1
|
Eslami H, Darvishi A. Extracellular Matrix Viscoelasticity: A Dynamic Regulator of Cellular Behavior. Ann Biomed Eng 2025:10.1007/s10439-025-03767-2. [PMID: 40526357 DOI: 10.1007/s10439-025-03767-2] [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: 12/09/2024] [Accepted: 05/26/2025] [Indexed: 06/19/2025]
Abstract
The extracellular matrix (ECM) is a three-dimensional network of polysaccharides and proteins that provides biochemical signals and structural rigidity to cells. In addition to structural support, the ECM provides dynamic mechanical properties such as viscoelasticity. Scientifically, viscoelasticity is the time-dependent response of materials under stress. Due to the viscoelastic behavior of the ECM, viscoelasticity plays a critical role in regulating and stimulating cellular behaviors such as adhesion, proliferation, differentiation, and migration, as well as tissue morphogenesis and remodeling. Scientific knowledge about viscoelasticity usually originates from materials science where physical parameters are well defined. Hence, understanding materials' physical/mechanical behaviors (especially biomaterials in biological contexts) and mathematical modeling related to viscoelasticity are important to achieve useful results. On the other hand, recent advances in the development of biophysical instruments for measuring viscoelasticity have revealed a strong link between material properties and physiological systems and have emerged as important diagnostic tools for investigating the viscoelastic behavior of cells and tissues. Therefore, this review, focusing on matrix viscoelasticity, explains the viscoelastic nature of cells/tissues, examines the impact of matrix viscoelasticity on cellular processes, reviews the role of engineered biomaterials in improving cellular behaviors, and finally discusses modern experimental techniques for measuring viscoelasticity at the cellular level.
Collapse
Affiliation(s)
- Hossein Eslami
- Department of Biomedical Engineering, Meybod University, Meybod, Iran.
| | - Ahmad Darvishi
- Department of Biomedical Engineering, Meybod University, Meybod, Iran.
| |
Collapse
|
2
|
Scutte A, Harrison K, Gregory T, Quashie D, Ramakrishnan S, Ali J. Rheological Characterization and 3D Fabrication of Artificial Bacterial Biofilms. ACS Biomater Sci Eng 2025. [PMID: 40378299 DOI: 10.1021/acsbiomaterials.5c00223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Biofilms are significantly involved in the progression of many diseases, such as cancer and upper respiratory infections, due to their ability to adhere to soft tissues. Factors influencing biofilm development have been extensively studied on planar substrates; however, there is limited understanding regarding biofilm growth and interactions within 3D matrices. Developing biofilm models that closely mimic natural bacterial communities' chemical and mechanical properties in soft tissues is essential for developing next-generation antibacterial compounds and therapeutics, as 3D biofilms are more complex and less susceptible to treatment than their 2D counterparts. Here, to understand environmental viscoelastic effects on biofilms within 3D matrix environments, two types of alginate-based hydrogels are formulated and used to encapsulatevarying concentrations of Salmonella Typhimurium. We explore the effects of increasing S. Typhimurium concentrations on hydrogel rheological properties and assess the impact of printing parameters on bacterial viability. Results show that hydrogels exhibit shear thinning behavior and that increasing the bacterial concentration up to 1 × 107 CFU mL-1 has no significant effect on the hydrogel precursor moduli and low shear viscosity. However, increasing the bacterial concentration to 1 × 1010 CFU mL-1 significantly decreases the hydrogel shear viscosity and modulus. Utilizing extrusion-based bioprinting, the optimal printing parameters (Pr > 0.8) have minimal effects on bacterial viability (>80%) over a 4 day incubation period. Additionally, we find that lower concentrations of bacteria form larger aggregates over time than hydrogels with higher cell concentrations. We show that biofilm growth in 3D depends on both initial bacterial density and matrix rigidity. Further development of physicochemically tuned bioprinted bacterial communities will aid our understanding of bacterial interactions within their 3D environments and enable the use of in vitro tissue models that incorporate biofilms for high-throughput therapeutic screening.
Collapse
Affiliation(s)
- Annie Scutte
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - Kiram Harrison
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - Tyler Gregory
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - David Quashie
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - Subramanian Ramakrishnan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - Jamel Ali
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| |
Collapse
|
3
|
Kasmi N, Pieruccioni L, Pitot E, Fourquaux I, Wodrinski A, Gibot L, Fitremann J. The potential of carbohydrate supramolecular hydrogels for long-term 3D culture of primary fibroblasts. J Mater Chem B 2025; 13:4386-4405. [PMID: 40084972 DOI: 10.1039/d4tb02658f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
N-Alkyl-galactonamides, which are small synthetic molecules derived from galactose, self-assemble to give fibrous hydrogels. These molecules are biocompatible and, in a previous study, the cell culture of human neural stem cells was performed for 7 days on a gel of N-heptyl-D-galactonamide. With the objective of broadening the scope of these molecules as scaffolds for cell culture, in the present study, the culture of primary human dermal fibroblasts has been carried out on N-nonyl-D-galactonamide hydrogels. These supramolecular fibrillar hydrogels have a sufficient mechanical strength to withstand cell culture (≈50 kPa) and they are resistant enough on the long term to carry out the cell culture over at least 3 weeks. In contrast to N-heptyl-D-galactonamide, N-nonyl-D-galactonamide is insoluble in the culture medium. It avoids its dissolution at each renewal of the culture medium. The molecule is only slowly eliminated by other mechanisms (1/3rd in 3 weeks), which did not impair the cell culture on a monthly scale. The hydrogel's microstructure and how the cells organize on this scaffold have been studied using electron and two-photon microscopies. The gel is made of a quite homogeneous network with a width of ≈180 nm and hundreds of micrometer long fibers, except at the surface where a dense mat of heterogeneous fibers is formed. We focused on methods able to colocalize the cells and the gel fibers. Many cell clusters have elongated and multidirectionnal shapes, guided by the fibers. Chains of single cells are also found following the fibers from one cluster to another. N-Nonyl-D-galactonamide fibers, which have the advantage of not being autofluorescent, do not mask the fluorescence of cells. But interestingly, they give a strong second harmonic generation (SHG) signal, due to their well-organized lamellar structure. We also made a special effort to visualize the penetration of cells within the depth of the hydrogels, in 3D, notably by sectioning the hydrogels, despite their softness. It was found that most of the cells stayed at the surface, but several cells grew within the supramolecular fiber network between 50 and 100 μm depth.
Collapse
Affiliation(s)
- Nadia Kasmi
- Laboratoire Softmat, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, Toulouse, France.
| | - Laetitia Pieruccioni
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVT, Toulouse, France
| | - Eve Pitot
- Cytometry and Imaging Core facility, Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Isabelle Fourquaux
- Centre de Microscopie Electronique Appliquée à la Biologie (CMEAB), Faculté de Médecine Rangueil, Université de Toulouse III - Paul Sabatier, Toulouse, France
| | - Alexandre Wodrinski
- Laboratoire Softmat, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, Toulouse, France.
| | - Laure Gibot
- Laboratoire Softmat, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, Toulouse, France.
| | - Juliette Fitremann
- Laboratoire Softmat, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, Toulouse, France.
| |
Collapse
|
4
|
Narasimhan BN, Fraley SI. Matrix degradation enhances stress relaxation, regulating cell adhesion and spreading. Proc Natl Acad Sci U S A 2025; 122:e2416771122. [PMID: 40131951 PMCID: PMC12002262 DOI: 10.1073/pnas.2416771122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/09/2025] [Indexed: 03/27/2025] Open
Abstract
In native extracellular matrices (ECM), cells utilize matrix metalloproteinases (MMPs) to degrade and remodel their microenvironment. Accordingly, synthetic matrices have been engineered to permit MMP-mediated cleavage, facilitating cell spreading, migration, and interactions. However, the interplay between matrix degradability and mechanical properties remains underexplored. We hypothesized that MMP activity induces immediate mechanical alterations in the ECM, which are subsequently detected by cells. We observed that both fibrillar collagen and synthetic degradable matrices exhibit enhanced stress relaxation following MMP exposure. Cells responded to these variations in relaxation by modulating their spreading and focal adhesions. Furthermore, we demonstrated that stress relaxation and cell spreading can be precisely controlled through the rational design of matrix degradability. These findings establish a fundamental link between matrix degradability and stress relaxation, with potential implications for a broad spectrum of biological applications.
Collapse
|
5
|
El Kommos A, Magesh P, Lattanze S, Perros A, Andreopoulos F, Travascio F, Jackson A. Hybrid Hydrogels Augmented via Additive Network Integration (HANI) for Meniscal Tissue Engineering Applications. Gels 2025; 11:223. [PMID: 40277659 PMCID: PMC12027216 DOI: 10.3390/gels11040223] [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: 02/17/2025] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 04/26/2025] Open
Abstract
Orthopedic soft tissue injuries, such as those to the fibrocartilaginous meniscus in the knee, present a significant clinical challenge, impacting millions globally and often requiring surgical interventions that fail to fully restore mechanical function. Current bioengineered meniscal replacement options that incorporate synthetic and/or natural scaffolds have limitations in biomechanical performance and biological integration. This study introduces a novel scaffold fabrication approach, termed Hybrid Hydrogels Augmented via Additive Network Integration (HANI) with great potential for meniscal tissue engineering applications. HANI scaffolds combine cross-linked gelatin-based hydrogels with polycaprolactone (PCL) additive networks, created via Fused Deposition Modeling (FDM), to enhance mechanical strength and replicate the anisotropic properties of the meniscus. Custom Stereolithography (SLA)-printed molds ensure precise dimensional control and seamless incorporation of PCL networks within the hydrogel matrix. The mechanical evaluation of HANI scaffolds showed improvements in compressive stiffness, stress relaxation behavior, and load-bearing capacity, especially with circumferential and 3D PCL reinforcements, when compared to hydrogel scaffolds without additive networks. These findings highlight HANI's potential as a cost-effective, scalable, and tunable scaffold fabrication approach for meniscal tissue engineering applications.
Collapse
Affiliation(s)
- Anthony El Kommos
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA; (A.E.K.); (S.L.); (A.P.)
| | - Praveen Magesh
- Department of Mechanical and Aerospace Engineering, University of Miami, Coral Gables, FL 33146, USA; (P.M.); (F.T.)
| | - Samantha Lattanze
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA; (A.E.K.); (S.L.); (A.P.)
| | - Andrew Perros
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA; (A.E.K.); (S.L.); (A.P.)
| | - Fotios Andreopoulos
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA; (A.E.K.); (S.L.); (A.P.)
- Department of Surgery, University of Miami, Miami, FL 33136, USA
| | - Francesco Travascio
- Department of Mechanical and Aerospace Engineering, University of Miami, Coral Gables, FL 33146, USA; (P.M.); (F.T.)
- Department of Orthopaedic Surgery, University of Miami, Miami, FL 33136, USA
| | - Alicia Jackson
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA; (A.E.K.); (S.L.); (A.P.)
| |
Collapse
|
6
|
Piantino M, Muller Q, Nakadozono C, Yamada A, Matsusaki M. Towards more realistic cultivated meat by rethinking bioengineering approaches. Trends Biotechnol 2025; 43:364-382. [PMID: 39271415 DOI: 10.1016/j.tibtech.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024]
Abstract
Cultivated meat (CM) refers to edible lab-grown meat that incorporates cultivated animal cells. It has the potential to address some issues associated with real meat (RM) production, including the ethical and environmental impact of animal farming, and health concerns. Recently, various biomanufacturing methods have been developed to attempt to recreate realistic meat in the laboratory. We therefore overview recent achievements and challenges in the production of CM. We also discuss the issues that need to be addressed and suggest additional recommendations and potential criteria to help to bridge the gap between CM and RM from an engineering standpoint.
Collapse
Affiliation(s)
- Marie Piantino
- Consortium for Future Innovation by Cultured Meat, Osaka, Japan
| | - Quentin Muller
- Consortium for Future Innovation by Cultured Meat, Osaka, Japan
| | - Chika Nakadozono
- Consortium for Future Innovation by Cultured Meat, Osaka, Japan; Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan; Shimadzu Analytical Innovation Research Laboratories, Osaka University, Osaka, Japan; Shimadzu Corporation, Kyoto, Japan
| | - Asuka Yamada
- Consortium for Future Innovation by Cultured Meat, Osaka, Japan; Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan; Toppan Holdings Inc., Business Development Division, Technical Research Institute, Saitama, Japan
| | - Michiya Matsusaki
- Consortium for Future Innovation by Cultured Meat, Osaka, Japan; Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan.
| |
Collapse
|
7
|
Gionet-Gonzales M, Gathman G, Rosas J, Kunisaki KY, Inocencio DGP, Hakami N, Milburn GN, Pitenis AA, Campbell KS, Pruitt BL, Stowers RS. Stress relaxation rates of myocardium from failing and non-failing hearts. Biomech Model Mechanobiol 2025; 24:265-280. [PMID: 39741200 PMCID: PMC11846740 DOI: 10.1007/s10237-024-01909-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/06/2024] [Indexed: 01/02/2025]
Abstract
The heart is a dynamic pump whose function is influenced by its mechanical properties. The viscoelastic properties of the heart, i.e., its ability to exhibit both elastic and viscous characteristics upon deformation, influence cardiac function. Viscoelastic properties change during heart failure (HF), but direct measurements of failing and non-failing myocardial tissue stress relaxation under constant displacement are lacking. Further, how consequences of tissue remodeling, such as fibrosis and fat accumulation, alter the stress relaxation remains unknown. To address this gap, we conducted stress relaxation tests on porcine myocardial tissue to establish baseline properties of cardiac tissue. We found porcine myocardial tissue to be fast relaxing, characterized by stress relaxation tests on both a rheometer and microindenter. We then measured human left ventricle (LV) epicardium and endocardium tissue from non-failing, ischemic HF and non-ischemic HF patients by microindentation. Analyzing by patient groups, we found that ischemic HF samples had slower stress relaxation than non-failing endocardium. Categorizing the data by stress relaxation times, we found that slower stress relaxing tissues were correlated with increased collagen deposition and increased α-smooth muscle actin (α-SMA) stress fibers, a marker of fibrosis and cardiac fibroblast activation, respectively. In the epicardium, analyzing by patient groups, we found that ischemic HF had faster stress relaxation than non-ischemic HF and non-failing. When categorizing by stress relaxation times, we found that faster stress relaxation correlated with Oil Red O staining, a marker for adipose tissue. These data show that changes in stress relaxation vary across the different layers of the heart during ischemic versus non-ischemic HF. These findings reveal how the viscoelasticity of the heart changes, which will lead to better modeling of cardiac mechanics for in vitro and in silico HF models.
Collapse
Affiliation(s)
- Marissa Gionet-Gonzales
- Bioengineering, University of California, Santa Barbara, Santa Barbara, United States
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States
| | - Gianna Gathman
- Bioengineering, University of California, Santa Barbara, Santa Barbara, United States
| | - Jonah Rosas
- Materials, University of California, Santa Barbara, Santa Barbara, United States
| | - Kyle Y Kunisaki
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States
| | | | - Niki Hakami
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States
| | | | - Angela A Pitenis
- Materials, University of California, Santa Barbara, Santa Barbara, United States
| | | | - Beth L Pruitt
- Bioengineering, University of California, Santa Barbara, Santa Barbara, United States.
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States.
| | - Ryan S Stowers
- Bioengineering, University of California, Santa Barbara, Santa Barbara, United States.
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States.
| |
Collapse
|
8
|
Rossi A, Bassi G, Cunha C, Baldisserri C, Ravaglia N, Gardini D, Molinari F, Lista F, Teran FJ, Piperno A, Montesi M, Panseri S. Magnetically induced anisotropic structure in an injectable hydrogel for skeletal muscle regeneration. J Colloid Interface Sci 2025; 678:334-345. [PMID: 39298986 DOI: 10.1016/j.jcis.2024.09.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Skeletal muscle integrity and its intrinsic aligned architecture are crucial for locomotion, postural support, and respiration functions, impacting overall quality of life. However, volumetric muscle loss (VML) can exceed intrinsic regenerative potential, leading to fibrosis and impairments. Autologous muscle grafting, the current gold standard, is constrained by tissue availability and success rates. Therefore, innovative strategies like cell-based therapies and scaffold-based approaches are needed. Our minimally invasive approach involves a tunable injectable hydrogel capable of achieving an aligned architecture post-injection via a low-intensity static magnetic field (SMF). Our hydrogel formulation uses gellan gum as the backbone polymer, enriched with essential extracellular matrix components such as hyaluronic acid and collagen type I, enhancing bio-functionality. To achieve an aligned architectural biomimicry, collagen type I is coupled with iron oxide magnetic nanoparticles, creating magnetic collagen bundles (MagC) that align within the hydrogel when exposed to a SMF. An extensive study was performed to characterize MagC and assess the hydrogel's stability, mechanical properties, and biological response in vitro and in vivo. The proposed system, fully composed of natural polymers, exhibited mechanical properties similar to human skeletal muscle and demonstrated effective biological performances, supporting its potential as a safe and patient-friendly treatment for VML.
Collapse
Affiliation(s)
- Arianna Rossi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy.
| | - Giada Bassi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; University of G. D'Annunzio, Department of Neurosciences, Imaging and Clinical Sciences, Via Luigi Polacchi, 11, 66100 Chieti, Italy
| | - Carla Cunha
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Carlo Baldisserri
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy
| | - Noemi Ravaglia
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy
| | - Davide Gardini
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy
| | - Filippo Molinari
- Defense Institute for Biomedical Sciences, IGESAN, Via di Santo Stefano Rotondo 4, 00184 Rome, Italy
| | - Florigio Lista
- Defense Institute for Biomedical Sciences, IGESAN, Via di Santo Stefano Rotondo 4, 00184 Rome, Italy
| | - Francisco J Teran
- iMdea Nanociencia, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain; Nanotech Solutions, Ctra Madrid 23, 40150 Villacastín, Spain
| | - Anna Piperno
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy
| | - Monica Montesi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy
| | - Silvia Panseri
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy.
| |
Collapse
|
9
|
Chang TL, Borelli AN, Cutler AA, Olwin BB, Anseth KS. Myofibers cultured in viscoelastic hydrogels reveal the effects of integrin-binding and mechanosensing on muscle satellite cells. Acta Biomater 2025; 192:48-60. [PMID: 39615561 PMCID: PMC11949280 DOI: 10.1016/j.actbio.2024.11.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024]
Abstract
Quiescent skeletal muscle satellite cells (SCs) located on myofibers activate in response to muscle injury to regenerate muscle; however, identifying the role of specific matrix signals on SC behavior in vivo is difficult. Therefore, we developed a viscoelastic hydrogel with tunable properties to encapsulate myofibers while maintaining stem cell niche polarity and SC-myofiber interactions to investigate how matrix signals, including viscoelasticity and the integrin-binding ligand arginyl-glycyl-aspartic acid (RGD), influence SC behavior during muscle regeneration. Viscoelastic hydrogels support myofiber culture while preserving SC stemness for up to 72 hours post-encapsulation, minimizing myofiber hypercontraction and SC hyperproliferation compared to Matrigel. Pax7 is continuously expressed in SCs on myofibers embedded in hydrogels with higher stress relaxation while SCs differentiate when embedded in elastic hydrogels. Increasing RGD concentrations activates SCs and translocates YAP/TAZ to the nucleus as revealed by photo-expansion microscopy. Deleting YAP/TAZ abrogates RGD-mediated activation of SCs, and thus, YAP/TAZ mediates RGD ligand-induced SC activation and subsequent proliferation. STATEMENT OF SIGNIFICANCE: Satellite cells (SCs) are responsible for muscle maintenance and regeneration, but how the extracellular matrix regulates SC function is less understood and would benefit from new biomaterial models that can recapitulate the complexity of SC niche in vitro. Upon isolation of myofibers, SCs exit quiescence, becoming activated. To circumvent this issue, we developed a viscoelastic hydrogel for encapsulating myofibers, which maintains SC quiescence and limits differentiation, allowing the study of RGD effects. We showed that increasing RGD concentration promotes activation and suppresses differentiation. Finally, to allow high resolution imaging for resolving the subcellular localization of YAP/TAZ transcriptional co-activators, we applied photo-expansion microscopy and gel-to-gel transfer techniques to quantify YAP/TAZ nuclear-cytoplasmic ratio, revealing that RGD-mediated activation relies on YAP/TAZ nuclear translocation.
Collapse
Affiliation(s)
- Tze-Ling Chang
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder CO, 80303, USA; The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Alexandra N Borelli
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder CO, 80303, USA; The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Alicia A Cutler
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Bradley B Olwin
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder CO, 80303, USA; The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA.
| |
Collapse
|
10
|
Amitrano A, Yuan Q, Agarwal B, Sen A, Dance YW, Zuo Y, Phillip JM, Gu L, Konstantopoulos K. Extracellular fluid viscosity regulates human mesenchymal stem cell lineage and function. SCIENCE ADVANCES 2025; 11:eadr5023. [PMID: 39742493 PMCID: PMC11691697 DOI: 10.1126/sciadv.adr5023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025]
Abstract
Human mesenchymal stem cells (hMSCs) respond to mechanical stimuli, including stiffness and viscoelasticity. To date, it is unknown how extracellular fluid viscosity affects hMSC function on substrates of different stiffness and viscoelasticity. While hMSCs assume an adipogenic phenotype on gels of low stiffness and prescribed stress relaxation times, elevated fluid viscosity is sufficient to bias hMSCs toward an osteogenic phenotype. Elevated viscosity induces Arp2/3-dependent actin remodeling, enhances NHE1 activity, and promotes hMSC spreading via up-regulation of integrin-linked kinase. The resulting increase in membrane tension triggers the activation of transient receptor potential cation vanilloid 4 to facilitate calcium influx, thereby stimulating RhoA/ROCK and driving YAP-dependent RUNX2 translocation to the nucleus, leading to osteogenic differentiation. hMSCs on soft gels at elevated relative to basal viscosity favor an M2 macrophage phenotype. This study establishes fluid viscosity as a key physical cue that imprints osteogenic memory in hMSCs and promotes an immunosuppressive phenotype.
Collapse
Affiliation(s)
- Alice Amitrano
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Qinling Yuan
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bhawana Agarwal
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anindya Sen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yoseph W. Dance
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yi Zuo
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Deparment of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jude M. Phillip
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | - Luo Gu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Deparment of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| |
Collapse
|
11
|
Moheimani H, Stealey S, Neal S, Ferchichi E, Zhang J, Foston M, Setton LA, Genin G, Huebsch N, Zustiak SP. Tunable Viscoelasticity of Alginate Hydrogels via Serial Autoclaving. Adv Healthc Mater 2024; 13:e2401550. [PMID: 39075933 PMCID: PMC11671294 DOI: 10.1002/adhm.202401550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/02/2024] [Indexed: 07/31/2024]
Abstract
Alginate hydrogels are widely used as biomaterials for cell culture and tissue engineering due to their biocompatibility and tunable mechanical properties. Reducing alginate molecular weight is an effective strategy for modulating hydrogel viscoelasticity and stress relaxation behavior, which can significantly impact cell spreading and fate. However, current methods like gamma irradiation to produce low molecular weight alginates suffer from high cost and limited accessibility. Here, a facile and cost-effective approach to reduce alginate molecular weight in a highly controlled manner using serial autoclaving is presented. Increasing the number of autoclave cycles results in proportional reductions in intrinsic viscosity, hydrodynamic radius, and molecular weight of the polymer while maintaining its chemical composition. Hydrogels fabricated from mixtures of the autoclaved alginates exhibit tunable mechanical properties, with inclusion of lower molecular weight alginate leading to softer gels with faster stress relaxation behaviors. The method is demonstrated by establishing how viscoelastic relaxation affects the spreading of encapsulated fibroblasts and glioblastoma cells. Results establish repetitive autoclaving as an easily accessible technique to generate alginates with a range of molecular weights and to control the viscoelastic properties of alginate hydrogels, and demonstrate utility across applications in mechanobiology, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Hamidreza Moheimani
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, MO, 63130
| | - Samuel Stealey
- Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, Saint Louis, MO, 63103
| | - Sydney Neal
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130
| | - Eya Ferchichi
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, MO, 63130
| | - Jialang Zhang
- Department of Energy, Environmental & Chemical Engineering, Washington University in Saint Louis, Saint Louis, MO, 63130
| | - Marcus Foston
- Department of Energy, Environmental & Chemical Engineering, Washington University in Saint Louis, Saint Louis, MO, 63130
| | - Lori A. Setton
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130
| | - Guy Genin
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, MO, 63130
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130
| | - Nathaniel Huebsch
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, MO, 63130
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130
| | - Silviya P. Zustiak
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, MO, 63130
- Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, Saint Louis, MO, 63103
| |
Collapse
|
12
|
Li G, Gao F, Yang D, Lin L, Yu W, Tang J, Yang R, Jin M, Gu Y, Wang P, Lu E. ECM-mimicking composite hydrogel for accelerated vascularized bone regeneration. Bioact Mater 2024; 42:241-256. [PMID: 39285909 PMCID: PMC11404060 DOI: 10.1016/j.bioactmat.2024.08.035] [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: 05/15/2024] [Revised: 08/07/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Bioactive hydrogel materials have great potential for applications in bone tissue engineering. However, fabrication of functional hydrogels that mimic the natural bone extracellular matrix (ECM) remains a challenge, because they need to provide mechanical support and embody physiological cues for angiogenesis and osteogenesis. Inspired by the features of ECM, we constructed a dual-component composite hydrogel comprising interpenetrating polymer networks of gelatin methacryloyl (GelMA) and deoxyribonucleic acid (DNA). Within the composite hydrogel, the GelMA network serves as the backbone for mechanical and biological stability, whereas the DNA network realizes dynamic capabilities (e.g., stress relaxation), thereby promoting cell proliferation and osteogenic differentiation. Furthermore, functional aptamers (Apt19S and AptV) are readily attached to the DNA network to recruit bone marrow mesenchymal stem cells (BMSCs) and achieve sustained release of loaded vascular endothelial growth factor towards angiogenesis. Our results showed that the composite hydrogel could facilitate the adhesion of BMSCs, promote osteogenic differentiation by activating focal adhesion kinase (FAK)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/β-Catenin signaling pathway, and eventually enhance vascularized bone regeneration. This study shows that the multifunctional composite hydrogel of GelMA and DNA can successfully simulate the biological functions of natural bone ECM and has great potential for repairing bone defects.
Collapse
Affiliation(s)
- Guanglong Li
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Fei Gao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Donglei Yang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Lu Lin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Weijun Yu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Jiaqi Tang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Ruhan Yang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Min Jin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Yuting Gu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Pengfei Wang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| |
Collapse
|
13
|
Liu Z, Ling SD, Liang K, Chen Y, Niu Y, Sun L, Li J, Du Y. Viscoelasticity of ECM and cells-origin, measurement and correlation. MECHANOBIOLOGY IN MEDICINE 2024; 2:100082. [PMID: 40395221 PMCID: PMC12082326 DOI: 10.1016/j.mbm.2024.100082] [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: 03/09/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 05/22/2025]
Abstract
The extracellular matrix (ECM) and cells are crucial components of natural tissue microenvironments, and they both demonstrate dynamic mechanical properties, particularly viscoelastic behaviors, when exposed to external stress or strain over time. The capacity to modify the mechanical properties of cells and ECM is crucial for gaining insight into the development, physiology, and pathophysiology of living organisms. As an illustration, researchers have developed hydrogels with diverse compositions to mimic the properties of the native ECM and use them as substrates for cell culture. The behavior of cultured cells can be regulated by modifying the viscoelasticity of hydrogels. Moreover, there is widespread interest across disciplines in accurately measuring the mechanical properties of cells and the surrounding ECM, as well as exploring the interactive relationship between these components. Nevertheless, the lack of standardized experimental methods, conditions, and other variables has hindered systematic comparisons and summaries of research findings on ECM and cell viscoelasticity. In this review, we delve into the origins of ECM and cell viscoelasticity, examine recently developed methods for measuring ECM and cell viscoelasticity, and summarize the potential interactions between cell and ECM viscoelasticity. Recent research has shown that both ECM and cell viscoelasticity experience alterations during in vivo pathogenesis, indicating the potential use of tailored viscoelastic ECM and cells in regenerative medicine.
Collapse
Affiliation(s)
- Zhiqiang Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Si Da Ling
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yihan Chen
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yudi Niu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Lei Sun
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Junyang Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
14
|
Huerta-López C, Clemente-Manteca A, Velázquez-Carreras D, Espinosa FM, Sanchez JG, Martínez-del-Pozo Á, García-García M, Martín-Colomo S, Rodríguez-Blanco A, Esteban-González R, Martín-Zamora FM, Gutierrez-Rus LI, Garcia R, Roca-Cusachs P, Elosegui-Artola A, del Pozo MA, Herrero-Galán E, Sáez P, Plaza GR, Alegre-Cebollada J. Cell response to extracellular matrix viscous energy dissipation outweighs high-rigidity sensing. SCIENCE ADVANCES 2024; 10:eadf9758. [PMID: 39546608 PMCID: PMC11567001 DOI: 10.1126/sciadv.adf9758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024]
Abstract
The mechanics of the extracellular matrix (ECM) determine cell activity and fate through mechanoresponsive proteins including Yes-associated protein 1 (YAP). Rigidity and viscous relaxation have emerged as the main mechanical properties of the ECM steering cell behavior. However, how cells integrate coexisting ECM rigidity and viscosity cues remains poorly understood, particularly in the high-stiffness regime. Here, we have exploited engineered stiff viscoelastic protein hydrogels to show that, contrary to current models of cell-ECM interaction, substrate viscous energy dissipation attenuates mechanosensing even when cells are exposed to higher effective rigidity. This unexpected behavior is however readily captured by a pull-and-hold model of molecular clutch-based cell mechanosensing, which also recapitulates opposite cellular response at low rigidities. Consistent with predictions of the pull-and-hold model, we find that myosin inhibition can boost mechanosensing on cells cultured on dissipative matrices. Together, our work provides general mechanistic understanding on how cells respond to the viscoelastic properties of the ECM.
Collapse
Affiliation(s)
- Carla Huerta-López
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | | | | | | | - Juan G. Sanchez
- Instituto de Ciencia de Materiales de Madrid, CSIC, 28049 Madrid, Spain
| | - Álvaro Martínez-del-Pozo
- Departamento de Bioquímica y Biología Molecular, Facultad de CC. Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María García-García
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Sara Martín-Colomo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | | | | | | | | | - Ricardo Garcia
- Instituto de Ciencia de Materiales de Madrid, CSIC, 28049 Madrid, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Alberto Elosegui-Artola
- Cell and Tissue Mechanobiology Laboratory, Francis Crick Institute, London, 1 Midland Road, NW1 1AT, UK
- Department of Physics, King’s College London, London, WC2R 2LS, UK
| | - Miguel A. del Pozo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Elías Herrero-Galán
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Pablo Sáez
- Laboratori de Càlcul Numèric (LaCàN), Universitat Politècnica de Catalunya–BarcelonaTech, Barcelona, Spain
- Institut de Matemàtiques de la UPC-BarcelonaTech (IMTech), Barcelona, Spain
| | - Gustavo R. Plaza
- ETSI de Caminos and Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | | |
Collapse
|
15
|
Wheeler EE, Leach JK. Tissue-Engineered Three-Dimensional Platforms for Disease Modeling and Therapeutic Development. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39345164 DOI: 10.1089/ten.teb.2024.0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Three-dimensional (3D) tissue-engineered models are under investigation to recapitulate tissue architecture and functionality, thereby overcoming limitations of traditional two-dimensional cultures and preclinical animal models. This review highlights recent developments in 3D platforms designed to model diseases in vitro that affect numerous tissues and organs, including cardiovascular, gastrointestinal, bone marrow, neural, reproductive, and pulmonary systems. We discuss current technologies for engineered tissue models, highlighting the advantages, limitations, and important considerations for modeling tissues and diseases. Lastly, we discuss future advancements necessary to enhance the reliability of 3D models of tissue development and disease.
Collapse
Affiliation(s)
- Erika E Wheeler
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California, Davis, California, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California, Davis, California, USA
| |
Collapse
|
16
|
Rossi A, Furlani F, Bassi G, Cunha C, Lunghi A, Molinari F, Teran FJ, Lista F, Bianchi M, Piperno A, Montesi M, Panseri S. Contactless magnetically responsive injectable hydrogel for aligned tissue regeneration. Mater Today Bio 2024; 27:101110. [PMID: 39211510 PMCID: PMC11360152 DOI: 10.1016/j.mtbio.2024.101110] [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: 04/14/2024] [Revised: 05/20/2024] [Accepted: 05/31/2024] [Indexed: 09/04/2024] Open
Abstract
Cellular alignment plays a pivotal role in several human tissues, including skeletal muscle, spinal cord and tendon. Various techniques have been developed to control cellular alignment using 3D biomaterials. However, the majority of 3D-aligned scaffolds require invasive surgery for implantation. In contrast, injectable hydrogels provide a non-invasive delivery method, gaining considerable attention for the treatment of diverse conditions, including osteochondral lesions, volumetric muscle loss, and traumatic brain injury. We engineered a biomimetic hydrogel with magnetic responsiveness by combining gellan gum, hyaluronic acid, collagen, and magnetic nanoparticles (MNPs). Collagen type I was paired with MNPs to form magnetic collagen bundles (MCollB), allowing the orientation control of these bundles within the hydrogel matrix through the application of a remote low-intensity magnetic field. This resulted in the creation of an anisotropic architecture. The hydrogel mechanical properties were comparable to those of human soft tissues, such as skeletal muscle, and proof of the aligned hydrogel concept was demonstrated. In vitro findings confirmed the absence of toxicity and pro-inflammatory effects. Notably, an increased fibroblast cell proliferation and pro-regenerative activation of macrophages were observed. The in-vivo study further validated the hydrogel biocompatibility and demonstrated the feasibility of injection with rapid in situ gelation. Consequently, this magnetically controlled injectable hydrogel exhibits significant promise as a minimally invasive, rapid gelling and effective treatment for regenerating various aligned human tissues.
Collapse
Affiliation(s)
- Arianna Rossi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy. Via Granarolo 64, 48018. Faenza, Italy
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences. Viale Ferdinando Stagno d'Alcontres, 31, 98166, Messina, Italy
| | - Franco Furlani
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy. Via Granarolo 64, 48018. Faenza, Italy
| | - Giada Bassi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy. Via Granarolo 64, 48018. Faenza, Italy
- University of G. D'Annunzio, Department of Neurosciences, Imaging and Clinical Sciences. Via Luigi Polacchi, 11, 66100 Chieti, Italy
| | - Carla Cunha
- i3S - Instituto de Investigação e Inovação em Saúde. Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Alice Lunghi
- Center for Translational Neurophysiology of Speech and Communication, Istituto Italiano di Tecnologia 44121 Ferrara, Italy
- Section of Physiology, Università di Ferrara 44121 Ferrara, Italy
| | - Filippo Molinari
- Defense Institute for Biomedical Sciences, IGESAN, Via di Santo Stefano Rotondo 4, 00184 Rome, Italy
| | - Francisco J. Teran
- iMdea Nanociencia, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain
- Nanotech Solutions, Ctra Madrid23, 40150 Villacastín, Spain
| | - Florigio Lista
- Defense Institute for Biomedical Sciences, IGESAN, Via di Santo Stefano Rotondo 4, 00184 Rome, Italy
| | - Michele Bianchi
- Department of Life Sciences, Università degli Studi di Modena e Reggio Emilia 44125 Modena, Italy
| | - Anna Piperno
- University of Messina, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences. Viale Ferdinando Stagno d'Alcontres, 31, 98166, Messina, Italy
| | - Monica Montesi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy. Via Granarolo 64, 48018. Faenza, Italy
| | - Silvia Panseri
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy. Via Granarolo 64, 48018. Faenza, Italy
| |
Collapse
|
17
|
Narasimhan BN, Fraley SI. Degradability tunes ECM stress relaxation and cellular mechanics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605514. [PMID: 39131364 PMCID: PMC11312499 DOI: 10.1101/2024.07.28.605514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
In native extracellular matrices (ECM), cells can use matrix metalloproteinases (MMPs) to degrade and remodel their surroundings. Likewise, synthetic matrices have been engineered to facilitate MMP-mediated cleavage that enables cell spreading, migration, and interactions. However, the intersection of matrix degradability and mechanical properties has not been fully considered. We hypothesized that immediate mechanical changes result from the action of MMPs on the ECM and that these changes are sensed by cells. Using atomic force microscopy (AFM) to measure cell-scale mechanical properties, we find that both fibrillar collagen and synthetic degradable matrices exhibit enhanced stress relaxation after MMP exposure. Cells respond to these relaxation differences by altering their spreading and focal adhesions. We demonstrate that stress relaxation can be tuned through the rational design of matrix degradability. These findings establish a fundamental link between matrix degradability and stress relaxation, which may impact a range of biological applications.
Collapse
Affiliation(s)
| | - Stephanie I. Fraley
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
18
|
Simpson A, Krissanaprasit A, Chester D, Koehler C, LaBean TH, Brown AC. Utilizing multiscale engineered biomaterials to examine TGF-β-mediated myofibroblastic differentiation. Wound Repair Regen 2024; 32:234-245. [PMID: 38459905 PMCID: PMC11111354 DOI: 10.1111/wrr.13168] [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: 08/30/2023] [Revised: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 03/11/2024]
Abstract
Cells integrate many mechanical and chemical cues to drive cell signalling responses. Because of the complex nature and interdependency of alterations in extracellular matrix (ECM) composition, ligand density, mechanics, and cellular responses it is difficult to tease out individual and combinatorial contributions of these various factors in driving cell behavior in homeostasis and disease. Tuning of material viscous and elastic properties, and ligand densities, in combinatorial fashions would enhance our understanding of how cells process complex signals. For example, it is known that increased ECM mechanics and transforming growth factor beta (TGF-β) receptor (TGF-β-R) spacing/clustering independently drive TGF-β signalling and associated myofibroblastic differentiation. However, it remains unknown how these inputs orthogonally contribute to cellular outcomes. Here, we describe the development of a novel material platform that combines microgel thin films with controllable viscoelastic properties and DNA origami to probe how viscoelastic properties and nanoscale spacing of TGF-β-Rs contribute to TGF-β signalling and myofibroblastic differentiation. We found that highly viscous materials with non-fixed TGF-β-R spacing promoted increased TGF-β signalling and myofibroblastic differentiation. This is likely due to the ability of cells to better cluster receptors on these surfaces. These results provide insight into the contribution of substrate properties and receptor localisation on downstream signalling. Future studies allow for exploration into other receptor-mediated processes.
Collapse
Affiliation(s)
- Aryssa Simpson
- Joint Department of Biomedical Engineering of University of North Carolina - Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Abhichart Krissanaprasit
- Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Daniel Chester
- Joint Department of Biomedical Engineering of University of North Carolina - Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Cynthia Koehler
- Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Thomas H LaBean
- Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Ashley C Brown
- Joint Department of Biomedical Engineering of University of North Carolina - Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
19
|
Zhang J, Li X, Tian Y, Zou J, Gan D, Deng D, Jiao C, Yin Y, Tian B, Wu R, Chen F, He X. Harnessing Mechanical Stress with Viscoelastic Biomaterials for Periodontal Ligament Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309562. [PMID: 38460171 PMCID: PMC11095218 DOI: 10.1002/advs.202309562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/15/2024] [Indexed: 03/11/2024]
Abstract
The viscoelasticity of mechanically sensitive tissues such as periodontal ligaments (PDLs) is key in maintaining mechanical homeostasis. Unfortunately, PDLs easily lose viscoelasticity (e.g., stress relaxation) during periodontitis or dental trauma, which disrupt cell-extracellular matrix (ECM) interactions and accelerates tissue damage. Here, Pluronic F127 diacrylate (F127DA) hydrogels with PDL-matched stress relaxation rates and high elastic moduli are developed. The hydrogel viscoelasticity is modulated without chemical cross-linking by controlling precursor concentrations. Under cytomechanical loading, F127DA hydrogels with fast relaxation rates significantly improved the fibrogenic differentiation potential of PDL stem cells (PDLSCs), while cells cultured on F127DA hydrogels with various stress relaxation rates exhibited similar fibrogenic differentiation potentials with limited cell spreading and traction forces under static conditions. Mechanically, faster-relaxing F127DA hydrogels leveraged cytomechanical loading to activate PDLSC mechanotransduction by upregulating integrin-focal adhesion kinase pathway and thus cytoskeletal rearrangement, reinforcing cell-ECM interactions. In vivo experiments confirm that faster-relaxing F127DA hydrogels significantly promoted PDL repair and reduced abnormal healing (e.g., root resorption and ankyloses) in delayed replantation of avulsed teeth. This study firstly investigated how matrix nonlinear viscoelasticity influences the fibrogenesis of PDLSCs under mechanical stimuli, and it reveals the underlying mechanobiology, which suggests novel strategies for PDL regeneration.
Collapse
Affiliation(s)
- Jiu‐Jiu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Xuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Yi Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Jie‐Kang Zou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Dian Gan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Dao‐Kun Deng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Chen Jiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Yuan Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Bei‐Min Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Rui‐Xin Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Fa‐Ming Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| | - Xiao‐Tao He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical UniversityXi'an710032China
| |
Collapse
|
20
|
Nguyen J, Wang L, Lei W, Hu Y, Gulati N, Chavez-Madero C, Ahn H, Ginsberg HJ, Krawetz R, Brandt M, Betz T, Gilbert PM. Culture substrate stiffness impacts human myoblast contractility-dependent proliferation and nuclear envelope wrinkling. J Cell Sci 2024; 137:jcs261666. [PMID: 38345101 PMCID: PMC11033523 DOI: 10.1242/jcs.261666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/04/2024] [Indexed: 03/28/2024] Open
Abstract
Understanding how biophysical and biochemical microenvironmental cues together influence the regenerative activities of muscle stem cells and their progeny is crucial in strategizing remedies for pathological dysregulation of these cues in aging and disease. In this study, we investigated the cell-level influences of extracellular matrix (ECM) ligands and culture substrate stiffness on primary human myoblast contractility and proliferation within 16 h of plating and found that tethered fibronectin led to stronger stiffness-dependent responses compared to laminin and collagen. A proteome-wide analysis further uncovered cell metabolism, cytoskeletal and nuclear component regulation distinctions between cells cultured on soft and stiff substrates. Interestingly, we found that softer substrates increased the incidence of myoblasts with a wrinkled nucleus, and that the extent of wrinkling could predict Ki67 (also known as MKI67) expression. Nuclear wrinkling and Ki67 expression could be controlled by pharmacological manipulation of cellular contractility, offering a potential cellular mechanism. These results provide new insights into the regulation of human myoblast stiffness-dependent contractility response by ECM ligands and highlight a link between myoblast contractility and proliferation.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Lu Wang
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Wen Lei
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Yechen Hu
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| | - Nitya Gulati
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Carolina Chavez-Madero
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Henry Ahn
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Li Ka Shing Knowledge Institute, Saint Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Howard J. Ginsberg
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Li Ka Shing Knowledge Institute, Saint Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Roman Krawetz
- McCaig Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Matthias Brandt
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University Münster, 48149 Münster, Germany
| | - Timo Betz
- Third Institute of Physics – Biophysics, Georg August University Göttingen, 37077 Göttingen, Germany
| | - Penney M. Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| |
Collapse
|
21
|
Nguyen J, Gilbert PM. Decoding the forces that shape muscle stem cell function. Curr Top Dev Biol 2024; 158:279-306. [PMID: 38670710 DOI: 10.1016/bs.ctdb.2024.02.009] [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: 04/28/2024]
Abstract
Skeletal muscle is a force-producing organ composed of muscle tissues, connective tissues, blood vessels, and nerves, all working in synergy to enable movement and provide support to the body. While robust biomechanical descriptions of skeletal muscle force production at the body or tissue level exist, little is known about force application on microstructures within the muscles, such as cells. Among various cell types, skeletal muscle stem cells reside in the muscle tissue environment and play a crucial role in driving the self-repair process when muscle damage occurs. Early evidence indicates that the fate and function of skeletal muscle stem cells are controlled by both biophysical and biochemical factors in their microenvironments, but much remains to accomplish in quantitatively describing the biophysical muscle stem cell microenvironment. This book chapter aims to review current knowledge on the influence of biophysical stresses and landscape properties on muscle stem cells in heath, aging, and diseases.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
22
|
Shi N, Wang J, Tang S, Zhang H, Wei Z, Li A, Ma Y, Xu F. Matrix Nonlinear Viscoelasticity Regulates Skeletal Myogenesis through MRTF Nuclear Localization and Nuclear Mechanotransduction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305218. [PMID: 37847903 DOI: 10.1002/smll.202305218] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/30/2023] [Indexed: 10/19/2023]
Abstract
Mechanically sensitive tissues (e.g., skeletal muscles) greatly need mechanical stimuli during the development and maturation. The extracellular matrix (ECM) mediates these signals through nonlinear viscoelasticity of collagen networks that are predominant components of the ECM. However, the interactions between cells and ECM form a feedback loop, and it has not yet been possible to determine the degree to which, if any, of the features of matrix nonlinear viscoelasticity affect skeletal muscle development and regeneration. In this study, a nonlinear viscoelastic feature (i.e., strain-enhanced stress relaxation (SESR)) in normal skeletal muscles is observed, which however is almost absent in diseased muscles from Duchenne muscular dystrophy mice. It is recapitulated such SESR feature in vitro and separated the effects of mechanical strain and ECM viscoelasticity on myoblast response by developing a collagen-based hydrogel platform. Both strain and stress relaxation induce myogenic differentiation and myotube formation by C2C12 myoblasts, and myogenesis is more promoted by applying SESR. This promotion can be explained by the effects of SESR on actin polymerization-mediated myocardin related transcription factor (MRTF) nuclear localization and nuclear mechanotransduction. This study represents the first attempt to investigate the SESR phenomenon in skeletal muscles and reveal underlying mechanobiology, which will provide new opportunities for the tissue injury treatments.
Collapse
Affiliation(s)
- Nianyuan Shi
- 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
| | - Jing Wang
- 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
| | - Shaoxin Tang
- 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
| | - Hui Zhang
- 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
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Zhao Wei
- 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
| | - 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
| | - 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
| | - 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
|
23
|
Wang H, Yang J, Tian W, Peng K, Xue Y, Zhao H, Ma X, Shi R, Chen Y. A sodium alginate/carboxymethyl chitosan dual-crosslinked injectable hydrogel scaffold with tunable softness/hardness for bone regeneration. Int J Biol Macromol 2024; 257:128700. [PMID: 38072347 DOI: 10.1016/j.ijbiomac.2023.128700] [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: 08/14/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 01/27/2024]
Abstract
Recently, injectable dual-crosslinked (DC) hydrogel scaffolds have attracted many attentions as a class of excellent bone regeneration biomaterials with in-situ tunable functions. However, the design of injectable DC hydrogels with cell behavior-compatible network structure and mechanical property remains a bottleneck. Herein, based on the in-situ gelling method, we constructed an injectable CMCS/PEG+SA/CaCl2 (CPSC) chemical/physical DC hydrogel scaffold with tunable softness/hardness mechanical properties and good biocompatibility. The formation mechanism and properties of the CPSC hydrogel scaffold were investigated by FTIR, XRD, rheometry, and mechanical testing. It is found that proper softness/hardness mechanical properties can be obtained by adjusting the secondary network structure of the hydrogel. The CPSC hydrogel scaffold prepared under optimal conditions can effectively promote cell infiltration, nutrient transport, and the osteogenic differentiation of rat bone mesenchymal stem cells (rBMSCs). The in vivo experiments show that the rBMSCs-loaded injectable CPSC hydrogels with appropriate mechanical properties can effectively promote bone reconstruction. This study has provided important guidance for the construction of injectable DC hydrogels with adjustable softness/hardness to promote osteogenesis for bone defect repair.
Collapse
Affiliation(s)
- Hui Wang
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Jueying Yang
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Wei Tian
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing 100035, China
| | - Kelin Peng
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Yun Xue
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing 100035, China
| | - Haosen Zhao
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xilan Ma
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Rui Shi
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing 100035, China.
| | - Yu Chen
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China; Sports & Medicine Integration Research Center (SMIRC), Capital University of Physical Education and Sports, Beijing 100191, China.
| |
Collapse
|
24
|
Zhang S, Liu J, Feng F, Jia Y, Xu F, Wei Z, Zhang M. Rational design of viscoelastic hydrogels for periodontal ligament remodeling and repair. Acta Biomater 2024; 174:69-90. [PMID: 38101557 DOI: 10.1016/j.actbio.2023.12.017] [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: 08/16/2023] [Revised: 11/14/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
The periodontal ligament (PDL) is a distinctive yet critical connective tissue vital for maintaining the integrity and functionality of tooth-supporting structures. However, PDL repair poses significant challenges due to the complexity of its mechanical microenvironment encompassing hard-soft-hard tissues, with the viscoelastic properties of the PDL being of particular interest. This review delves into the significant role of viscoelastic hydrogels in PDL regeneration, underscoring their utility in simulating biomimetic three-dimensional microenvironments. We review the intricate relationship between PDL and viscoelastic mechanical properties, emphasizing the role of tissue viscoelasticity in maintaining mechanical functionality. Moreover, we summarize the techniques for characterizing PDL's viscoelastic behavior. From a chemical bonding perspective, we explore various crosslinking methods and characteristics of viscoelastic hydrogels, along with engineering strategies to construct viscoelastic cell microenvironments. We present a detailed analysis of the influence of the viscoelastic microenvironment on cellular mechanobiological behavior and fate. Furthermore, we review the applications of diverse viscoelastic hydrogels in PDL repair and address current challenges in the field of viscoelastic tissue repair. Lastly, we propose future directions for the development of innovative hydrogels that will facilitate not only PDL but also systemic ligament tissue repair. STATEMENT OF SIGNIFICANCE.
Collapse
Affiliation(s)
- Songbai Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR 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, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Jingyi 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, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Fan Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Yuanbo Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Zhao Wei
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Min Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
25
|
Johnson N, Filler AC, Sethi A, Smith LR, Leach JK. Skeletal Muscle Spheroids as Building Blocks for Engineered Muscle Tissue. ACS Biomater Sci Eng 2024; 10:497-506. [PMID: 38113146 PMCID: PMC10777344 DOI: 10.1021/acsbiomaterials.3c01078] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Spheroids exhibit enhanced cell-cell interactions that facilitate improved survival and mimic the physiological cellular environment in vivo. Cell spheroids have been successfully used as building blocks for engineered tissues, yet the viability of this approach with skeletal muscle spheroids is poorly understood, particularly when incorporated into three-dimensional (3D) constructs. Bioprinting is a promising strategy to recapitulate the hierarchical organization of native tissue that is fundamental to its function. However, the influence of bioprinting on skeletal muscle cell spheroids and their function are yet to be interrogated. Using C2C12 mouse myoblasts and primary bovine muscle stem cells (MuSCs), we characterized spheroid formation as a function of duration and cell seeding density. We then investigated the potential of skeletal muscle spheroids entrapped in alginate bioink as tissue building blocks for bioprinting myogenic tissue. Both C2C12 and primary bovine MuSCs formed spheroids of similar sizes and remained viable after bioprinting. Spheroids of both cell types fused into larger tissue clusters over time within alginate and exhibited tissue formation comparable to monodisperse cells. Compared to monodisperse cells in alginate gels, C2C12 spheroids exhibited greater MyHC expression after 2 weeks, while cells within bovine MuSC spheroids displayed increased cell spreading. Both monodisperse and MuSC spheroids exhibited increased expression of genes denoting mid- and late-stage myogenic differentiation. Together, these data suggest that skeletal muscle spheroids have the potential for generating myogenic tissue via 3D bioprinting and reveal areas of research that could enhance myogenesis and myogenic differentiation in future studies.
Collapse
Affiliation(s)
- Nicholas Johnson
- Department
of Orthopaedic Surgery, UC Davis Health, Sacramento, California 95817, United States
- Department
of Biomedical Engineering, UC Davis, Davis, California 95616, United States
| | - Andrea C. Filler
- Department
of Orthopaedic Surgery, UC Davis Health, Sacramento, California 95817, United States
- Department
of Biomedical Engineering, UC Davis, Davis, California 95616, United States
| | - Akash Sethi
- Department
of Molecular and Cellular Biology, UC Davis, Davis, California 95616, United States
| | - Lucas R. Smith
- Department
of Neurobiology, Physiology and Behavior, UC Davis, Davis, California 95616, United States
| | - J. Kent Leach
- Department
of Orthopaedic Surgery, UC Davis Health, Sacramento, California 95817, United States
- Department
of Biomedical Engineering, UC Davis, Davis, California 95616, United States
| |
Collapse
|
26
|
Galindo JM, San-Millán MI, Castillo-Sarmiento CA, Ballesteros-Yáñez I, Vázquez E, Merino S, Herrero MA. Optimization of 3D Synthetic Scaffolds for Neuronal Tissue Engineering Applications. Chemistry 2024; 30:e202302481. [PMID: 37823243 DOI: 10.1002/chem.202302481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023]
Abstract
The increasing prevalence of neurodegenerative diseases has spurred researchers to develop advanced 3D models that accurately mimic neural tissues. Hydrogels stand out as ideal candidates as their properties closely resemble those of the extracellular matrix. A critical challenge in this regard is to comprehend the influence of the scaffold's mechanical properties on cell growth and differentiation, thus enabling targeted modifications. In light of this, a synthesis and comprehensive analysis of acrylamide-based hydrogels incorporating a peptide has been conducted. Adequate cell adhesion and development is achieved due to their bioactive nature and specific interactions with cellular receptors. The integration of a precisely controlled physicochemical hydrogel matrix and inclusion of the arginine-glycine-aspartic acid peptide sequence has endowed this system with an optimal structure, thus providing a unique ability to interact effectively with biomolecules. The analysis fully examined essential properties governing cell behavior, including pore size, mechanical characteristics, and swelling ability. Cell-viability experiments were performed to assess the hydrogel's biocompatibility, while the incorporation of grow factors aimed to promote the differentiation of neuroblastoma cells. The results underscore the hydrogel's ability to stimulate cell viability and differentiation in the presence of the peptide within the matrix.
Collapse
Affiliation(s)
- Josué M Galindo
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Ms Irene San-Millán
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | | | | | - Ester Vázquez
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Sonia Merino
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - M Antonia Herrero
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| |
Collapse
|
27
|
Fan F, Su B, Kolodychak A, Ekwueme E, Alderfer L, Saha S, Webber MJ, Hanjaya-Putra D. Hyaluronic Acid Hydrogels with Phototunable Supramolecular Cross-Linking for Spatially Controlled Lymphatic Tube Formation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58181-58195. [PMID: 38065571 PMCID: PMC10739586 DOI: 10.1021/acsami.3c12514] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/22/2023]
Abstract
The dynamics of the extracellular matrix (ECM) influences stem cell differentiation and morphogenesis into complex lymphatic networks. While dynamic hydrogels with stress relaxation properties have been developed, many require detailed chemical processing to tune viscoelasticity, offering a limited opportunity for in situ and spatiotemporal control. Here, a hyaluronic acid (HA) hydrogel is reported with viscoelasticity that is controlled and spatially tunable using UV light to direct the extent of supramolecular and covalent cross-linking interactions. This is achieved using UV-mediated photodimerization of a supramolecular ternary complex of pendant trans-Brooker's Merocyanine (BM) guests and a cucurbit[8]uril (CB[8]) macrocycle. The UV-mediated conversion of this supramolecular complex to its covalent photodimerized form is catalyzed by CB[8], offering a user-directed route to spatially control hydrogel dynamics in combination with orthogonal photopatterning by UV irradiation through photomasks. This material thus achieves spatial heterogeneity of substrate dynamics, recreating features of native ECM without the need for additional chemical reagents. Moreover, these dynamic hydrogels afford spatial control of substrate mechanics to direct human lymphatic endothelial cells (LECs) to form lymphatic cord-like structures (CLS). Specifically, cells cultured on viscoelastic supramolecular hydrogels have enhanced formation of CLS, arising from increased expression of key lymphatic markers, such as LYVE-1, Podoplanin, and Prox1, compared to static elastic hydrogels prepared from fully covalent cross-linking. Viscoelastic hydrogels promote lymphatic CLS formation through the expression of Nrp2, VEGFR2, and VEGFR3 to enhance the VEGF-C stimulation. Overall, viscoelastic supramolecular hydrogels offer a facile route to spatially control lymphatic CLS formation, providing a tool for future studies of basic lymphatic biology and tissue engineering applications.
Collapse
Affiliation(s)
- Fei Fan
- Bioengineering
Graduate Program, Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Bo Su
- Chemical
and Biomolecular Engineering, University
of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Alexander Kolodychak
- Chemical
and Biomolecular Engineering, University
of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Ephraim Ekwueme
- Bioengineering
Graduate Program, Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Laura Alderfer
- Bioengineering
Graduate Program, Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Sanjoy Saha
- Bioengineering
Graduate Program, Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Matthew J. Webber
- Chemical
and Biomolecular Engineering, University
of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Donny Hanjaya-Putra
- Bioengineering
Graduate Program, Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
28
|
Lemarié L, Dargar T, Grosjean I, Gache V, Courtial EJ, Sohier J. Human Induced Pluripotent Spheroids' Growth Is Driven by Viscoelastic Properties and Macrostructure of 3D Hydrogel Environment. Bioengineering (Basel) 2023; 10:1418. [PMID: 38136009 PMCID: PMC10740696 DOI: 10.3390/bioengineering10121418] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Stem cells, particularly human iPSCs, constitute a powerful tool for tissue engineering, notably through spheroid and organoid models. While the sensitivity of stem cells to the viscoelastic properties of their direct microenvironment is well-described, stem cell differentiation still relies on biochemical factors. Our aim is to investigate the role of the viscoelastic properties of hiPSC spheroids' direct environment on their fate. To ensure that cell growth is driven only by mechanical interaction, bioprintable alginate-gelatin hydrogels with significantly different viscoelastic properties were utilized in differentiation factor-free culture medium. Alginate-gelatin hydrogels of varying concentrations were developed to provide 3D environments of significantly different mechanical properties, ranging from 1 to 100 kPa, while allowing printability. hiPSC spheroids from two different cell lines were prepared by aggregation (⌀ = 100 µm, n > 1 × 104), included and cultured in the different hydrogels for 14 days. While spheroids within dense hydrogels exhibited limited growth, irrespective of formulation, porous hydrogels prepared with a liquid-liquid emulsion method displayed significant variations of spheroid morphology and growth as a function of hydrogel mechanical properties. Transversal culture (adjacent spheroids-laden alginate-gelatin hydrogels) clearly confirmed the separate effect of each hydrogel environment on hiPSC spheroid behavior. This study is the first to demonstrate that a mechanically modulated microenvironment induces diverse hiPSC spheroid behavior without the influence of other factors. It allows one to envision the combination of multiple formulations to create a complex object, where the fate of hiPSCs will be independently controlled by their direct microenvironment.
Collapse
Affiliation(s)
- Lucas Lemarié
- SEGULA Technologies, 69100 Villeurbanne, France;
- 3d.FAB, CNRS UMR 5246, ICBMS (Institute of Molecular and Supramolecular Chemistry and Biochemistry), Université Lyon 1, 69622 Villeurbanne, France;
- CNRS UMR 5305, LBTI (Tissue Biology and Therapeutic Engineering Laboratory), 69007 Lyon, France
| | - Tanushri Dargar
- CNRS UMR5261, INSERM U1315, INMG-PNMG (NeuroMyoGene Institute, Physiopathology and Genetics of the Neuron and the Muscle), Université Lyon 1, 69008 Lyon, France; (T.D.); (I.G.); (V.G.)
| | - Isabelle Grosjean
- CNRS UMR5261, INSERM U1315, INMG-PNMG (NeuroMyoGene Institute, Physiopathology and Genetics of the Neuron and the Muscle), Université Lyon 1, 69008 Lyon, France; (T.D.); (I.G.); (V.G.)
| | - Vincent Gache
- CNRS UMR5261, INSERM U1315, INMG-PNMG (NeuroMyoGene Institute, Physiopathology and Genetics of the Neuron and the Muscle), Université Lyon 1, 69008 Lyon, France; (T.D.); (I.G.); (V.G.)
| | - Edwin J. Courtial
- 3d.FAB, CNRS UMR 5246, ICBMS (Institute of Molecular and Supramolecular Chemistry and Biochemistry), Université Lyon 1, 69622 Villeurbanne, France;
| | - Jérôme Sohier
- CNRS UMR 5305, LBTI (Tissue Biology and Therapeutic Engineering Laboratory), 69007 Lyon, France
| |
Collapse
|
29
|
Piazza F, Sacco P, Marsich E, Baj G, Brun F, Asaro F, Grassi G, Grassi M, Donati I. Cell Activities on Viscoelastic Substrates Show an Elastic Energy Threshold and Correlate with the Linear Elastic Energy Loss in the Strain‐Softening Region. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202307224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Indexed: 02/06/2025]
Abstract
Energy‐sensing in viscoelastic substrates has recently been shown to be an important regulator of cellular activities, modulating mechanical transmission and transduction processes. Here, this study fine‐tunes the elastic energy of viscoelastic hydrogels with different physical and chemical compositions and shows that this has an impact on cell response in 2D cell cultures. This study shows that there is a threshold value for elastic energy (≈0.15 J m−3) above which cell adhesion is impaired. When hydrogels leave the linear stress–strain range, they show softening (plastic) behavior typical of soft tissues. This study identifies a correlation between the theoretical linear elastic energy loss in the strain‐softening region and the number of cells adhering to the substrate. This also has implications for the formation of vinculin‐rich anchorage points and the ability of cells to remodel the substrate through traction forces. Overall, the results reported in this study support that the relationship between cell activities and energy‐sensing in viscoelastic substrates is an important aspect to consider in the development of reliable ex vivo models of human tissues that mimic both normal and pathological conditions.
Collapse
Affiliation(s)
- Francesco Piazza
- Department of Life Sciences University of Trieste Via Licio Giorgieri 5 I‐34127 Trieste Italy
| | - Pasquale Sacco
- Department of Life Sciences University of Trieste Via Licio Giorgieri 5 I‐34127 Trieste Italy
| | - Eleonora Marsich
- Department of Medicine Surgery and Health Sciences University of Trieste Piazza dell'Ospitale 1 I‐34129 Trieste Italy
| | - Gabriele Baj
- Department of Life Sciences University of Trieste Via Licio Giorgieri 5 I‐34127 Trieste Italy
| | - Francesco Brun
- Department of Engineering and Architecture University of Trieste Via A. Valerio 6/1 I‐34127 Trieste Italy
| | - Fioretta Asaro
- Department of Chemical and Pharmaceutical Sciences University of Trieste Via Licio Giorgieri 1 I‐34127 Trieste Italy
| | - Gabriele Grassi
- Department of Life Sciences University of Trieste Via Licio Giorgieri 5 I‐34127 Trieste Italy
| | - Mario Grassi
- Department of Engineering and Architecture University of Trieste Via A. Valerio 6/1 I‐34127 Trieste Italy
| | - Ivan Donati
- Department of Life Sciences University of Trieste Via Licio Giorgieri 5 I‐34127 Trieste Italy
| |
Collapse
|
30
|
Lv H, Deng G, Lai J, Yu Y, Chen F, Yao J. Advances in 3D Bioprinting of Biomimetic and Engineered Meniscal Grafts. Macromol Biosci 2023; 23:e2300199. [PMID: 37436941 DOI: 10.1002/mabi.202300199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/14/2023]
Abstract
The meniscus plays a crucial role in loads distribution and protection of articular cartilage. Meniscal injury can result in cartilage degeneration, loss of mechanical stability in the knee joint and ultimately lead to arthritis. Surgical interventions provide only short-term pain relief but fail to repair or regenerate the injured meniscus. Emerging tissue engineering approaches based on 3D bioprinting provide alternatives to current surgical methods for meniscus repair. In this review, the current bioprinting techniques employed in developing engineered meniscus grafts are summarized and discuss the latest strategies for mimicking the gradient structure, composition, and viscoelastic properties of native meniscus. Recent progress is highlighted in gene-activated matrices for meniscus regeneration as well. Finally, a perspective is provided on the future development of 3D bioprinting for meniscus repair, emphasizing the potential of this technology to revolutionize meniscus regeneration and improve patient outcomes.
Collapse
Affiliation(s)
- Haiyuan Lv
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Guotao Deng
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jiaqi Lai
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yin Yu
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Fei Chen
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jun Yao
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
31
|
Lewns FK, Tsigkou O, Cox LR, Wildman RD, Grover LM, Poologasundarampillai G. Hydrogels and Bioprinting in Bone Tissue Engineering: Creating Artificial Stem-Cell Niches for In Vitro Models. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301670. [PMID: 37087739 PMCID: PMC11478930 DOI: 10.1002/adma.202301670] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Advances in bioprinting have enabled the fabrication of complex tissue constructs with high speed and resolution. However, there remains significant structural and biological complexity within tissues that bioprinting is unable to recapitulate. Bone, for example, has a hierarchical organization ranging from the molecular to whole organ level. Current bioprinting techniques and the materials employed have imposed limits on the scale, speed, and resolution that can be achieved, rendering the technique unable to reproduce the structural hierarchies and cell-matrix interactions that are observed in bone. The shift toward biomimetic approaches in bone tissue engineering, where hydrogels provide biophysical and biochemical cues to encapsulated cells, is a promising approach to enhancing the biological function and development of tissues for in vitro modeling. A major focus in bioprinting of bone tissue for in vitro modeling is creating dynamic microenvironmental niches to support, stimulate, and direct the cellular processes for bone formation and remodeling. Hydrogels are ideal materials for imitating the extracellular matrix since they can be engineered to present various cues whilst allowing bioprinting. Here, recent advances in hydrogels and 3D bioprinting toward creating a microenvironmental niche that is conducive to tissue engineering of in vitro models of bone are reviewed.
Collapse
Affiliation(s)
| | - Olga Tsigkou
- Department of MaterialsUniversity of ManchesterManchesterM1 5GFUK
| | - Liam R. Cox
- School of ChemistryUniversity of BirminghamBirminghamB15 2TTUK
| | - Ricky D. Wildman
- Faculty of EngineeringUniversity of NottinghamNottinghamNG7 2RDUK
| | - Liam M. Grover
- Healthcare Technologies InstituteSchool of Chemical EngineeringUniversity of BirminghamBirminghamB15 2TTUK
| | | |
Collapse
|
32
|
Zhang Y, Wang Z, Sun Q, Li Q, Li S, Li X. Dynamic Hydrogels with Viscoelasticity and Tunable Stiffness for the Regulation of Cell Behavior and Fate. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5161. [PMID: 37512435 PMCID: PMC10386333 DOI: 10.3390/ma16145161] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
The extracellular matrix (ECM) of natural cells typically exhibits dynamic mechanical properties (viscoelasticity and dynamic stiffness). The viscoelasticity and dynamic stiffness of the ECM play a crucial role in biological processes, such as tissue growth, development, physiology, and disease. Hydrogels with viscoelasticity and dynamic stiffness have recently been used to investigate the regulation of cell behavior and fate. This article first emphasizes the importance of tissue viscoelasticity and dynamic stiffness and provides an overview of characterization techniques at both macro- and microscale. Then, the viscoelastic hydrogels (crosslinked via ion bonding, hydrogen bonding, hydrophobic interactions, and supramolecular interactions) and dynamic stiffness hydrogels (softening, stiffening, and reversible stiffness) with different crosslinking strategies are summarized, along with the significant impact of viscoelasticity and dynamic stiffness on cell spreading, proliferation, migration, and differentiation in two-dimensional (2D) and three-dimensional (3D) cell cultures. Finally, the emerging trends in the development of dynamic mechanical hydrogels are discussed.
Collapse
Affiliation(s)
- Yuhang Zhang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China (Q.L.)
- National Center for International Joint Research of Micro-Nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Zhuofan Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China (Q.L.)
- 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
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China (Q.L.)
- National Center for International Joint Research of Micro-Nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Shaohui Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China (Q.L.)
- National Center for International Joint Research of Micro-Nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
33
|
Shou Y, Teo XY, Wu KZ, Bai B, Kumar ARK, Low J, Le Z, Tay A. Dynamic Stimulations with Bioengineered Extracellular Matrix-Mimicking Hydrogels for Mechano Cell Reprogramming and Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300670. [PMID: 37119518 PMCID: PMC10375194 DOI: 10.1002/advs.202300670] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/10/2023] [Indexed: 06/19/2023]
Abstract
Cells interact with their surrounding environment through a combination of static and dynamic mechanical signals that vary over stimulus types, intensity, space, and time. Compared to static mechanical signals such as stiffness, porosity, and topography, the current understanding on the effects of dynamic mechanical stimulations on cells remains limited, attributing to a lack of access to devices, the complexity of experimental set-up, and data interpretation. Yet, in the pursuit of emerging translational applications (e.g., cell manufacturing for clinical treatment), it is crucial to understand how cells respond to a variety of dynamic forces that are omnipresent in vivo so that they can be exploited to enhance manufacturing and therapeutic outcomes. With a rising appreciation of the extracellular matrix (ECM) as a key regulator of biofunctions, researchers have bioengineered a suite of ECM-mimicking hydrogels, which can be fine-tuned with spatiotemporal mechanical cues to model complex static and dynamic mechanical profiles. This review first discusses how mechanical stimuli may impact different cellular components and the various mechanobiology pathways involved. Then, how hydrogels can be designed to incorporate static and dynamic mechanical parameters to influence cell behaviors are described. The Scopus database is also used to analyze the relative strength in evidence, ranging from strong to weak, based on number of published literatures, associated citations, and treatment significance. Additionally, the impacts of static and dynamic mechanical stimulations on clinically relevant cell types including mesenchymal stem cells, fibroblasts, and immune cells, are evaluated. The aim is to draw attention to the paucity of studies on the effects of dynamic mechanical stimuli on cells, as well as to highlight the potential of using a cocktail of various types and intensities of mechanical stimulations to influence cell fates (similar to the concept of biochemical cocktail to direct cell fate). It is envisioned that this progress report will inspire more exciting translational development of mechanoresponsive hydrogels for biomedical applications.
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
| | - Xin Yong Teo
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Kenny Zhuoran Wu
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Bingyu Bai
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Arun R. K. Kumar
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore117597Singapore
| | - Jessalyn Low
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Zhicheng Le
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
| | - Andy Tay
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
- NUS Tissue Engineering ProgramNational University of SingaporeSingapore117510Singapore
| |
Collapse
|
34
|
Kalashnikov N, Moraes C. Substrate viscoelasticity affects human macrophage morphology and phagocytosis. SOFT MATTER 2023; 19:2438-2445. [PMID: 36930245 DOI: 10.1039/d2sm01683d] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Viscoelasticity is an inherent characteristic of many living tissues and, in an attempt to better recapitulate this aspect in cell culture, hydrogel biomaterials have been engineered to exhibit time-dependent energy-dissipative mechanical behavior. Viscoelastic hydrogel culture platforms have been instrumental in understanding the biological effects of viscoelasticity. Although viscoelasticity has been shown to regulate fundamental cell processes such as spreading and differentiation in adherent cells, the influence of viscoelasticity on macrophage behavior has not been explored. Here, we use a tunable viscoelastic polyacrylamide hydrogel culture system to demonstrate that viscoelasticity is an important biophysical regulator of macrophage function. After biologically validating our system with HS-5 fibroblasts to show behavior consistent with existing reports, we seed human THP-1 monocytes on these viscoelastic substrates and differentiate them into macrophages. THP-1 macrophages become smaller and rounder, and less efficient at phagocytosis on more viscous polyacrylamide hydrogel substrates. Since macrophages play key roles in mounting responses such as inflammation and fibrosis, these results indicate that viscoelasticity is an important parameter in the design of immunomodulatory biomaterials.
Collapse
Affiliation(s)
- Nikita Kalashnikov
- Department of Chemical Engineering, McGill University, Montreal, Canada.
| | - Christopher Moraes
- Department of Chemical Engineering, McGill University, Montreal, Canada.
- Department of Biological and Biomedical Engineering, McGill University, Montreal, Canada
- Goodman Cancer Research Center, McGill University, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| |
Collapse
|
35
|
Farsheed AC, Thomas AJ, Pogostin BH, Hartgerink JD. 3D Printing of Self-Assembling Nanofibrous Multidomain Peptide Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210378. [PMID: 36604310 PMCID: PMC10023392 DOI: 10.1002/adma.202210378] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/13/2022] [Indexed: 05/25/2023]
Abstract
3D printing has become one of the primary fabrication strategies used in biomedical research. Recent efforts have focused on the 3D printing of hydrogels to create structures that better replicate the mechanical properties of biological tissues. These pose a unique challenge, as soft materials are difficult to pattern in three dimensions with high fidelity. Currently, a small number of biologically derived polymers that form hydrogels are frequently reused for 3D printing applications. Thus, there exists a need for novel hydrogels with desirable biological properties that can be used as 3D printable inks. In this work, the printability of multidomain peptides (MDPs), a class of self-assembling peptides that form a nanofibrous hydrogel at low concentrations, is established. MDPs with different charge functionalities are optimized as distinct inks and are used to create complex 3D structures, including multi-MDP prints. Additionally, printed MDP constructs are used to demonstrate charge-dependent differences in cellular behavior in vitro. This work presents the first time that self-assembling peptides have been used to print layered structures with overhangs and internal porosity. Overall, MDPs are a promising new class of 3D printable inks that are uniquely peptide-based and rely solely on supramolecular mechanisms for assembly.
Collapse
Affiliation(s)
- Adam C Farsheed
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA
| | - Adam J Thomas
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | - Brett H Pogostin
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA
| | - Jeffrey D Hartgerink
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| |
Collapse
|
36
|
Miranda Alarcón YS, Jazwinska D, Lymon T, Khalili A, Browe D, Newton B, Pellegrini M, Cohen RI, Shreiber DI, Freeman JW. The Use of Collagen Methacrylate in Actuating Polyethylene Glycol Diacrylate-Acrylic Acid Scaffolds for Muscle Regeneration. Ann Biomed Eng 2023; 51:1165-1180. [PMID: 36853478 DOI: 10.1007/s10439-023-03139-8] [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: 05/25/2022] [Accepted: 01/03/2023] [Indexed: 03/01/2023]
Abstract
After muscle loss or injury, skeletal muscle tissue has the ability to regenerate and return its function. However, large volume defects in skeletal muscle tissue pose a challenge to regenerate due to the absence of regenerative elements such as biophysical and biochemical cues, making the development of new treatments necessary. One potential solution is to utilize electroactive polymers that can change size or shape in response to an external electric field. Poly(ethylene glycol) diacrylate (PEGDA) is one such polymer, which holds great potential as a scaffold for muscle tissue regeneration due to its mechanical properties. In addition, the versatile chemistry of this polymer allows for the conjugation of new functional groups to enhance its electroactive properties and biocompatibility. Herein, we have developed an electroactive copolymer of PEGDA and acrylic acid (AA) in combination with collagen methacrylate (CMA) to promote cell adhesion and proliferation. The electroactive properties of the CMA + PEGDA:AA constructs were investigated through actuation studies. Furthermore, the biological properties of the hydrogel were investigated in a 14-day in vitro study to evaluate myosin light chain (MLC) expression and metabolic activity of C2C12 mouse myoblast cells. The addition of CMA improved some aspects of material bioactivity, such as MLC expression in C2C12 mouse myoblast cells. However, the incorporation of CMA in the PEGDA:AA hydrogels reduced the sample movement when placed under an electric field, possibly due to steric hindrance from the CMA. Further research is needed to optimize the use of CMA in combination with PEGDA:AA as a potential scaffold for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
| | - Dorota Jazwinska
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Terrence Lymon
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Amin Khalili
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Daniel Browe
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Brandon Newton
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Michael Pellegrini
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Rick I Cohen
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Joseph W Freeman
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
37
|
Pizzolitto C, Scognamiglio F, Sacco P, Lipari S, Romano M, Donati I, Marsich E. Immediate stress dissipation in dual cross-link hydrogels controls osteogenic commitment of mesenchymal stem cells. Carbohydr Polym 2023; 302:120369. [PMID: 36604049 DOI: 10.1016/j.carbpol.2022.120369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/21/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022]
Abstract
In vitro studies of mesenchymal stem cells (MSCs) differentiation have been predominantly performed with non-physiologically elastic materials. Here we report the effect of different viscoplastic ECM mimics on the osteogenic engagement of MSCs in 2D. We have developed soft hydrogels, composed of a lactose-modified chitosan, using a combination of permanent and temporary cross-links. The presence of temporary cross-links has a minor effect on the shear modulus of the hydrogels, but causes an immediate relaxation (dissipation) of the applied stress. This material property leads to early osteogenic commitment of MSCs, as evidenced by gene expression of runt-related transcription factor 2 (RUNX2), type 1 collagen (COL1A1), osteocalcin (OCN), alkaline phosphatase enzyme activity (ALP) and calcium deposit formation. In contrast, cells cultured on purely elastic hydrogels with only permanent cross-link begin to differentiate only after a longer period of time, indicating a dissipation-mediated mechano-sensing in the osteogenic commitment of MSCs.
Collapse
Affiliation(s)
- Chiara Pizzolitto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, I-34129 Trieste, Italy
| | - Francesca Scognamiglio
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, I-34129 Trieste, Italy
| | - Pasquale Sacco
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, I-34129 Trieste, Italy; Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy; AREA Science Park, loc. Padriciano 99, I-34149 Trieste, Italy.
| | - Sara Lipari
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Maurizio Romano
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Ivan Donati
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Eleonora Marsich
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, I-34129 Trieste, Italy
| |
Collapse
|
38
|
Viscoelastic hydrogels for interrogating pancreatic cancer-stromal cell interactions. Mater Today Bio 2023; 19:100576. [PMID: 36816601 PMCID: PMC9929443 DOI: 10.1016/j.mtbio.2023.100576] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/05/2023] Open
Abstract
The tumor microenvironment (TME) is known to direct cancer cell growth, migration, invasion into the matrix and distant tissues, and to confer drug resistance in cancer cells. While multiple aspects of TME have been studied using in vitro, ex vivo, and in vivo tumor models and engineering tools, the influence of matrix viscoelasticity on pancreatic cancer cells and its associated TME remained largely unexplored. In this contribution, we synthesized a new biomimetic hydrogel with tunable matrix stiffness and stress-relaxation for evaluating the effect of matrix viscoelasticity on pancreatic cancer cell (PCC) behaviors in vitro. Using three simple monomers and Reverse-Addition Fragmentation Chain-Transfer (RAFT) polymerization, we synthesized a new class of phenylboronic acid containing polymers (e.g., poly (OEGA-s-HEAA-s-APBA) or PEHA). Norbornene group was conjugated to HEAA on PEHA via carbic anhydride, affording a new NB and BA dually modified polymer - PEHNBA amenable for orthogonal thiol-norbornene photopolymerization and boronate ester diol complexation. The former provided tunable matrix elasticity, while the latter gave rise to matrix stress-relaxation (or viscoelasticity). The new PEHNBA polymers were shown to be highly cytocompatible for in situ encapsulation of PCCs and cancer-associated fibroblasts (CAFs). Furthermore, we demonstrated that hydrogels with high stress-relaxation promoted spreading of CAFs, which in turns promoted PCC proliferation and spreading in the viscoelastic matrix. Compared with elastic matrix, viscoelastic gels upregulated the secretion of soluble proteins known to promote epithelial-mesenchymal transition (EMT). This study demonstrated the crucial influence of matrix viscoelasticity on pancreatic cancer cell fate and provided an engineered viscoelastic matrix for future studies and applications related to TME.
Collapse
|
39
|
Scognamiglio F, Cok M, Piazza F, Marsich E, Pacor S, Aarstad OA, Aachmann FL, Donati I. Hydrogels based on methylated-alginates as a platform to investigate the effect of material properties on cell activity. The role of material compliance. Carbohydr Polym 2023; 311:120745. [PMID: 37028873 DOI: 10.1016/j.carbpol.2023.120745] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/14/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Alginate-based hydrogels with tunable mechanical properties are developed by chemical methylation of the polysaccharide backbone, which was performed either in homogeneous phase (in solution) or in heterogeneous phase (on hydrogels). Nuclear Magnetic Resonance (NMR) and Size Exclusion Chromatography (SEC-MALS) analyses of methylated alginates allow to identify the presence and location of methyl groups on the polysaccharide, and to investigate the influence of methylation on the stiffness of the polymer chains. The methylated polysaccharides are employed for the manufacturing of calcium-reticulated hydrogels for cell growth in 3D. The rheological characterization shows that the shear modulus of hydrogels is dependent on the amount of cross-linker used. Methylated alginates represent a platform to explore the effect of mechanical properties on cell activity. As an example, the effect of compliance is investigated using hydrogels displaying similar shear modulus. An osteosarcoma cell line (MG-63) was encapsulated in the alginate hydrogels and the effect of material compliance on cell proliferation and localization of YAP/TAZ protein complex is investigated by flow cytometry and immunohistochemistry, respectively. The results point out that an increase of material compliance leads to an increase of the proliferative rate of cells and correlates with the translocation of YAP/TAZ inside the cell nucleus.
Collapse
Affiliation(s)
- Francesca Scognamiglio
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy.
| | - Michela Cok
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy
| | - Francesco Piazza
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy
| | - Eleonora Marsich
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34129 Trieste, Italy
| | - Sabrina Pacor
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy
| | - Olav A Aarstad
- Norwegian Biopolymer Laboratory (NOBIPOL), Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Sem Sælands vei 6/8, 7491 Trondheim, Norway
| | - Finn L Aachmann
- Norwegian Biopolymer Laboratory (NOBIPOL), Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Sem Sælands vei 6/8, 7491 Trondheim, Norway
| | - Ivan Donati
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy
| |
Collapse
|
40
|
Qiu Z, Zhu H, Wang Y, Kasimu A, Li D, He J. Functionalized alginate-based bioinks for microscale electrohydrodynamic bioprinting of living tissue constructs with improved cellular spreading and alignment. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00225-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
41
|
Viscoelastic modeling of the stress relaxation behavior for the bionic extracellular matrix polymer scaffold. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
42
|
Choi JH, In Kim S, Seo JS, Tumursukh NE, Kim SE, Choe SH, Kim SJ, Park S, Song JE, Khang G. Fast stress relaxing gellan gum that enhances the microenvironment and secreting function of bone mesenchymal stem cells. Int J Biol Macromol 2022; 222:2144-2157. [DOI: 10.1016/j.ijbiomac.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/19/2022] [Accepted: 10/02/2022] [Indexed: 11/05/2022]
|
43
|
Chen X, Sun T, Wei Z, Chen Z, Wang H, Huang Q, Fukuda T, Shi Q. A clamp-free micro-stretching system for evaluating the viscoelastic response of cell-laden microfibers. Biosens Bioelectron 2022; 214:114517. [DOI: 10.1016/j.bios.2022.114517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 12/24/2022]
|
44
|
Interactions between infernan and calcium: From the molecular level to the mechanical properties of microgels. Carbohydr Polym 2022; 292:119629. [DOI: 10.1016/j.carbpol.2022.119629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/05/2022] [Accepted: 05/14/2022] [Indexed: 11/20/2022]
|
45
|
Fabrication and Evaluation of Gellan Gum/Hyaluronic Acid Hydrogel for Retinal Tissue Engineering Biomaterial and the Influence of Substrate Stress Relaxation on Retinal Pigment Epithelial Cells. Molecules 2022; 27:molecules27175512. [PMID: 36080277 PMCID: PMC9458149 DOI: 10.3390/molecules27175512] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Cell therapies for age-related macular degeneration (AMD) treatment have been developed by integrating hydrogel-based biomaterials. Until now, cell activity has been observed only in terms of the modulus of the hydrogel. In addition, cell behavior has only been observed in the 2D environment of the hydrogel and the 3D matrix. As time-dependent stress relaxation is considered a significant mechanical cue for the control of cellular activities, it is important to optimize hydrogels for retinal tissue engineering (TE) by applying this viewpoint. Herein, a gellan Gum (GG)/Hyaluronic acid (HA) hydrogel was fabricated using a facile physical crosslinking method. The physicochemical and mechanical properties were controlled by forming a different composition of GG and HA. The characterization was performed by conducting a mass swelling study, a sol fraction study, a weight loss test, a viscosity test, an injection force study, a compression test, and a stress relaxation analysis. The biological activity of the cells encapsulated in 3D constructs was evaluated by conducting a morphological study, a proliferation test, a live/dead analysis, histology, immunofluorescence staining, and a gene expression study to determine the most appropriate material for retinal TE biomaterial. Hydrogels with moderate amounts of HA showed improved physicochemical and mechanical properties suitable for injection into the retina. Moreover, the time-dependent stress relaxation property of the GG/HA hydrogel was enhanced when the appropriate amount of HA was loaded. In addition, the cellular compatibility of the GG/HA hydrogel in in vitro experiments was significantly improved in the fast-relaxing hydrogel. Overall, these results demonstrate the remarkable potential of GG/HA hydrogel as an injectable hydrogel for retinal TE and the importance of the stress relaxation property when designing retinal TE hydrogels. Therefore, we believe that GG/HA hydrogel is a prospective candidate for retinal TE biomaterial.
Collapse
|
46
|
Ling SD, Liu Z, Ma W, Chen Z, Du Y, Xu J. A Novel Step-T-Junction Microchannel for the Cell Encapsulation in Monodisperse Alginate-Gelatin Microspheres of Varying Mechanical Properties at High Throughput. BIOSENSORS 2022; 12:bios12080659. [PMID: 36005055 PMCID: PMC9406195 DOI: 10.3390/bios12080659] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022]
Abstract
Cell encapsulation has been widely employed in cell therapy, characterization, and analysis, as well as many other biomedical applications. While droplet-based microfluidic technology is advantageous in cell microencapsulation because of its modularity, controllability, mild conditions, and easy operation when compared to other state-of-art methods, it faces the dilemma between high throughput and monodispersity of generated cell-laden microdroplets. In addition, the lack of a biocompatible method of de-emulsification transferring cell-laden hydrogel from cytotoxic oil phase into cell culture medium also hurtles the practical application of microfluidic technology. Here, a novel step-T-junction microchannel was employed to encapsulate cells into monodisperse microspheres at the high-throughput jetting regime. An alginate–gelatin co-polymer system was employed to enable the microfluidic-based fabrication of cell-laden microgels with mild cross-linking conditions and great biocompatibility, notably for the process of de-emulsification. The mechanical properties of alginate-gelatin hydrogel, e.g., stiffness, stress–relaxation, and viscoelasticity, are fully adjustable in offering a 3D biomechanical microenvironment that is optimal for the specific encapsulated cell type. Finally, the encapsulation of HepG2 cells into monodisperse alginate–gelatin microgels with the novel microfluidic system and the subsequent cultivation proved the maintenance of the long-term viability, proliferation, and functionalities of encapsulated cells, indicating the promising potential of the as-designed system in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Si Da Ling
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Zhiqiang Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences Tsinghua University, Beijing 100084, China
| | - Wenjun Ma
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Zhuo Chen
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Correspondence: (Z.C.); (Y.D.); (J.X.)
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences Tsinghua University, Beijing 100084, China
- Correspondence: (Z.C.); (Y.D.); (J.X.)
| | - Jianhong Xu
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Correspondence: (Z.C.); (Y.D.); (J.X.)
| |
Collapse
|
47
|
Liu D, Zhang H, Dong X, Sang L, Qi M. Effect of viscoelastic properties of cellulose nanocrystal/collagen hydrogels on chondrocyte behaviors. Front Bioeng Biotechnol 2022; 10:959409. [PMID: 36032700 PMCID: PMC9403537 DOI: 10.3389/fbioe.2022.959409] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Cartilage tissue engineering technology provides a solution for treating osteoarthritis. Based on the viscoelastic nature of articular cartilage, many viscoelastic hydrogel scaffolds have been developed for investigating the effects on chondrocyte behaviors. However, cellulose nanocrystal/collagen (CNC/COL) hydrogels have not been used as a viscoelastic microenvironment to study chondrocyte growth. Here, we prepared CNC/COL hydrogels with tunable viscoelastic properties and investigated their influences on chondrocyte behaviors. The results showed that CNC and COL within the hydrogels are bonded by hydrogen bonds. The hydrogels had a microporous structure, and the viscoelastic properties were enhanced by increasing the concentration of CNC. Moreover, enhancing the hydrogel viscoelastic properties, including stress relaxation, creep, storage modulus, and loss modulus, promoted the cell shape change, proliferation, and matrix deposition and reduced the IL-1β level. Using a principal component analysis (PCA), stress relaxation was assessed to have the strongest correlation with chondrocytes behaviors, with an authority weight value of 62.547%. More importantly, FAK and YAP were involved in the chondrocytes’ response to the rapid relaxing hydrogel by immunofluorescence staining.
Collapse
Affiliation(s)
- Donglei Liu
- School of Basic Medicine, Binzhou Medical University, Yantai, China
- School of Materials Science and Engineering, Dalian University of Technology, Dalian, China
| | - Hao Zhang
- School of Materials Science and Engineering, Dalian University of Technology, Dalian, China
- Department of Orthopedics, Central Hospital of Dalian University of Technology, Dalian, China
- Changchun SinoBiomaterials Co., Ltd., Changchun, China
- *Correspondence: Hao Zhang, ; Xufeng Dong,
| | - Xufeng Dong
- School of Materials Science and Engineering, Dalian University of Technology, Dalian, China
- *Correspondence: Hao Zhang, ; Xufeng Dong,
| | - Lin Sang
- School of Automotive Engineering, Dalian University of Technology, Dalian, China
| | - Min Qi
- School of Materials Science and Engineering, Dalian University of Technology, Dalian, China
| |
Collapse
|
48
|
Zhang R, Zhang D, Sun X, Song X, Yan KC, Liang H. Polyvinyl alcohol/gelatin hydrogels regulate cell adhesion and chromatin accessibility. Int J Biol Macromol 2022; 219:672-684. [PMID: 35952815 DOI: 10.1016/j.ijbiomac.2022.08.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 12/24/2022]
Abstract
Cell adhesion has a critical influence on various processes such as cancer metastasis and wound healing. Many substrates have been used for studying cell adhesion and its related biological processes, it is still highly desirable to have a simply prepared and low-cost substrate suitable for regulating cell adhesion. In this study, we produced a series of polyvinyl alcohol/gelatin hydrogels with different gelatin concentrations via dry-annealing method. Our data showed that the protein adsorbing capability was enhanced and cell adhesion area and the ratio of non-spherical cells were increased with the increment of gelatin concentration. We also observed that varying cell adhesion conditions induced by polyvinyl alcohol /gelatin hydrogels resulted in expression level changes of genes involved in mechanotransduction from extracellular matrices (ECM) to the nucleus. In particular, we detected a widespread increase in chromatin accessibility under poor cell adhesion condition. This work provides a useful hydrogel system for regulating cell adhesion and opens up new possibilities for the design of biomaterials for cell adhesion study.
Collapse
Affiliation(s)
- Ran Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Duo Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Xingyue Sun
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Xiaoyuan Song
- MOE Key Laboratory for Cellular Dynamics, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Karen Chang Yan
- Mechanical Engineering and Biomedical Engineering, The College of New Jersey, Ewing, NJ, USA.
| | - Haiyi Liang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China; School of Civil Engineering, Anhui Jianzhu University, Hefei, China; IAT-Chungu Joint Laboratory for Additive Manufacturing, Anhui Chungu 3D printing Institute of Intelligent Equipment and Industrial Technology, Wuhu, China.
| |
Collapse
|
49
|
Norris SCP, Kawecki NS, Davis AR, Chen KK, Rowat AC. Emulsion-templated microparticles with tunable stiffness and topology: Applications as edible microcarriers for cultured meat. Biomaterials 2022; 287:121669. [PMID: 35853359 PMCID: PMC9834440 DOI: 10.1016/j.biomaterials.2022.121669] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 06/27/2022] [Accepted: 07/02/2022] [Indexed: 01/16/2023]
Abstract
Cultured meat has potential to diversify methods for protein production, but innovations in production efficiency will be required to make cultured meat a feasible protein alternative. Microcarriers provide a strategy to culture sufficient volumes of adherent cells in a bioreactor that are required for meat products. However, cell culture on inedible microcarriers involves extra downstream processing to dissociate cells prior to consumption. Here, we present edible microcarriers that can support the expansion and differentiation of myogenic cells in a single bioreactor system. To fabricate edible microcarriers with a scalable process, we used water-in-oil emulsions as templates for gelatin microparticles. We also developed a novel embossing technique to imprint edible microcarriers with grooved topology in order to test if microcarriers with striated surface texture can promote myoblast proliferation and differentiation in suspension culture. In this proof-of-concept demonstration, we showed that edible microcarriers with both smooth and grooved surface topologies supported the proliferation and differentiation of mouse myogenic C2C12 cells in a suspension culture. The grooved edible microcarriers showed a modest increase in the proliferation and alignment of myogenic cells compared to cells cultured on smooth, spherical microcarriers. During the expansion phase, we also observed the formation of cell-microcarrier aggregates or 'microtissues' for cells cultured on both smooth and grooved microcarriers. Myogenic microtissues cultured with smooth and grooved microcarriers showed similar characteristics in terms of myotube length, myotube volume fraction, and expression of myogenic markers. To establish feasibility of edible microcarriers for cultured meat, we showed that edible microcarriers supported the production of myogenic microtissue from C2C12 or bovine satellite muscle cells, which we harvested by centrifugation into a cookable meat patty that maintained its shape and exhibited browning during cooking. These findings demonstrate the potential of edible microcarriers for the scalable production of cultured meat in a single bioreactor.
Collapse
Affiliation(s)
- Sam C P Norris
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - N Stephanie Kawecki
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ashton R Davis
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kathleen K Chen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
50
|
Prouvé E, Rémy M, Feuillie C, Molinari M, Chevallier P, Drouin B, Laroche G, Durrieu MC. Interplay of matrix stiffness and stress relaxation in directing osteogenic differentiation of mesenchymal stem cells. Biomater Sci 2022; 10:4978-4996. [PMID: 35801706 DOI: 10.1039/d2bm00485b] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aim of this study is to investigate the impact of the stiffness and stress relaxation of poly(acrylamide-co-acrylic acid) hydrogels on the osteogenic differentiation of human mesenchymal stem cells (hMSCs). Varying the amount of the crosslinker and the ratio between the monomers enabled the obtainment of hydrogels with controlled mechanical properties, as characterized using unconfined compression and atomic force microscopy (AFM). Subsequently, the surface of the hydrogels was functionalized with a mimetic peptide of the BMP-2 protein, in order to favor the osteogenic differentiation of hMSCs. Finally, hMSCs were cultured on the hydrogels with different stiffness and stress relaxation: 15 kPa - 15%, 60 kPa - 15%, 140 kPa - 15%, 100 kPa - 30%, and 140 kPa - 70%. The cells on hydrogels with stiffnesses from 60 kPa to 140 kPa presented a star-like shape, typical of osteocytes, which has only been reported by our group for two-dimensional substrates. Then, the extent of hMSC differentiation was evaluated by using immunofluorescence and by quantifying the expression of both osteoblast markers (Runx-2 and osteopontin) and osteocyte markers (E11, DMP1, and sclerostin). It was found that a stiffness of 60 kPa led to a higher expression of osteocyte markers as compared to stiffnesses of 15 and 140 kPa. Finally, the strongest expression of osteoblast and osteocyte differentiation markers was observed for the hydrogel with a high relaxation of 70% and a stiffness of 140 kPa.
Collapse
Affiliation(s)
- Emilie Prouvé
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux, Université Laval, 1065 Avenue de la médecine, Québec G1V 0A6, Canada. .,Axe médecine régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada.,Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| | - Murielle Rémy
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| | - Cécile Feuillie
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| | - Michael Molinari
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| | - Pascale Chevallier
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux, Université Laval, 1065 Avenue de la médecine, Québec G1V 0A6, Canada. .,Axe médecine régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada
| | - Bernard Drouin
- Axe médecine régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada
| | - Gaétan Laroche
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux, Université Laval, 1065 Avenue de la médecine, Québec G1V 0A6, Canada. .,Axe médecine régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada
| | - Marie-Christine Durrieu
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN), Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,CNRS, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.,Bordeaux INP, CBMN UMR5248, Allée Geoffroy Saint Hilaire - Bât B14, 33600 Pessac, France.
| |
Collapse
|