID: 35038587 DOI: 10.1016/j.actbio.2022.01.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 02/03/2023]
Abstract
The dynamics of cell mechanics and epigenetic signatures direct cell behaviour and fate, thus influencing regenerative outcomes. In recent years, the utilisation of 2D geometric (i.e. square, circle, hexagon, triangle or round-shaped) substrates for investigating cell mechanics in response to the extracellular microenvironment have gained increasing interest in regenerative medicine due to their tunable physicochemical properties. In contrast, there is relatively limited knowledge of cell mechanobiology and epigenetics in the context of 3D biomaterial matrices, i.e., hydrogels and scaffolds. Scaffold geometry provides biophysical signals that trigger a nucleus response (regulation of gene expression) and modulates cell behaviour and function. In this review, we explore the potential of additive manufacturing to incorporate multi length-scale geometry features on a scaffold. Then, we discuss how scaffold geometry direct cell and nuclear mechanosensing. We further discuss how cell epigenetics, particularly DNA/histone methylation and histone acetylation, are modulated by scaffold features that lead to specific gene expression and ultimately influence the outcome of tissue regeneration. Overall, we highlight that geometry of different magnitude scales can facilitate the assembly of cells and multicellular tissues into desired functional architectures through the mechanotransduction pathway. Moving forward, the challenge confronting biomedical engineers is the distillation of the vast knowledge to incorporate multiscaled geometrical features that would collectively elicit a favourable tissue regeneration response by harnessing the design flexibility of additive manufacturing. STATEMENT OF SIGNIFICANCE: It is well-established that cells sense and respond to their 2D geometric microenvironment by transmitting extracellular physiochemical forces through the cytoskeleton and biochemical signalling to the nucleus, facilitating epigenetic changes such as DNA methylation, histone acetylation, and microRNA expression. In this context, the current review presents a unique perspective and highlights the importance of 3D architectures (dimensionality and geometries) on cell and nuclear mechanics and epigenetics. Insight into current challenges around the study of mechanobiology and epigenetics utilising additively manufactured 3D scaffold geometries will progress biomaterials research in this space.
Collapse
|
37
|
Bonany M, del-Mazo-Barbara L, Espanol M, Ginebra MP. Microsphere incorporation as a strategy to tune the biological performance of bioinks. J Tissue Eng 2022; 13:20417314221119895. [PMID: 36199978 PMCID: PMC9527984 DOI: 10.1177/20417314221119895] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Although alginate is widely used as a matrix in the formulation of cell-laden inks, this polymer often requires laborious processing strategies due to its lack of cell adhesion moieties. The main objective of the present work was to explore the incorporation of microspheres into alginate-based bioinks as a simple and tuneable way to solve the cell adhesion problems, while adding extra biological functionality and improving their mechanical properties. To this end, three types of microspheres with different mineral contents (i.e. gelatine with 0% of hydroxyapatite, gelatine with 25 wt% of hydroxyapatite nanoparticles and 100 wt% of calcium -deficient hydroxyapatite) were synthesised and incorporated into the formulation of cell-laden inks. The results showed that the addition of microspheres generally improved the rheological properties of the ink, favoured cell proliferation and positively affected osteogenic cell differentiation. Furthermore, this differentiation was found to be influenced by the type of microsphere and the ability of the cells to migrate towards them, which was highly dependent on the stiffness of the bioink. In this regard, Ca2+ supplementation in the cell culture medium had a pronounced effect on the relaxation of the stiffness of these cell-loaded inks, influencing the overall cell performance. In conclusion, we have developed a powerful and tuneable strategy for the fabrication of alginate-based bioinks with enhanced biological characteristics by incorporating microspheres into the initial ink formulation.
Collapse
Affiliation(s)
- Mar Bonany
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Centre in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Biomedical Engineering Research Center (CREB), UPC, Barcelona, Spain
| | - Laura del-Mazo-Barbara
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Centre in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Biomedical Engineering Research Center (CREB), UPC, Barcelona, Spain
| | - Montserrat Espanol
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Centre in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Biomedical Engineering Research Center (CREB), UPC, Barcelona, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Barcelona Research Centre in Multiscale Science and Engineering, UPC, Barcelona, Spain
- Biomedical Engineering Research Center (CREB), UPC, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|
38
|
Yuan X, Li L, Liu H, Luo J, Zhao Y, Pan C, Zhang X, Chen Y, Gou M. Strategies for improving adipose-derived stem cells for tissue regeneration. BURNS & TRAUMA 2022; 10:tkac028. [PMID: 35992369 PMCID: PMC9382096 DOI: 10.1093/burnst/tkac028] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/27/2022] [Indexed: 11/13/2022]
Abstract
Abstract
Adipose-derived stem cells (ADSCs) have promising applications in tissue regeneration. Currently, there are only a few ADSC products that have been approved for clinical use. The clinical application of ADSCs still faces many challenges. Here, we review emerging strategies to improve the therapeutic efficacy of ADSCs in tissue regeneration. First, a great quantity of cells is often needed for the stem cell therapies, which requires the advanced cell expansion technologies. In addition cell-derived products are also required for the development of ‘cell-free’ therapies to overcome the drawbacks of cell-based therapies. Second, it is necessary to strengthen the regenerative functions of ADSCs, including viability, differentiation and paracrine ability, for the tissue repair and regeneration required for different physiological and pathophysiological conditions. Third, poor delivery efficiency also restricts the therapeutic effect of ADSCs. Effective methods to improve cell delivery include alleviating harsh microenvironments, enhancing targeting ability and prolonging cell retention. Moreover, we also point out some critical issues about the sources, effectiveness and safety of ADSCs. With these advanced strategies to improve the therapeutic efficacy of ADSCs, ADSC-based treatment holds great promise for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Xin Yuan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Li Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Haofan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Jing Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Yongchao Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Cheng Pan
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Xue Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Yuwen Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University , Chengdu, 610041, China
| |
Collapse
|
39
|
Wei L, Shi J. Insight Into Rho Kinase Isoforms in Obesity and Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:886534. [PMID: 35769086 PMCID: PMC9234286 DOI: 10.3389/fendo.2022.886534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity and associated complications increasingly jeopardize global health and contribute to the rapidly rising prevalence of type 2 diabetes mellitus and obesity-related diseases. Developing novel methods for the prevention and treatment of excess body adipose tissue expansion can make a significant contribution to public health. Rho kinase is a Rho-associated coiled-coil-containing protein kinase (Rho kinase or ROCK). The ROCK family including ROCK1 and ROCK2 has recently emerged as a potential therapeutic target for the treatment of metabolic disorders. Up-regulated ROCK activity has been involved in the pathogenesis of all aspects of metabolic syndrome including obesity, insulin resistance, dyslipidemia and hypertension. The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in both white and beige adipogenesis. Studies using ROCK pan-inhibitors in animal models of obesity, diabetes, and associated complications have demonstrated beneficial outcomes. Studies via genetically modified animal models further established isoform-specific roles of ROCK in the pathogenesis of metabolic disorders including obesity. However, most reported studies have been focused on ROCK1 activity during the past decade. Due to the progress in developing ROCK2-selective inhibitors in recent years, a growing body of evidence indicates more attention should be devoted towards understanding ROCK2 isoform function in metabolism. Hence, studying individual ROCK isoforms to reveal their specific roles and principal mechanisms in white and beige adipogenesis, insulin sensitivity, energy balancing regulation, and obesity development will facilitate significant breakthroughs for systemic treatment with isoform-selective inhibitors. In this review, we give an overview of ROCK functions in the pathogenesis of obesity and insulin resistance with a particular focus on the current understanding of ROCK isoform signaling in white and beige adipogenesis, obesity and thermogenesis in adipose tissue and other major metabolic organs involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lei Wei
- *Correspondence: Lei Wei, ; Jianjian Shi,
| | | |
Collapse
|
40
|
Chin IL, Hool L, Choi YS. Interrogating cardiac muscle cell mechanobiology on stiffness gradient hydrogels. Biomater Sci 2021; 9:6795-6806. [PMID: 34542112 DOI: 10.1039/d1bm01061a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Extracellular matrix (ECM) remodeling is a major facet of cardiac development and disease, yet our understanding of cardiomyocyte mechanotransduction remains limited. To enhance our understanding of cardiomyocyte mechanosensation, we studied stiffness-driven changes to cell morphology and mechanomarker expression in H9C2 cells and neonatal rat cardiomyocytes (NRCMs). Linear stiffness gradient polyacrylamide hydrogels (2-33 kPa) coated with ECM proteins including Collagen I (Col), Fibronectin (Fn) or Laminin (Ln) were used to represent necrotic, healthy, and infarcted cardiac tissue on a continuous stiffness gradient. Cell size, cell shape and nuclear size were found to be mechanosensitive in H9C2 cells, as was the expression or nuclear translocalization of the mechanomarkers Lamin-A, YAP, and MRTF-A. Minor differences were observed between the different ECM coatings, with the same overarching stiffness-dependent trends being observed across Col, Fn and Ln coated hydrogels. Inhibition of mechanotransduction in H9C2 cells using blebbistatin or Y27632 resulted in disruptions to cell shape, nuclear shape, and nuclear size, however, trends in cell size and mechanomarker expression were not significantly attenuated. Mechanosensation in NRCMs was much less marked, with no significant changes in cell morphology being detected, although YAP did become increasingly nuclear localized with increasing stiffness. In α-actinin positive cells, striations formed with regular structure and frequency at all stiffnesses for Col and Fn coated hydrogels, but not Ln coated gels. In this study, we used our stiffness gradient hydrogels to comprehensively map the relationship between ECM stiffness and cardiac cell phenotype and found that less mature H9C2 cardiac cells are more sensitive to ECM changes than the more developed neonatal cardiomyocytes.
Collapse
Affiliation(s)
- Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| | - Livia Hool
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia. .,Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
41
|
Kamperman T, Henke S, Crispim JF, Willemen NGA, Dijkstra PJ, Lee W, Offerhaus HL, Neubauer M, Smink AM, de Vos P, de Haan BJ, Karperien M, Shin SR, Leijten J. Tethering Cells via Enzymatic Oxidative Crosslinking Enables Mechanotransduction in Non-Cell-Adhesive Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102660. [PMID: 34476848 PMCID: PMC8530967 DOI: 10.1002/adma.202102660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/10/2021] [Indexed: 05/14/2023]
Abstract
Cell-matrix interactions govern cell behavior and tissue function by facilitating transduction of biomechanical cues. Engineered tissues often incorporate these interactions by employing cell-adhesive materials. However, using constitutively active cell-adhesive materials impedes control over cell fate and elicits inflammatory responses upon implantation. Here, an alternative cell-material interaction strategy that provides mechanotransducive properties via discrete inducible on-cell crosslinking (DOCKING) of materials, including those that are inherently non-cell-adhesive, is introduced. Specifically, tyramine-functionalized materials are tethered to tyrosines that are naturally present in extracellular protein domains via enzyme-mediated oxidative crosslinking. Temporal control over the stiffness of on-cell tethered 3D microniches reveals that DOCKING uniquely enables lineage programming of stem cells by targeting adhesome-related mechanotransduction pathways acting independently of cell volume changes and spreading. In short, DOCKING represents a bioinspired and cytocompatible cell-tethering strategy that offers new routes to study and engineer cell-material interactions, thereby advancing applications ranging from drug delivery, to cell-based therapy, and cultured meat.
Collapse
Affiliation(s)
- Tom Kamperman
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
- Division of Engineering in MedicineBrigham and Women's HospitalHarvard Medical School65 Landsdowne StreetCambridgeMA02139USA
| | - Sieger Henke
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - João F. Crispim
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Niels G. A. Willemen
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Pieter J. Dijkstra
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Wooje Lee
- Optical SciencesMESA+ Institute for NanotechnologyUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Herman L. Offerhaus
- Optical SciencesMESA+ Institute for NanotechnologyUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Martin Neubauer
- Physical Chemistry IIUniversity of BayreuthUniversitätsstrasse 30D‐95447BayreuthGermany
| | - Alexandra M. Smink
- Department of Pathology and Medical BiologySection of ImmunoendocrinologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 1 (EA11)Groningen9713 GZThe Netherlands
| | - Paul de Vos
- Department of Pathology and Medical BiologySection of ImmunoendocrinologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 1 (EA11)Groningen9713 GZThe Netherlands
| | - Bart J. de Haan
- Department of Pathology and Medical BiologySection of ImmunoendocrinologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 1 (EA11)Groningen9713 GZThe Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Su Ryon Shin
- Division of Engineering in MedicineBrigham and Women's HospitalHarvard Medical School65 Landsdowne StreetCambridgeMA02139USA
| | - Jeroen Leijten
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| |
Collapse
|
42
|
Roth JG, Huang MS, Li TL, Feig VR, Jiang Y, Cui B, Greely HT, Bao Z, Paşca SP, Heilshorn SC. Advancing models of neural development with biomaterials. Nat Rev Neurosci 2021; 22:593-615. [PMID: 34376834 PMCID: PMC8612873 DOI: 10.1038/s41583-021-00496-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cells have emerged as a promising in vitro model system for studying the brain. Two-dimensional and three-dimensional cell culture paradigms have provided valuable insights into the pathogenesis of neuropsychiatric disorders, but they remain limited in their capacity to model certain features of human neural development. Specifically, current models do not efficiently incorporate extracellular matrix-derived biochemical and biophysical cues, facilitate multicellular spatio-temporal patterning, or achieve advanced functional maturation. Engineered biomaterials have the capacity to create increasingly biomimetic neural microenvironments, yet further refinement is needed before these approaches are widely implemented. This Review therefore highlights how continued progression and increased integration of engineered biomaterials may be well poised to address intractable challenges in recapitulating human neural development.
Collapse
Affiliation(s)
- Julien G Roth
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Thomas L Li
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Vivian R Feig
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Yuanwen Jiang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Henry T Greely
- Stanford Law School, Stanford University, Stanford, CA, USA
| | - Zhenan Bao
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Sergiu P Paşca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
43
|
Cacopardo L, Guazzelli N, Ahluwalia A. Characterising and engineering biomimetic materials for viscoelastic mechanotransduction studies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:912-925. [PMID: 34555953 PMCID: PMC9419958 DOI: 10.1089/ten.teb.2021.0151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The mechanical behavior of soft tissue extracellular matrix is time dependent. Moreover, it evolves over time due to physiological processes as well as aging and disease. Measuring and quantifying the time-dependent mechanical behavior of soft tissues and materials pose a challenge, not only because of their labile and hydrated nature but also because of the lack of a common definition of terms and understanding of models for characterizing viscoelasticity. Here, we review the most important measurement techniques and models used to determine the viscoelastic properties of soft hydrated materials—or hydrogels—underlining the difference between viscoelastic behavior and the properties and descriptors used to quantify viscoelasticity. We then discuss the principal factors, which determine tissue viscoelasticity in vivo and summarize what we currently know about cell response to time-dependent materials, outlining fundamental factors that have to be considered when interpreting results. Particular attention is given to the relationship between the different time scales involved (mechanical, cellular and observation time scales), as well as scaling principles, all of which must be considered when designing viscoelastic materials and performing experiments for biomechanics or mechanobiology applications. From this overview, key considerations and directions for furthering insights and applications in the emergent field of cell viscoelastic mechanotransduction are provided.
Collapse
Affiliation(s)
| | - Nicole Guazzelli
- University of Pisa, 9310, Research Center 'E.Piaggio', Pisa, Italy.,University of Pisa, 9310, Information Engineering Department, Pisa, Italy;
| | - Arti Ahluwalia
- University of Pisa, 9310, Pisa, Italy.,University of Pisa, 9310, Information Engineering Department, Pisa, Toscana, Italy.,Centro 3R (Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research), Pisa, Italy;
| |
Collapse
|
44
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
45
|
Zhao F, Cheng J, Zhang J, Yu H, Dai W, Yan W, Sun M, Ding G, Li Q, Meng Q, Liu Q, Duan X, Hu X, Ao Y. Comparison of three different acidic solutions in tendon decellularized extracellular matrix bio-ink fabrication for 3D cell printing. Acta Biomater 2021; 131:262-275. [PMID: 34157451 DOI: 10.1016/j.actbio.2021.06.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 06/05/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022]
Abstract
Decellularized extracellular matrix (dECM) hydrogels are being increasingly investigated for use in bio-inks for three-dimensional cell printing given their good cytocompatibility and biomimetic properties. The osmotic pressure and stiffness of bio-ink are important factors affecting the biological functions of printed cells. However, little attention has been given to the osmotic pressure and stiffness of the dECM bio-inks. Here, we compared three types of commonly used acidic solutions in the bio-fabrication of a tendon derived dECM bio-ink for 3D cell printing (0.5 M acetic acid, 0.1 M hydrochloric acid and 0.02 M hydrochloric acid). We found that low pH value of 0.1 M hydrochloric acid could accelerate the digestion process for dECM powders. This could lead to a much softer dECM hydrogel with storage modulus less than 100 Pa. This soft dECM hydrogel facilitated the spreading and proliferation of stem cells encapsulated within it. It also showed better tendon-inducing ability compared with two others much stiffer dECM hydrogels. However, this over-digested dECM hydrogel was more unstable as it could shrink with the culture time going on. For 0.5 M acetic acid made dECM bio-ink, the hyperosmotic state of the bio-ink led to much lower cellular viability rates. Postprocess (Dilution or dialysis) to tailor the osmotic pressure of hydrogels could be a necessary step before mixed with cells. Thus, kindly choosing the type and concentration of acidic solution is necessary for dECM bio-ink preparation. And a balance should be made between the digestion period, strength of acidic solution, as well as the size and concentration of the dECM powders. STATEMENT OF SIGNIFICANCE: The dECM bio-ink has been widely used in 3D cell printing for tissue engineering and organ modelling. In this study, we found that different types of acid have different digestion and dissolution status for the dECM materials. A much softer tendon derived dECM hydrogel with lower stiffness could facilitate the cellular spreading, proliferation and tendon differentiation. We also demonstrated that the osmotic pressure should be taken care of in the preparation of dECM bio-ink with 0.5 M acetic acid. Thus, kindly choosing the type and concentration of acidic solution is necessary for dECM bio-ink preparation.
Collapse
|
46
|
Khalil NN, McCain ML. Engineering the Cellular Microenvironment of Post-infarct Myocardium on a Chip. Front Cardiovasc Med 2021; 8:709871. [PMID: 34336962 PMCID: PMC8316619 DOI: 10.3389/fcvm.2021.709871] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/14/2021] [Indexed: 01/02/2023] Open
Abstract
Myocardial infarctions are one of the most common forms of cardiac injury and death worldwide. Infarctions cause immediate necrosis in a localized region of the myocardium, which is followed by a repair process with inflammatory, proliferative, and maturation phases. This repair process culminates in the formation of scar tissue, which often leads to heart failure in the months or years after the initial injury. In each reparative phase, the infarct microenvironment is characterized by distinct biochemical, physical, and mechanical features, such as inflammatory cytokine production, localized hypoxia, and tissue stiffening, which likely each contribute to physiological and pathological tissue remodeling by mechanisms that are incompletely understood. Traditionally, simplified two-dimensional cell culture systems or animal models have been implemented to elucidate basic pathophysiological mechanisms or predict drug responses following myocardial infarction. However, these conventional approaches offer limited spatiotemporal control over relevant features of the post-infarct cellular microenvironment. To address these gaps, Organ on a Chip models of post-infarct myocardium have recently emerged as new paradigms for dissecting the highly complex, heterogeneous, and dynamic post-infarct microenvironment. In this review, we describe recent Organ on a Chip models of post-infarct myocardium, including their limitations and future opportunities in disease modeling and drug screening.
Collapse
Affiliation(s)
- Natalie N Khalil
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
47
|
Xiang S, Li Z, Fritch MR, Li L, Velankar S, Liu Y, Sohn J, Baker N, Lin H, Tuan RS. Caveolin-1 mediates soft scaffold-enhanced adipogenesis of human mesenchymal stem cells. Stem Cell Res Ther 2021; 12:347. [PMID: 34127047 PMCID: PMC8201886 DOI: 10.1186/s13287-021-02356-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/26/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human bone marrow-derived mesenchymal stem cells (hBMSCs) can differentiate into adipocytes upon stimulation and are considered an appropriate cell source for adipose tissue engineering. In addition to biochemical cues, the stiffness of a substrate that cells attach to has also been shown to affect hBMSC differentiation potential. Of note, most current studies are conducted on monolayer cultures which do not directly inform adipose tissue engineering, where 3-dimensional (3D) scaffolds are often used to create proper tissue architecture. In this study, we aim to examine the adipogenic differentiation of hBMSCs within soft or stiff scaffolds and investigate the molecular mechanism mediating the response of hBMSCs to substrate stiffness in 3D culture, specifically the involvement of the integral membrane protein, caveolin-1 (CAV1), known to regulate signaling in MSCs via compartmentalizing and concentrating signaling molecules. METHODS By adjusting the photo-illumination time, photocrosslinkable gelatin scaffolds with the same polymer concentration but different stiffnesses were created. hBMSCs were seeded within soft and stiff scaffolds, and their response to adipogenic induction under different substrate mechanical conditions was characterized. The functional involvement of CAV1 was assessed by suppressing its expression level using CAV1-specific siRNA. RESULTS The soft and stiff scaffolds used in this study had a compressive modulus of ~0.5 kPa and ~23.5 kPa, respectively. hBMSCs showed high viability in both scaffold types, but only spread out in the soft scaffolds. hBMSCs cultured in soft scaffolds displayed significantly higher adipogenesis, as revealed by histology, qRT-PCR, and immunostaining. Interestingly, a lower CAV1 level was observed in hBMSCs in the soft scaffolds, concomitantly accompanied by increased levels of Yes-associated protein (YAP) and decreased YAP phosphorylation, when compared to cells seeded in the stiff scaffolds. Interestingly, reducing CAV1 expression with siRNA was shown to further enhance hBMSC adipogenesis, which may function through activation of the YAP signaling pathway. CONCLUSIONS Soft biomaterials support superior adipogenesis of encapsulated hBMSCs in 3D culture, which is partially mediated by the CAV1-YAP axis. Suppressing CAV1 expression levels represents a robust method in the promotion of hBMSC adipogenesis.
Collapse
Affiliation(s)
- Shiqi Xiang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhong Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Madalyn R Fritch
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - La Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sachin Velankar
- Department of Chem/Petroleum Engineering and Mechanical Engineering & Materials Science, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
| | - Yuwei Liu
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jihee Sohn
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Present Address: Biogen, Boston, Massachusetts, USA
| | - Natasha Baker
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Present Address: Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, Pennsylvania, USA
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA. .,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. .,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA.
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA. .,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. .,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA. .,Present Address: Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
48
|
Stowers RS. Advances in Extracellular Matrix-Mimetic Hydrogels to Guide Stem Cell Fate. Cells Tissues Organs 2021; 211:703-720. [PMID: 34082418 DOI: 10.1159/000514851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/11/2021] [Indexed: 01/25/2023] Open
Abstract
In the fields of regenerative medicine and tissue engineering, stem cells offer vast potential for treating or replacing diseased and damaged tissue. Much progress has been made in understanding stem cell biology, yielding protocols for directing stem cell differentiation toward the cell type of interest for a specific application. One particularly interesting and powerful signaling cue is the extracellular matrix (ECM) surrounding stem cells, a network of biopolymers that, along with cells, makes up what we define as a tissue. The composition, structure, biochemical features, and mechanical properties of the ECM are varied in different tissues and developmental stages, and serve to instruct stem cells toward a specific lineage. By understanding and recapitulating some of these ECM signaling cues through engineered ECM-mimicking hydrogels, stem cell fate can be directed in vitro. In this review, we will summarize recent advances in material systems to guide stem cell fate, highlighting innovative methods to capture ECM functionalities and how these material systems can be used to provide basic insight into stem cell biology or make progress toward therapeutic objectives.
Collapse
Affiliation(s)
- Ryan S Stowers
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California, USA
| |
Collapse
|
49
|
Hepburn MS, Foo KY, Wijesinghe P, Munro PRT, Chin L, Kennedy BF. Speckle-dependent accuracy in phase-sensitive optical coherence tomography. OPTICS EXPRESS 2021; 29:16950-16968. [PMID: 34154247 DOI: 10.1364/oe.417954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/26/2021] [Indexed: 05/25/2023]
Abstract
Phase-sensitive optical coherence tomography (OCT) is used to measure motion in a range of techniques, such as Doppler OCT and optical coherence elastography (OCE). In phase-sensitive OCT, motion is typically estimated using a model of the OCT signal derived from a single reflector. However, this approach is not representative of turbid samples, such as tissue, which exhibit speckle. In this study, for the first time, we demonstrate, through theory and experiment that speckle significantly lowers the accuracy of phase-sensitive OCT in a manner not accounted for by the OCT signal-to-noise ratio (SNR). We describe how the inaccuracy in speckle reduces phase difference sensitivity and introduce a new metric, speckle brightness, to quantify the amount of constructive interference at a given location in an OCT image. Experimental measurements show an almost three-fold degradation in sensitivity between regions of high and low speckle brightness at a constant OCT SNR. Finally, we apply these new results in compression OCE to demonstrate a ten-fold improvement in strain sensitivity, and a five-fold improvement in contrast-to-noise by incorporating independent speckle realizations. Our results show that speckle introduces a limit to the accuracy of phase-sensitive OCT and that speckle brightness should be considered to avoid erroneous interpretation of experimental data.
Collapse
|
50
|
Li J, Hepburn MS, Chin L, Mowla A, Kennedy BF. Analysis of sensitivity in quantitative micro-elastography. BIOMEDICAL OPTICS EXPRESS 2021; 12:1725-1745. [PMID: 33796383 PMCID: PMC7984799 DOI: 10.1364/boe.417829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 05/11/2023]
Abstract
Quantitative micro-elastography (QME), a variant of compression optical coherence elastography (OCE), is a technique to image tissue elasticity on the microscale. QME has been proposed for a range of applications, most notably tumor margin assessment in breast-conserving surgery. However, QME sensitivity, a key imaging metric, has yet to be systematically analyzed. Consequently, it is difficult to optimize imaging performance and to assess the potential of QME in new application areas. To address this, we present a framework for analyzing sensitivity that incorporates the three main steps in QME image formation: mechanical deformation, its detection using optical coherence tomography (OCT), and signal processing used to estimate elasticity. Firstly, we present an analytical model of QME sensitivity, validated by experimental data, and demonstrate that sub-kPa elasticity sensitivity can be achieved in QME. Using silicone phantoms, we demonstrate that sensitivity is dependent on friction, OCT focus depth, and averaging methods in signal processing. For the first time, we show that whilst lubrication of layer improves accuracy by reducing surface friction, it reduces sensitivity due to the time-dependent effect of lubricant exudation from the layer boundaries resulting in increased friction. Furthermore, we demonstrate how signal processing in QME provides a trade-off between sensitivity and resolution that can be used to optimize imaging performance. We believe that our framework to analyze sensitivity can help to sustain the development of QME and, also, that it can be readily adapted to other OCE techniques.
Collapse
Affiliation(s)
- Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
- Australian Research Council Centre for Personalized Therapeutics Technologies, Australia
| | - Matt S. Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
| | - Lixin Chin
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
| | - Alireza Mowla
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
| | - Brendan F. Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, 35, Stirling Highway, Perth, Western Australia, 6009, Australia
- Australian Research Council Centre for Personalized Therapeutics Technologies, Australia
| |
Collapse
|