1
|
Ioannidis K, Dimopoulos A, Decoene I, Guilliams M, Svitina H, Storozhuk L, de Oliveira‐Silva R, Basov S, Thanh NTK, Mourdikoudis S, Van Bael MJ, Smeets B, Sakellariou D, Papantoniou I. 4D Biofabrication of Magnetically Augmented Callus Assembloid Implants Enables Rapid Endochondral Ossification via Activation of Mechanosensitive Pathways. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413680. [PMID: 39998420 PMCID: PMC12005758 DOI: 10.1002/advs.202413680] [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: 10/25/2024] [Revised: 01/31/2025] [Indexed: 02/26/2025]
Abstract
The use of magnetic-driven strategies for non-contact manipulation of engineered living modules opens up new possibilities for tissue engineering. The integration of magnetic nanoparticles (MNPs) with cartilaginous microtissues enables model-driven 4D bottom-up biofabrication of remotely actuated assembloids, providing unique properties to mechanoresponsive tissues, particularly skeletal constructs. However, for clinical use, the long-term effects of magnetic stimulation on phenotype and in vivo functionality need further exploration. Magnetic-driven biofabrication includes both rapid processes, such as guided microtissue assembly, and slower biological processes, like extracellular matrix secretion. This work explores the interplay between magnetic fields and MNP-loaded cartilaginous microtissues through mathematical modeling and experimental approaches, investigating long-term stimulation effects on ECM maturation and chondrogenic hypertrophy. Transcriptomic analysis reveal that magnetic stimulation activated mechanosensitive pathways and catabolic processes, driving accelerated cartilage-to-bone transitions via endochondral ossification, outcomes not observed in non-stimulated controls. This study paves the way for pre-programmed, remotely actuated skeletal assembloids with superior bone-forming capacity for regenerating challenging bone fractures.
Collapse
Affiliation(s)
- Konstantinos Ioannidis
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Andreas Dimopoulos
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Isaak Decoene
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Maya Guilliams
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
- MeBioS division, Biosystems DepartmentKU LeuvenKasteelpark, Arenberg 30Leuven3001Belgium
| | - Hanna Svitina
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| | - Liudmyla Storozhuk
- Healthcare Biomagnetics and Nanomaterials Laboratories, Department of Medical Physics and Biomedical EngineeringUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- London Centre for NanotechnologyUniversity College London17‐19 Gordon StreetLondonWC1H 0AHUK
| | - Rodrigo de Oliveira‐Silva
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular SystemsKU LeuvenCelestijnenlaan 200F, PB 2454Leuven3001Belgium
| | - Sergey Basov
- Quantum Solid State Physics, Department of Physics and AstronomyKU LeuvenCelestijnenlaan 200DLeuven3001Belgium
| | - Nguyen Thi Kim Thanh
- Healthcare Biomagnetics and Nanomaterials Laboratories, Department of Medical Physics and Biomedical EngineeringUniversity College London21 Albemarle StreetLondonW1S 4BSUK
- Biophysics Group, Department of Physics and AstronomyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Stefanos Mourdikoudis
- CINBIO, Department of Physical Chemistry, Campus Universitario, Lagoas MarcosendeUniversidade de VigoVigo36310Spain
| | - Margriet J. Van Bael
- Quantum Solid State Physics, Department of Physics and AstronomyKU LeuvenCelestijnenlaan 200DLeuven3001Belgium
| | - Bart Smeets
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
- MeBioS division, Biosystems DepartmentKU LeuvenKasteelpark, Arenberg 30Leuven3001Belgium
| | - Dimitrios Sakellariou
- Membrane Separations, Adsorption, Catalysis, and Spectroscopy for Sustainable Solutions (cMACS), Department of Microbial and Molecular SystemsKU LeuvenCelestijnenlaan 200F, PB 2454Leuven3001Belgium
| | - Ioannis Papantoniou
- Prometheus Translational Division of Skeletal Tissue EngineeringKU Leuven, O&N1, Herestraat 49, PB 813Leuven3000Belgium
- Skeletal Biology and Engineering Research Centre, Department of Development & RegenerationKU LeuvenO&N1, Herestraat 49, PB 813Leuven3000Belgium
| |
Collapse
|
2
|
Yan X, Fu S, Xie Y, Zhang C, Wu X. Piezo1-driven mechanotransduction as a key regulator of cartilage degradation in early osteoarthritis. BIOMOLECULES & BIOMEDICINE 2025; 25:905-913. [PMID: 39388709 PMCID: PMC11959384 DOI: 10.17305/bb.2024.11156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease characterized by pain and cartilage damage in its later stages, while early OA is marked by the loss of cartilage's mechanical function. Recent studies suggest that Piezo1, a mechanotransducer, may contribute to cartilage degradation under abnormal physical stress. This study investigates the mechanism by which Piezo1 mediates the loss of cartilage's mechanical properties. Using rat chondrocytes cultured in a 3D in vitro model, we found that fluid flow-induced physical stress activates constitutively expressed Piezo1, leading to increased catabolic activity and apoptosis, which, in turn, disrupts the matrix structure. Ex vivo cartilage experiments further demonstrated that the mechanical stress-induced loss of cartilage's physical properties (approximately 10% reduction in relaxation modulus) is mediated by Piezo1 and depends on cell viability. Notably, Piezo1 agonists alone did not alter the mechanical behavior of cartilage tissue. In vivo, using an OA rat model induced by anterior cruciate ligament transection, we observed cartilage integrity degradation and loss of mechanical properties, which were partially mitigated by Piezo1 inhibition. RNA sequencing revealed significant modulation of the PI3K signaling and matrix regulation pathways. Collectively, this study demonstrates that Piezo1-mediated catabolic activity in chondrocytes is a key driver of the loss of cartilage's mechanical function during the relaxation phase.
Collapse
Affiliation(s)
- Xu Yan
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Su Fu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Xie
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunlin Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuejian Wu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Wright C, Zotter SF, Tung WS, Reikersdorfer K, Homer A, Kheir N, Paschos N. Current Concepts and Clinical Applications in Cartilage Tissue Engineering. Tissue Eng Part A 2025; 31:87-99. [PMID: 39812645 DOI: 10.1089/ten.tea.2024.0300] [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: 01/16/2025] Open
Abstract
Cartilage injuries are extremely common in the general population, and conventional interventions have failed to produce optimal results. Tissue engineering (TE) technology has been developed to produce neocartilage for use in a variety of cartilage-related conditions. However, progress in the field of cartilage TE has historically been difficult due to the high functional demand and avascular nature of the tissue. Recent advancements in cell sourcing, biostimulation, and scaffold technology have revolutionized the field and made the clinical application of this technology a reality. Cartilage engineering technology will continue to expand its horizons to fully integrate three-dimensional printing, gene editing, and optimal cell sourcing in the future. This review focuses on the recent advancements in the field of cartilage TE and the landscape of clinical treatments for a variety of cartilage-related conditions.
Collapse
Affiliation(s)
- Connor Wright
- University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Orthopaedics, Massachusetts General Brigham, Boston, MA, USA
| | | | - Wei Shao Tung
- Department of Orthopaedics, Massachusetts General Brigham, Boston, MA, USA
| | - Kristen Reikersdorfer
- Department of Orthopaedics, Massachusetts General Brigham, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Andrew Homer
- Department of Orthopaedics, Massachusetts General Brigham, Boston, MA, USA
| | - Nadim Kheir
- Department of Orthopaedics, Massachusetts General Brigham, Boston, MA, USA
| | - Nikolaos Paschos
- Department of Orthopaedics, Massachusetts General Brigham, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Liu Z, Zhou H, Wu Q, Luo T, Tu H, Sa G, Yang X. Constructing condylar cartilage organoid to explore primary cilia functions. Heliyon 2024; 10:e35972. [PMID: 39281559 PMCID: PMC11395755 DOI: 10.1016/j.heliyon.2024.e35972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/18/2024] Open
Abstract
An organoid culture system better recapitulates the cellular structure, function, and interaction between cells and the extracellular matrix (ECM) than a two-dimensional (2D) culture system. We here constructed a condylar cartilage organoid to explore the regulatory role of primary cilia. Similar to the natural condylar cartilage, the condylar cartilage organoid exhibited abundant ECM and comprised superficial, proliferative, and hypertrophic zones. Primary cilia in the condylar cartilage organoid were shorter on average than those in the 2D culture chondrocytes, but their average length was equivalent to those in the natural condylar cartilage. Notably, primary cilia in each zone of the condylar cartilage organoid had an average length similar to that of primary cilia in the natural condylar cartilage. According to transcriptomic and biochemical analyses, the expression of cilia-related genes and cilia-related Hedgehog (HH) signaling differed between the condylar cartilage organoid and 2D culture systems. IFT88 knockdown promoted the protein levels of COL-Ⅹ, TRPV4, and HH signaling molecules in the condylar cartilage organoid, but decreased them in the 2D culture system. Notably, the protein levels of COL-Ⅹ, TRPV4, and HH signaling molecules increased in the superficial zone of the si IFT88 condylar cartilage organoid compared with the condylar cartilage organoid. However, the protein levels of aforementioned molecules were not significantly different in proliferative and hypertrophic zones. Collectively, we successfully constructed the condylar cartilage organoid with a better tissue structure and abundant ECM. Moreover, the condylar cartilage organoid is more suitable for exploring primary cilia functions.
Collapse
Affiliation(s)
- Zhan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, PR China
| | - Haoyu Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, PR China
| | - Qingwei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, PR China
| | - Tianhao Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, PR China
| | - Hanlin Tu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, PR China
| | - Guoliang Sa
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, PR China
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Xuewen Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, PR China
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Wuhan University, Wuhan, PR China
| |
Collapse
|
5
|
Yang C, Chen R, Chen C, Yang F, Xiao H, Geng B, Xia Y. Tissue engineering strategies hold promise for the repair of articular cartilage injury. Biomed Eng Online 2024; 23:92. [PMID: 39261876 PMCID: PMC11389311 DOI: 10.1186/s12938-024-01260-w] [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: 05/07/2024] [Accepted: 06/18/2024] [Indexed: 09/13/2024] Open
Abstract
Articular cartilage damage and wear can result in cartilage degeneration, ultimately culminating in osteoarthritis. Current surgical interventions offer limited capacity for cartilage tissue regeneration and offer only temporary alleviation of symptoms. Tissue engineering strategies are increasingly recognized as promising modalities for cartilage restoration. Currently, various biological scaffolds utilizing tissue engineering materials are extensively employed in both fundamental and clinical investigations of cartilage repair. In order to optimize the cartilage repair ability of tissue engineering scaffolds, researchers not only optimize the structure and properties of scaffolds from the perspective of materials science and manufacturing technology to enhance their histocompatibility, but also adopt strategies such as loading cells, cytokines, and drugs to promote cartilage formation. This review provides an overview of contemporary tissue engineering strategies employed in cartilage repair, as well as a synthesis of existing preclinical and clinical research. Furthermore, the obstacles faced in the translation of tissue engineering strategies to clinical practice are discussed, offering valuable guidance for researchers seeking to address these challenges.
Collapse
Affiliation(s)
- Chenhui Yang
- Department of Orthopedics, Lanzhou University Second Hospital, No.82, Cuyingmen, Chengguan District, Lanzhou, 730000, Gansu, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730000, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou, 730000, China
- Department of Orthopedic, Tianshui Hand and Foot Surgery Hospital, Tianshui, 741000, China
| | - Rongjin Chen
- Department of Orthopedics, Lanzhou University Second Hospital, No.82, Cuyingmen, Chengguan District, Lanzhou, 730000, Gansu, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730000, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou, 730000, China
| | - Changshun Chen
- Department of Orthopedics, Lanzhou University Second Hospital, No.82, Cuyingmen, Chengguan District, Lanzhou, 730000, Gansu, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730000, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou, 730000, China
| | - Fei Yang
- Department of Orthopedics, Lanzhou University Second Hospital, No.82, Cuyingmen, Chengguan District, Lanzhou, 730000, Gansu, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730000, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou, 730000, China
| | - Hefang Xiao
- Department of Orthopedics, Lanzhou University Second Hospital, No.82, Cuyingmen, Chengguan District, Lanzhou, 730000, Gansu, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730000, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou, 730000, China
| | - Bin Geng
- Department of Orthopedics, Lanzhou University Second Hospital, No.82, Cuyingmen, Chengguan District, Lanzhou, 730000, Gansu, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730000, China
- The Second School of Clinical Medical, Lanzhou University, Lanzhou, 730000, China
| | - Yayi Xia
- Department of Orthopedics, Lanzhou University Second Hospital, No.82, Cuyingmen, Chengguan District, Lanzhou, 730000, Gansu, China.
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- The Second School of Clinical Medical, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
6
|
Nordberg RC, Donahue RP, Espinosa MG, Salinas EY, Hu JC, Athanasiou KA. A cell bank paradigm for preclinical evaluation of an analogous cellular product for an allogeneic cell therapy. Biofabrication 2024; 16:10.1088/1758-5090/ad4de2. [PMID: 38768586 PMCID: PMC11534090 DOI: 10.1088/1758-5090/ad4de2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/20/2024] [Indexed: 05/22/2024]
Abstract
Toward the translation of allogeneic cell therapy products, cell banks are needed not only to manufacture the final human product but also during the preclinical evaluation of an animal-based analogous cellular product (ACP). These cell banks need to be established at both the master cell bank (MCB) level and the working cell bank (WCB) level. Inasmuch as most of the development of cell therapy products is at academic centers, it is imperative that academic researchers understand how to establish MCBs and WCBs within an academic environment. To illustrate this process, using articular cartilage as the model, a cell bank for an ACP was developed (MCBs at passage 2, WCBs at passage 5) to produce self-assembled neocartilage for preclinical evaluation (constructs at passage 7). The cell bank system is estimated to be able to produce between 160 000 and 400 000 constructs for each of the six MCBs. Overall, the ACP cell bank yielded constructs that are analogous to the intended human product, which is critical toward conducting preclinical evaluations of the ACP for inclusion in an Investigational New Drug application to the FDA.
Collapse
Affiliation(s)
- Rachel C. Nordberg
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, California 92617, USA
| | - Ryan P. Donahue
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, California 92617, USA
| | - M. Gabriela Espinosa
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, California 92617, USA
- Concordia University Irvine, 1530 Concordia West, Irvine, CA, USA 92612
| | - Evelia Y. Salinas
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, California 92617, USA
| | - Jerry C. Hu
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, California 92617, USA
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, California 92617, USA
| |
Collapse
|
7
|
Nordberg RC, Bielajew BJ, Takahashi T, Dai S, Hu JC, Athanasiou KA. Recent advancements in cartilage tissue engineering innovation and translation. Nat Rev Rheumatol 2024; 20:323-346. [PMID: 38740860 PMCID: PMC11524031 DOI: 10.1038/s41584-024-01118-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 05/16/2024]
Abstract
Articular cartilage was expected to be one of the first successfully engineered tissues, but today, cartilage repair products are few and they exhibit considerable limitations. For example, of the cell-based products that are available globally, only one is marketed for non-knee indications, none are indicated for severe osteoarthritis or rheumatoid arthritis, and only one is approved for marketing in the USA. However, advances in cartilage tissue engineering might now finally lead to the development of new cartilage repair products. To understand the potential in this field, it helps to consider the current landscape of tissue-engineered products for articular cartilage repair and particularly cell-based therapies. Advances relating to cell sources, bioactive stimuli and scaffold or scaffold-free approaches should now contribute to progress in therapeutic development. Engineering for an inflammatory environment is required because of the need for implants to withstand immune challenge within joints affected by osteoarthritis or rheumatoid arthritis. Bringing additional cartilage repair products to the market will require an understanding of the translational vector for their commercialization. Advances thus far can facilitate the future translation of engineered cartilage products to benefit the millions of patients who suffer from cartilage injuries and arthritides.
Collapse
Affiliation(s)
- Rachel C Nordberg
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Benjamin J Bielajew
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Takumi Takahashi
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Shuyan Dai
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
8
|
Manescu (Paltanea) V, Paltanea G, Antoniac A, Gruionu LG, Robu A, Vasilescu M, Laptoiu SA, Bita AI, Popa GM, Cocosila AL, Silviu V, Porumb A. Mechanical and Computational Fluid Dynamic Models for Magnesium-Based Implants. MATERIALS (BASEL, SWITZERLAND) 2024; 17:830. [PMID: 38399081 PMCID: PMC10890492 DOI: 10.3390/ma17040830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/26/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024]
Abstract
Today, mechanical properties and fluid flow dynamic analysis are considered to be two of the most important steps in implant design for bone tissue engineering. The mechanical behavior is characterized by Young's modulus, which must have a value close to that of the human bone, while from the fluid dynamics point of view, the implant permeability and wall shear stress are two parameters directly linked to cell growth, adhesion, and proliferation. In this study, we proposed two simple geometries with a three-dimensional pore network dedicated to a manufacturing route based on a titanium wire waving procedure used as an intermediary step for Mg-based implant fabrication. Implant deformation under different static loads, von Mises stresses, and safety factors were investigated using finite element analysis. The implant permeability was computed based on Darcy's law following computational fluid dynamic simulations and, based on the pressure drop, was numerically estimated. It was concluded that both models exhibited a permeability close to the human trabecular bone and reduced wall shear stresses within the biological range. As a general finding, the proposed geometries could be useful in orthopedics for bone defect treatment based on numerical analyses because they mimic the trabecular bone properties.
Collapse
Affiliation(s)
- Veronica Manescu (Paltanea)
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania;
| | - Gheorghe Paltanea
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania;
| | - Aurora Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Lucian Gheorghe Gruionu
- Faculty of Mechanics, University of Craiova, 13 Alexandru Ioan Cuza, RO-200585 Craiova, Romania;
| | - Alina Robu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Marius Vasilescu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Stefan Alexandru Laptoiu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Ana Iulia Bita
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Georgiana Maria Popa
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania; (G.M.P.); (A.L.C.); (V.S.)
| | - Andreea Liliana Cocosila
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania; (G.M.P.); (A.L.C.); (V.S.)
| | - Vlad Silviu
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania; (G.M.P.); (A.L.C.); (V.S.)
| | - Anca Porumb
- Department of Dental Medicine, Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania;
| |
Collapse
|
9
|
Jia Y, Le H, Wang X, Zhang J, Liu Y, Ding J, Zheng C, Chang F. Double-edged role of mechanical stimuli and underlying mechanisms in cartilage tissue engineering. Front Bioeng Biotechnol 2023; 11:1271762. [PMID: 38053849 PMCID: PMC10694366 DOI: 10.3389/fbioe.2023.1271762] [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: 08/02/2023] [Accepted: 10/11/2023] [Indexed: 12/07/2023] Open
Abstract
Mechanical stimuli regulate the chondrogenic differentiation of mesenchymal stem cells and the homeostasis of chondrocytes, thus affecting implant success in cartilage tissue engineering. The mechanical microenvironment plays fundamental roles in the maturation and maintenance of natural articular cartilage, and the progression of osteoarthritis Hence, cartilage tissue engineering attempts to mimic this environment in vivo to obtain implants that enable a superior regeneration process. However, the specific type of mechanical loading, its optimal regime, and the underlying molecular mechanisms are still under investigation. First, this review delineates the composition and structure of articular cartilage, indicating that the morphology of chondrocytes and components of the extracellular matrix differ from each other to resist forces in three top-to-bottom overlapping zones. Moreover, results from research experiments and clinical trials focusing on the effect of compression, fluid shear stress, hydrostatic pressure, and osmotic pressure are presented and critically evaluated. As a key direction, the latest advances in mechanisms involved in the transduction of external mechanical signals into biological signals are discussed. These mechanical signals are sensed by receptors in the cell membrane, such as primary cilia, integrins, and ion channels, which next activate downstream pathways. Finally, biomaterials with various modifications to mimic the mechanical properties of natural cartilage and the self-designed bioreactors for experiment in vitro are outlined. An improved understanding of biomechanically driven cartilage tissue engineering and the underlying mechanisms is expected to lead to efficient articular cartilage repair for cartilage degeneration and disease.
Collapse
Affiliation(s)
- Yao Jia
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
- The Second Bethune Clinical Medical College of Jilin University, Jilin, China
| | - Hanxiang Le
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
- The Fourth Treatment Area of Trauma Hip Joint Surgery Department, Tianjin Hospital, Tianjin, China
| | - Xianggang Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| | - Jiaxin Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| | - Yan Liu
- The Second Bethune Clinical Medical College of Jilin University, Jilin, China
| | - Jiacheng Ding
- The Second Bethune Clinical Medical College of Jilin University, Jilin, China
| | - Changjun Zheng
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| | - Fei Chang
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| |
Collapse
|
10
|
Stampoultzis T, Guo Y, Nasrollahzadeh N, Rana VK, Karami P, Pioletti DP. Low-oxygen tension augments chondrocyte sensitivity to biomimetic thermomechanical cues in cartilage-engineered constructs. iScience 2023; 26:107491. [PMID: 37599834 PMCID: PMC10432199 DOI: 10.1016/j.isci.2023.107491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/14/2023] [Accepted: 07/23/2023] [Indexed: 08/22/2023] Open
Abstract
Chondrocytes respond to various biophysical cues, including oxygen tension, transient thermal signals, and mechanical stimuli. However, understanding how these factors interact to establish a unique regulatory microenvironment for chondrocyte function remains unclear. Herein, we explore these interactions using a joint-simulating bioreactor that independently controls the culture's oxygen concentration, evolution of temperature, and mechanical loading. Our analysis revealed significant coupling between these signals, resulting in a remarkable ∼14-fold increase in collagen type II (COL2a) and aggrecan (ACAN) mRNA expression. Furthermore, dynamic thermomechanical stimulation enhanced glycosaminoglycan and COL2a protein synthesis, with the magnitude of the biosynthetic changes being oxygen dependent. Additionally, our mechanistic study highlighted the crucial role of SRY-box transcription factor 9 (SOX9) as a major regulator of chondrogenic response, specifically expressed in response to combined biophysical signals. These findings illuminate the integration of various mechanobiological cues by chondrocytes and provide valuable insights for improving the extracellular matrix content in cartilage-engineered constructs.
Collapse
Affiliation(s)
- Theofanis Stampoultzis
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Yanheng Guo
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Naser Nasrollahzadeh
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Vijay Kumar Rana
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Peyman Karami
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Dominique P. Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| |
Collapse
|
11
|
Zhang Y, Tawiah GK, Wu X, Zhang Y, Wang X, Wei X, Qiao X, Zhang Q. Primary cilium-mediated mechanotransduction in cartilage chondrocytes. Exp Biol Med (Maywood) 2023; 248:1279-1287. [PMID: 37897221 PMCID: PMC10625344 DOI: 10.1177/15353702231199079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023] Open
Abstract
Osteoarthritis (OA) is one of the most prevalent joint disorders associated with the degradation of articular cartilage and an abnormal mechanical microenvironment. Mechanical stimuli, including compression, shear stress, stretching strain, osmotic challenge, and the physical properties of the matrix microenvironment, play pivotal roles in the tissue homeostasis of articular cartilage. The primary cilium, as a mechanosensory and chemosensory organelle, is important for detecting and transmitting both mechanical and biochemical signals in chondrocytes within the matrix microenvironment. Growing evidence indicates that primary cilia are critical for chondrocytes signaling transduction and the matrix homeostasis of articular cartilage. Furthermore, the ability of primary cilium to regulate cellular signaling is dynamic and dependent on the cellular matrix microenvironment. In the current review, we aim to elucidate the key mechanisms by which primary cilia mediate chondrocytes sensing and responding to the matrix mechanical microenvironment. This might have potential therapeutic applications in injuries and OA-associated degeneration of articular cartilage.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Histology and Embryology, Shanxi Medical University, Jinzhong 030604, Shanxi, China
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Godfred K Tawiah
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Xiaoan Wu
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Yanjun Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Xiaohu Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaochun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaohong Qiao
- Department of Histology and Embryology, Shanxi Medical University, Jinzhong 030604, Shanxi, China
- Department of Orthopaedics, Lvliang Hospital Affiliated to Shanxi Medical University, Lvliang 033099, Shanxi, China
| | - Quanyou Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| |
Collapse
|
12
|
Chen YW, Lin YH, Lin TL, Lee KXA, Yu MH, Shie MY. 3D-biofabricated chondrocyte-laden decellularized extracellular matrix-contained gelatin methacrylate auxetic scaffolds under cyclic tensile stimulation for cartilage regeneration. Biofabrication 2023; 15:045007. [PMID: 37429300 DOI: 10.1088/1758-5090/ace5e1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/10/2023] [Indexed: 07/12/2023]
Abstract
Three-dimensional (3D) hydrogel constructs can mimic features of the extracellular matrix (ECM) and have tailorable physicochemical properties to support and maintain the regeneration of articular cartilage. Various studies have shown that mechanical cues affect the cellular microenvironment and thereby influence cellular behavior. In this study, we fabricated an auxetic scaffold to investigate the effect of 3D tensile stimulation on chondrocyte behavior. Different concentrations of decellularized extracellular matrix (dECM) were mixed with fish gelatin methacrylate (FGelMa) and employed for the preparation of dECM/FGelMa auxetic bio-scaffolds using 3D biofabrication technology. We show that when human chondrocytes (HCs) were incorporated into these scaffolds, their proliferation and the expression of chondrogenesis-related markers increased with dECM content. The function of HC was influenced by cyclic tensile stimulation, as shown by increased production of the chondrogenesis-related markers, collagen II and glycosaminoglycans, with the involvement of the yes-associated protein 1 signaling pathway. The biofabricated auxetic scaffold represents an excellent platform for exploring interactions between cells and their mechanical microenvironment.
Collapse
Affiliation(s)
- Yi-Wen Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung 404332, Taiwan
- High Performance Materials Institute for x-Dimensional Printing, Asia University, Taichung City 41354, Taiwan
| | - Yen-Hong Lin
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung 404332, Taiwan
| | - Tsung-Li Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Department of Orthopedics, China Medical University Hospital, Taichung 404332, Taiwan
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung 406040, Taiwan
| | - Kai-Xing Alvin Lee
- Department of Orthopedics, China Medical University Hospital, Taichung 404332, Taiwan
| | - Min-Hua Yu
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung 404332, Taiwan
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung 406040, Taiwan
| | - Ming-You Shie
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung 404332, Taiwan
- Department of Biomedical Engineering, China Medical University, Taichung 406040, Taiwan
- School of Dentistry, China Medical University, Taichung 406040, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
13
|
Niu X, Xu Z, Di M, Huang D, Li X. Bioreactor strategies for tissue-engineered osteochondral constructs: Advantages, present situations and future trends. COMPOSITES PART B: ENGINEERING 2023; 259:110736. [DOI: 10.1016/j.compositesb.2023.110736] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
|
14
|
Lamandé SR, Ng ES, Cameron TL, Kung LHW, Sampurno L, Rowley L, Lilianty J, Patria YN, Stenta T, Hanssen E, Bell KM, Saxena R, Stok KS, Stanley EG, Elefanty AG, Bateman JF. Modeling human skeletal development using human pluripotent stem cells. Proc Natl Acad Sci U S A 2023; 120:e2211510120. [PMID: 37126720 PMCID: PMC10175848 DOI: 10.1073/pnas.2211510120] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 04/04/2023] [Indexed: 05/03/2023] Open
Abstract
Chondrocytes and osteoblasts differentiated from induced pluripotent stem cells (iPSCs) will provide insights into skeletal development and genetic skeletal disorders and will generate cells for regenerative medicine applications. Here, we describe a method that directs iPSC-derived sclerotome to chondroprogenitors in 3D pellet culture then to articular chondrocytes or, alternatively, along the growth plate cartilage pathway to become hypertrophic chondrocytes that can transition to osteoblasts. Osteogenic organoids deposit and mineralize a collagen I extracellular matrix (ECM), mirroring in vivo endochondral bone formation. We have identified gene expression signatures at key developmental stages including chondrocyte maturation, hypertrophy, and transition to osteoblasts and show that this system can be used to model genetic cartilage and bone disorders.
Collapse
Affiliation(s)
- Shireen R. Lamandé
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Elizabeth S. Ng
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Trevor L. Cameron
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Louise H. W. Kung
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Lisa Sampurno
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Lynn Rowley
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Jinia Lilianty
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Yudha Nur Patria
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
- Department of Child Health, Universitas Gadjah Mada, Yogyakarta55281, Indonesia
| | - Tayla Stenta
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Eric Hanssen
- Ian Holmes Imaging Center and Department of Biochemistry and Pharmacology, Bio21 Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | - Katrina M. Bell
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Ritika Saxena
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Kathryn S. Stok
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
| | - Edouard G. Stanley
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Andrew G. Elefanty
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - John F. Bateman
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
15
|
Zhou Z, Wang W, Wang J, Wang H, Xia Y, Zhang W, Lai Y, Lin X, Huang Y, Zou X, Stoddart MJ, Li Z, Tian W, Liu S, Wu X, Gao M, Li J, Yang L, Chen D. Function-oriented design: A novel strategy for advanced biomedical materials. JOURNAL OF MATERIALS SCIENCE & TECHNOLOGY 2023; 145:197-209. [DOI: 10.1016/j.jmst.2022.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
|
16
|
Li J, Wang X, Li X, Liu D, Zhai L, Wang X, Kang R, Yokota H, Yang L, Zhang P. Mechanical Loading Promotes the Migration of Endogenous Stem Cells and Chondrogenic Differentiation in a Mouse Model of Osteoarthritis. Calcif Tissue Int 2023; 112:363-376. [PMID: 36566445 DOI: 10.1007/s00223-022-01052-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/15/2022] [Indexed: 12/26/2022]
Abstract
Osteoarthritis (OA) is a major health problem, characterized by progressive cartilage degeneration. Previous works have shown that mechanical loading can alleviate OA symptoms by suppressing catabolic activities. This study evaluated whether mechanical loading can enhance anabolic activities by facilitating the recruitment of stem cells for chondrogenesis. We evaluated cartilage degradation in a mouse model of OA through histology with H&E and safranin O staining. We also evaluated the migration and chondrogenic ability of stem cells using in vitro assays, including immunohistochemistry, immunofluorescence, and Western blot analysis. The result showed that the OA mice that received mechanical loading exhibited resilience to cartilage damage. Compared to the OA group, mechanical loading promoted the expression of Piezo1 and the migration of stem cells was promoted via the SDF-1/CXCR4 axis. Also, the chondrogenic differentiation was enhanced by the upregulation of SOX9, a transcription factor important for chondrogenesis. Collectively, the results revealed that mechanical loading facilitated cartilage repair by promoting the migration and chondrogenic differentiation of endogenous stem cells. This study provided new insights into the loading-driven engagement of endogenous stem cells and the enhancement of anabolic responses for the treatment of OA.
Collapse
Affiliation(s)
- Jie Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaoyu Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China
| | - Daquan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China
| | - Lidong Zhai
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Xuetong Wang
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ran Kang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Lei Yang
- Center for Health Sciences and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300131, China
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China.
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
17
|
Stampoultzis T, Guo Y, Nasrollahzadeh N, Karami P, Pioletti DP. Mimicking Loading-Induced Cartilage Self-Heating in Vitro Promotes Matrix Formation in Chondrocyte-Laden Constructs with Different Mechanical Properties. ACS Biomater Sci Eng 2023; 9:651-661. [PMID: 36625682 PMCID: PMC9930743 DOI: 10.1021/acsbiomaterials.2c00723] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023]
Abstract
Articular cartilage presents a mechanically sensitive tissue. Chondrocytes, the sole cell type residing in the tissue, perceive and react to physical cues as signals that significantly modulate their behavior. Hyaline cartilage is a connective tissue with high dissipative capabilities, able to increase its temperature during daily activities, thus providing a dynamic thermal milieu for the residing chondrocytes. This condition, self-heating, which is still chiefly ignored among the scientific community, adds a new thermal dimension in cartilage mechanobiology. Motivated by the lack of studies exploring this dynamic temperature increase as a potential stimulus in cartilage-engineered constructs, we aimed to elucidate whether loading-induced evolved temperature serves as an independent or complementary regulatory cue for chondrocyte function. In particular, we evaluated the chondrocytes' response to thermal and/or mechanical stimulation in two types of scaffolds exhibiting dissipation levels close to healthy and degenerated articular cartilage. It was found, in both scaffold groups, that the combination of dynamic thermal and mechanical stimuli induced superior effects in the expression of major chondrogenic genes, such as SOX9 and LOXL2, compared to either signal alone. Similar effects were also observed in proteoglycan accumulation over time, along with increased mRNA transcription and synthesis of TRPV4, and for the first time demonstrated in chondrocytes, TREK1 ion channels. Conversely, the chondrogenic response of cells to isolated thermal or mechanical cues was generally scaffold-type dependent. Nonetheless, the significance of thermal stimulus as a chondro-inductive signal was better supported in both studied groups. Our data indicates that the temperature evolution is necessary for chondrocytes to more effectively perceive and translate applied mechanical loading.
Collapse
Affiliation(s)
- Theofanis Stampoultzis
- Laboratory
of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne 1015, Switzerland
| | - Yanheng Guo
- Laboratory
of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne 1015, Switzerland
| | - Naser Nasrollahzadeh
- Laboratory
of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne 1015, Switzerland
| | - Peyman Karami
- Laboratory
of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne 1015, Switzerland
| | - Dominique P. Pioletti
- Laboratory
of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne 1015, Switzerland
| |
Collapse
|
18
|
Dehghan-Baniani D, Mehrjou B, Chu PK, Lee WYW, Wu H. Recent Advances in "Functional Engineering of Articular Cartilage Zones by Polymeric Biomaterials Mediated with Physical, Mechanical, and Biological/Chemical Cues". Adv Healthc Mater 2022; 12:e2202581. [PMID: 36571465 DOI: 10.1002/adhm.202202581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/19/2022] [Indexed: 12/27/2022]
Abstract
Articular cartilage (AC) plays an unquestionable role in joint movements but unfortunately the healing capacity is restricted due to its avascular and acellular nature. While cartilage tissue engineering has been lifesaving, it is very challenging to remodel the complex cartilage composition and architecture with gradient physio-mechanical properties vital to proper tissue functions. To address these issues, a better understanding of the intrinsic AC properties and how cells respond to stimuli from the external microenvironment must be better understood. This is essential in order to take one step closer to producing functional cartilaginous constructs for clinical use. Recently, biopolymers have aroused much attention due to their versatility, processability, and flexibility because the properties can be tailored to match the requirements of AC. This review highlights polymeric scaffolds developed in the past decade for reconstruction of zonal AC layers including the superficial zone, middle zone, and deep zone by means of exogenous stimuli such as physical, mechanical, and biological/chemical signals. The mimicked properties are reviewed in terms of the biochemical composition and organization, cell fate (morphology, orientation, and differentiation), as well as mechanical properties and finally, the challenges and potential ways to tackle them are discussed.
Collapse
Affiliation(s)
- Dorsa Dehghan-Baniani
- Department of Chemical and Biological Engineering Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.,Musculoskeletal Research Laboratory, SH Ho Scoliosis Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Babak Mehrjou
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Paul K Chu
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Wayne Yuk Wai Lee
- Musculoskeletal Research Laboratory, SH Ho Scoliosis Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China.,Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong SAR, China.,Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong SAR, China
| | - Hongkai Wu
- Department of Chemical and Biological Engineering Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.,Department of Chemistry and the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong SAR, China
| |
Collapse
|
19
|
Otarola GA, Hu JC, Athanasiou KA. Ion modulatory treatments toward functional self-assembled neocartilage. Acta Biomater 2022; 153:85-96. [PMID: 36113725 PMCID: PMC11575480 DOI: 10.1016/j.actbio.2022.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022]
Abstract
Signals that recapitulate in vitro the conditions found in vivo, such as hypoxia or mechanical forces, contribute to the generation of tissue-engineered hyaline-like tissues. The cell regulatory processes behind hypoxic and mechanical stimuli rely on ion concentration; iron is required to degrade the hypoxia inducible factor 1a (HIF1α) under normoxia, whereas the initiation of mechanotransduction requires the cytoplasmic increase of calcium concentration. In this work, we propose that ion modulation can be used to improve the biomechanical properties of self-assembled neocartilage constructs derived from rejuvenated expanded minipig rib chondrocytes. The objectives of this work were 1) to determine the effects of iron sequestration on self-assembled neocartilage constructs using two doses of the iron chelator deferoxamine (DFO), and 2) to evaluate the performance of the combined treatment of DFO and ionomycin, a calcium ionophore that triggers cytoplasmic calcium accumulation. This study employed a two-phase approach. In Phase I, constructs treated with a high dose of DFO (100 µM) exhibited an 87% increase in pyridinoline crosslinks, a 57% increase in the Young's modulus, and a 112% increase in the ultimate tensile strength (UTS) of the neotissue. In Phase II, the combined use of both ion modulators resulted in 150% and 176% significant increases in the Young's modulus and UTS of neocartilage constructs, respectively; for the first time, neocartilage constructs achieved a Young's modulus of 11.76±3.29 MPa and UTS of 4.20±1.24 MPa. The results of this work provide evidence that ion modulation can be employed to improve the biomechanical properties in engineered neotissues. STATEMENT OF SIGNIFICANCE: The translation of tissue-engineered products requires the development of strategies capable of producing biomimetic neotissues in a replicable, controllable, and cost-effective manner. Among other functions, Fe2+ and Ca2+ are involved in the control of the hypoxic response and mechanotransduction, respectively. Both stimuli, hypoxia and mechanical forces, are known to favor chondrogenesis. This study utilized ion modulators to improve the mechanical properties self-assembled neocartilage constructs derived from expanded and rejuvenated costal chondrocytes via Fe2+ sequestration and Ca2+ influx, alone or in combination. The results indicate that ion modulation induced tissue maturation and a significant improvement of the mechanical properties, and holds potential as a tool to mitigate the need for bioreactors and engineer hyaline-like tissues.
Collapse
Affiliation(s)
- Gaston A Otarola
- 3131 Engineering Hall, Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
| | - Jerry C Hu
- 3131 Engineering Hall, Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
| | - Kyriacos A Athanasiou
- 3131 Engineering Hall, Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA.
| |
Collapse
|
20
|
Nordberg RC, Kim AN, Hight JM, Meka RS, Elder BD, Hu JC, Athanasiou KA. Biochemical and biomechanical characterization of the cervical, thoracic, and lumbar facet joint cartilage in the Yucatan minipig. J Biomech 2022; 142:111238. [PMID: 35933954 PMCID: PMC9910803 DOI: 10.1016/j.jbiomech.2022.111238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/24/2022]
Abstract
Facet joint arthrosis causes pain in approximately 7 % of the U.S. population, but current treatments are palliative. The objective of this study was to elucidate structure-function relationships and aid in the development of future treatments for the facet joint. This study characterized the articular surfaces of cervical, thoracic, and lumbar facet cartilage from skeletally mature (18-24 mo) Yucatan minipigs. The minipig was selected as the animal model because it is recognized by the U.S. Food and Drug Administration (FDA) and the American Society for Testing and Materials (ASTM) as a translationally relevant model for spine-related indications. It was found that the thoracic facets had a ∼2 times higher aspect ratio than lumbar and cervical facets. Lumbar facets had 6.9-9.6 times higher % depth than the cervical and thoracic facets. Aggregate modulus values ranged from 135 to 262 kPa, much lower than reported aggregate modulus in the human knee (reported to be 530-701 kPa). The tensile Young's modulus values ranged from 6.7 to 20.3 MPa, with the lumbar superior facet being 304 % and 286 % higher than the cervical inferior and thoracic superior facets, respectively. Moreover, 3D reconstructions of entire vertebral segments were generated. The results of this study imply that structure-function relationships in the facet cartilage are different from other joint cartilages because biochemical properties are analogous to other articular cartilage sources whereas mechanical properties are not. By providing functional properties and a 3D database of minipig facet geometries, this work may supply design criteria for future facet tissue engineering efforts.
Collapse
Affiliation(s)
- Rachel C Nordberg
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, CA 92617, USA
| | - Andrew N Kim
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, CA 92617, USA
| | - Justin M Hight
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, CA 92617, USA
| | - Rithika S Meka
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, CA 92617, USA
| | - Benjamin D Elder
- Department of Neurosurgery, Orthopedics, and Biomedical Engineering, Mayo Clinic School of Medicine, 200 1st St. SW, Rochester, MN 55905, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, CA 92617, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, 3131 Engineering Hall, University of California, Irvine, CA 92617, USA.
| |
Collapse
|
21
|
Omar AM, Hassan MH, Daskalakis E, Ates G, Bright CJ, Xu Z, Powell EJ, Mirihanage W, Bartolo PJDS. Geometry-Based Computational Fluid Dynamic Model for Predicting the Biological Behavior of Bone Tissue Engineering Scaffolds. J Funct Biomater 2022; 13:104. [PMID: 35997442 PMCID: PMC9397055 DOI: 10.3390/jfb13030104] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 02/05/2023] Open
Abstract
The use of biocompatible and biodegradable porous scaffolds produced via additive manufacturing is one of the most common approaches in tissue engineering. The geometric design of tissue engineering scaffolds (e.g., pore size, pore shape, and pore distribution) has a significant impact on their biological behavior. Fluid flow dynamics are important for understanding blood flow through a porous structure, as they determine the transport of nutrients and oxygen to cells and the flushing of toxic waste. The aim of this study is to investigate the impact of the scaffold architecture, pore size and distribution on its biological performance using Computational Fluid Dynamics (CFD). Different blood flow velocities (BFV) induce wall shear stresses (WSS) on cells. WSS values above 30 mPa are detrimental to their growth. In this study, two scaffold designs were considered: rectangular scaffolds with uniform square pores (300, 350, and 450 µm), and anatomically designed circular scaffolds with a bone-like structure and pore size gradient (476-979 µm). The anatomically designed scaffolds provided the best fluid flow conditions, suggesting a 24.21% improvement in the biological performance compared to the rectangular scaffolds. The numerical observations are aligned with those of previously reported biological studies.
Collapse
Affiliation(s)
- Abdalla M. Omar
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (M.H.H.); (E.D.); (G.A.); (C.J.B.); (Z.X.); (E.J.P.)
| | - Mohamed H. Hassan
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (M.H.H.); (E.D.); (G.A.); (C.J.B.); (Z.X.); (E.J.P.)
| | - Evangelos Daskalakis
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (M.H.H.); (E.D.); (G.A.); (C.J.B.); (Z.X.); (E.J.P.)
| | - Gokhan Ates
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (M.H.H.); (E.D.); (G.A.); (C.J.B.); (Z.X.); (E.J.P.)
| | - Charlie J. Bright
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (M.H.H.); (E.D.); (G.A.); (C.J.B.); (Z.X.); (E.J.P.)
| | - Zhanyan Xu
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (M.H.H.); (E.D.); (G.A.); (C.J.B.); (Z.X.); (E.J.P.)
| | - Emily J. Powell
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (M.H.H.); (E.D.); (G.A.); (C.J.B.); (Z.X.); (E.J.P.)
| | - Wajira Mirihanage
- Department of Materials, The University of Manchester, Manchester M13 9PL, UK;
| | - Paulo J. D. S. Bartolo
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester M13 9PL, UK; (M.H.H.); (E.D.); (G.A.); (C.J.B.); (Z.X.); (E.J.P.)
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|
22
|
Salinas EY, Donahue RP, Herrera JM, Hu JC, Athanasiou KA. The functionality and translatability of neocartilage constructs are improved with the combination of fluid-induced shear stress and bioactive factors. FASEB J 2022; 36:e22225. [PMID: 35224777 PMCID: PMC9045489 DOI: 10.1096/fj.202101699r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 11/11/2022]
Abstract
Neocartilage tissue engineering aims to address the shortcomings of current clinical treatments for articular cartilage indications. However, advancement is required toward neocartilage functionality (mechanical and biochemical properties) and translatability (construct size, gross morphology, passage number, cell source, and cell type). Using fluid-induced shear (FIS) stress, a potent mechanical stimulus, over four phases, this work investigates FIS stress' efficacy toward creating large neocartilage derived from highly passaged minipig costal chondrocytes, a species relevant to the preclinical regulatory process. In Phase I, FIS stress application timing was investigated in bovine articular chondrocytes and found to improve the aggregate modulus of neocartilage by 151% over unstimulated controls when stimulated during the maturation stage. In Phase II, FIS stress stimulation was translated from bovine articular chondrocytes to expanded minipig costal chondrocytes, yielding a 46% improvement in aggregate modulus over nonstimulated controls. In Phase III, bioactive factors were combined with FIS stress to improve the shear modulus by 115% over bioactive factor-only controls. The translatability of neocartilage was improved in Phase IV by utilizing highly passaged cells to form constructs more than 9-times larger in the area (11 × 17 mm), yielding an improved aggregate modulus by 134% and a flat morphology compared to free-floating, bioactive factor-only controls. Overall, this study represents a significant step toward generating mechanically robust, large constructs necessary for animal studies, and eventually, human clinical studies.
Collapse
Affiliation(s)
- Evelia Y Salinas
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA
| | - Ryan P Donahue
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA
| | - Jessica M Herrera
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
23
|
Alexandrovskaya Y, Baum O, Sovetsky A, Matveyev A, Matveev L, Sobol E, Zaitsev V. Optical Coherence Elastography as a Tool for Studying Deformations in Biomaterials: Spatially-Resolved Osmotic Strain Dynamics in Cartilaginous Samples. MATERIALS (BASEL, SWITZERLAND) 2022; 15:904. [PMID: 35160851 PMCID: PMC8838169 DOI: 10.3390/ma15030904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/14/2022] [Accepted: 01/20/2022] [Indexed: 12/15/2022]
Abstract
This paper presents a recently developed variant of phase-resolved Optical Coherence Elastography (OCE) enabling non-contact visualization of transient local strains of various origins in biological tissues and other materials. In this work, we demonstrate the possibilities of this new technique for studying dynamics of osmotically-induced strains in cartilaginous tissue impregnated with optical clearing agents (OCA). For poroelastic water-containing biological tissues, application of non-isotonic OCAs, various contrast additives, as well as drug solutions administration, may excite transient spatially-inhomogeneous strain fields of high magnitude in the tissue bulk, initiating mechanical and structural alterations. The range of the strain reliably observed by OCE varied from ±10-3 to ±0.4 for diluted and pure glycerol, correspondingly. The OCE-technique used made it possible to reveal previously inaccessible details of the complex spatio-temporal evolution of alternating-sign osmotic strains at the initial stages of agent diffusion. Qualitatively different effects produced by particular hydrophilic OCAs, such as glycerol and iohexol, are discussed, as well as concentration-dependent differences. Overall, the work demonstrates the unique abilities of the new OCE-modality in providing a deeper insight in real-time kinetics of osmotically-induced strains relevant to a broad range of biomedical applications.
Collapse
Affiliation(s)
- Yulia Alexandrovskaya
- Institute of Photon Technologies, Federal Scientific Research Center “Crystallography and Photonics”, Russian Academy of Sciences, 2 Pionerskaya Street, Troitsk, 108840 Moscow, Russia;
| | - Olga Baum
- Institute of Photon Technologies, Federal Scientific Research Center “Crystallography and Photonics”, Russian Academy of Sciences, 2 Pionerskaya Street, Troitsk, 108840 Moscow, Russia;
| | - Alexander Sovetsky
- Institute of Applied Physics of the Russian Academy of Sciences, 46 Uljanova Street, 603950 Nizhny Novgorod, Russia; (A.S.); (A.M.); (L.M.); (V.Z.)
| | - Alexander Matveyev
- Institute of Applied Physics of the Russian Academy of Sciences, 46 Uljanova Street, 603950 Nizhny Novgorod, Russia; (A.S.); (A.M.); (L.M.); (V.Z.)
| | - Lev Matveev
- Institute of Applied Physics of the Russian Academy of Sciences, 46 Uljanova Street, 603950 Nizhny Novgorod, Russia; (A.S.); (A.M.); (L.M.); (V.Z.)
| | - Emil Sobol
- UCI Health Beckman Laser Institute & Medical Clinic, 1002 Health Sciences Rd., Irvine, CA 92612, USA;
| | - Vladimir Zaitsev
- Institute of Applied Physics of the Russian Academy of Sciences, 46 Uljanova Street, 603950 Nizhny Novgorod, Russia; (A.S.); (A.M.); (L.M.); (V.Z.)
| |
Collapse
|
24
|
Otarola G, Hu JC, Athanasiou KA. INTRACELLULAR CALCIUM AND SODIUM MODULATION OF SELF-ASSEMBLED NEOCARTILAGE USING COSTAL CHONDROCYTES. Tissue Eng Part A 2021; 28:595-605. [PMID: 34877888 DOI: 10.1089/ten.tea.2021.0169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ion signaling via Ca2+ and Na+ plays a key role in mechanotransduction and encourages a chondrogenic phenotype and tissue maturation. Here, we propose that the pleiotropic effects of Ca2+ and Na+ modulation can be used to induce maturation and improvement of neocartilage derived from re-differentiated expanded chondrocytes from minipig rib cartilage. Three ion modulators were employed: 1) 4α-phorbol-12,13-didecanoate (4-αPDD), an agonist of the Ca2+-permeable transient receptor potential vanilloid 4 (TRPV4), 2) ouabain, an inhibitor of the Na+/K+ pump, and 3) ionomycin, a Ca2+ ionophore. These ion modulators were used individually or in combination. While no beneficial effects were observed when using combinations of the ion modulators, single treatment of constructs with the three ion modulators resulted in multiple effects in structure-function relationships. The most significant findings were related to ionomycin. Treatment of neocartilage with ionomycin produced 61% and 115% increases in glycosaminoglycan and pyridinoline crosslink content, respectively, compared to the control. Moreover, treatment with this Ca2+ ionophore resulted in a 45% increase of the aggregate modulus, and a 63% increase in the tensile Young's modulus, resulting in aggregate and Young's moduli of 567 kPa and 8.43 MPa, respectively. These results support the use of ion modulation to develop biomimetic neocartilage using expanded re-differentiated costal chondrocytes.
Collapse
Affiliation(s)
- Gaston Otarola
- University of California, Irvine, BME, Irvine, California, United States;
| | - Jerry C Hu
- University of California, Irvine, BME, Irvine, California, United States;
| | | |
Collapse
|
25
|
Stampoultzis T, Karami P, Pioletti DP. Thoughts on cartilage tissue engineering: A 21st century perspective. Curr Res Transl Med 2021; 69:103299. [PMID: 34192658 DOI: 10.1016/j.retram.2021.103299] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/11/2021] [Accepted: 05/26/2021] [Indexed: 12/15/2022]
Abstract
In mature individuals, hyaline cartilage demonstrates a poor intrinsic capacity for repair, thus even minor defects could result in progressive degeneration, impeding quality of life. Although numerous attempts have been made over the past years for the advancement of effective treatments, significant challenges still remain regarding the translation of in vitro cartilage engineering strategies from bench to bedside. This paper reviews the latest concepts on engineering cartilage tissue in view of biomaterial scaffolds, tissue biofabrication, mechanobiology, as well as preclinical studies in different animal models. The current work is not meant to provide a methodical review, rather a perspective of where the field is currently focusing and what are the requirements for bridging the gap between laboratory-based research and clinical applications, in light of the current state-of-the-art literature. While remarkable progress has been accomplished over the last 20 years, the current sophisticated strategies have reached their limit to further enhance healthcare outcomes. Considering a clinical aspect together with expertise in mechanobiology, biomaterial science and biofabrication methods, will aid to deal with the current challenges and will present a milestone for the furtherance of functional cartilage engineering.
Collapse
Affiliation(s)
| | - Peyman Karami
- Laboratory of Biomechanical Orthopedics, EPFL, Lausanne, Switzerland.
| | | |
Collapse
|