1
|
Wang T, Maldonado CC, Huang BL, Budbazar E, Martin A, Layne MD, Murphy-Ullrich JE, Grinstaff MW, Albro MB. A Bio-inspired Latent TGF-β Conjugated Scaffold Improves Neocartilage Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636279. [PMID: 39975171 PMCID: PMC11838498 DOI: 10.1101/2025.02.03.636279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
In cartilage tissue engineering, active TGF-β is conventionally supplemented in culture medium at highly supraphysiologic doses to accelerate neocartilage development. While this approach enhances cartilage extracellular matrix (ECM) biosynthesis, it further promotes tissue features detrimental to hyaline cartilage function, including the induction of tissue swelling, hyperplasia, hypertrophy, and ECM heterogeneities. In contrast, during native cartilage development, chondrocytes are surrounded by TGF-β configured in a latent complex (LTGF-β), which undergoes cell-mediated activation, giving rise to moderated, physiologic dosing regimens that enhance ECM biosynthesis while avoiding detrimental features associated with TGF-β excesses. Here, we explore a bio-inspired strategy, consisting of LTGF-β-conjugated scaffolds, providing TGF-β exposure regimens that are moderated and uniformly administered throughout the construct. Specifically, we evaluate the performance of LTGF-β scaffolds to improve neocartilage development with bovine chondrocyte-seeded agarose constructs compared to outcomes from active TGF-β media supplementation (MS) at a physiologic 0.3 ng/mL dose (MS-0.3), supraphysiologic 10 ng/mL dose (MS-10), or TGF-β free. For small-size constructs (∅3×2 mm), LTGF-β scaffolds yield neocartilage that achieves native-matched mechanical properties (800-925 kPa) and sGAG content (6.6%-7.1%), while providing a cell morphology and collagen distribution more reminiscent of hyaline cartilage. LTGF-β scaffolds further afford an optimal chondrogenic phenotype, marked by a 12-to 28-fold reduction of COL-I expression relative to TGF-β-free and a 7-to 17-fold reduction of COL-X expression relative to MS-10. Further, for large-size constructs, which approach the dimensions needed for clinical cartilage repair, LTGF-β scaffolds significantly reduce mechanical and biochemical heterogeneities relative to MS-0.3 and MS-10. Overall, the use of LTGF-β scaffolds improves the composition, structure, material properties, and cell phenotype of neocartilage.
Collapse
|
2
|
Yan W, Cheng J, Wu H, Gao Z, Li Z, Cao C, Meng Q, Wu Y, Ren S, Zhao F, Wang H, Liu P, Wang J, Hu X, Ao Y. Vascular Smooth Muscle Cells Transdifferentiate into Chondrocyte-Like Cells and Facilitate Meniscal Fibrocartilage Regeneration. RESEARCH (WASHINGTON, D.C.) 2024; 7:0555. [PMID: 39717465 PMCID: PMC11665451 DOI: 10.34133/research.0555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024]
Abstract
The effective and translational strategy to regenerate knee meniscal fibrocartilage remained challenging. Herein, we first identified vascular smooth muscle cells (VSMCs) transdifferentiated into fibrochondrocytes and participated in spontaneous meniscal regeneration using smooth muscle cell lineage tracing transgenic mice meniscal defect model. Then, we identified low-intensity pulsed ultrasound (LIPUS) acoustic stimulus enhanced fibrochondrogenic transdifferentiation of VSMCs in vitro and in vivo. Mechanistically, LIPUS stimulus could up-regulate mechanosensitive ion channel Piezo1 expression and then activate the transforming growth factor β1 (TGFβ1) signal, following repression of the Notch signal, consequently enhancing fibrochondrogenic transdifferentiation of VSMCs. Finally, we demonstrated that the regular LIPUS stimulus enhanced anisotropic native-like meniscal fibrocartilage tissue regeneration in a beagle canine subtotal meniscectomy model at 6 months postoperatively. The single-cell RNA sequencing analysis confirmed the role of VSMC fibrochondrogenic transdifferentiation in meniscal regeneration.
Collapse
Affiliation(s)
- Wenqiang Yan
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Jin Cheng
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Haoda Wu
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Zeyuan Gao
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Zong Li
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Chenxi Cao
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Qingyang Meng
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yue Wu
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Shuang Ren
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Fengyuan Zhao
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Hongde Wang
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Ping Liu
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Jianquan Wang
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yingfang Ao
- Department of Sports Medicine, Peking University Third Hospital,
Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| |
Collapse
|
3
|
Dogru S, Alba GM, Pierce KC, Wang T, Kia DS, Albro MB. Cell mediated reactions create TGF-β delivery limitations in engineered cartilage. Acta Biomater 2024; 190:178-190. [PMID: 39447669 PMCID: PMC11614674 DOI: 10.1016/j.actbio.2024.10.032] [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: 06/17/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
During native cartilage development, endogenous TGF-β activity is tightly regulated by cell-mediated chemical reactions in the extracellular milieu (e.g., matrix and receptor binding), providing spatiotemporal control in a manner that is localized and short acting. These regulatory paradigms appear to be at odds with TGF-β delivery needs in tissue engineering (TE) where administered TGF-β is required to transport long distances or reside in tissues for extended durations. In this study, we perform a novel examination of the influence of cell-mediated reactions on the spatiotemporal distribution of administered TGF-β in cartilage TE applications. Reaction rates of TGF-β binding to cell-deposited ECM and TGF-β internalization by cell receptors are experimentally characterized in bovine chondrocyte-seeded tissue constructs. TGF-β binding to the construct ECM exhibits non-linear Brunauer-Emmett-Teller (BET) adsorption behavior, indicating that as many as seven TGF-β molecules can aggregate at a binding site. Cell-mediated TGF-β internalization rates exhibit a biphasic trend, following a Michaelis-Menten relation (Vmax = 2.4 molecules cell-1 s-1, Km = 1.7 ng mL-1) at low ligand doses (≤130 ng/mL), but exhibit an unanticipated non-saturating power trend at higher doses (≥130 ng/mL). Computational models are developed to illustrate the influence of these reactions on TGF-β spatiotemporal delivery profiles for conventional TGF-β administration platforms. For TGF-β delivery via supplementation in culture medium, these reactions give rise to pronounced steady state TGF-β spatial gradients; TGF-β concentration decays by ∼90 % at a depth of only 500 μm from the media-exposed surface. For TGF-β delivery via heparin-conjugated affinity scaffolds, cell mediated internalization reactions significantly reduce the TGF-β scaffold retention time (160-360-fold reduction) relative to acellular heparin scaffolds. This work establishes the significant limitations that cell-mediated chemical reactions engender for TGF-β delivery and highlights the need for novel delivery platforms that account for these reactions to achieve optimal TGF-β exposure profiles. STATEMENT OF SIGNIFICANCE: During native cartilage development, endogenous TGF-β activity is tightly regulated by cell-mediated chemical reactions in the extracellular milieu (e.g., matrix and receptor binding), providing spatiotemporal control in a manner that is localized and short acting. However, the effect of these reactions on the delivery of exogenous TGF-β to engineered cartilage tissues remains not well understood. In this study, we demonstrate that cell-mediated reactions significantly restrict the delivery of TGF-β to cells in engineered cartilage tissue constructs. For delivery via media supplementation, reactions significantly limit TGF-β penetration into constructs. For delivery via scaffold loading, reactions significantly limit TGF-β residence time in constructs. Overall, these results illustrate the impact of cell-mediated chemical reactions on TGF-β delivery profiles and support the importance of accounting for these reactions when designing TGF-β delivery platforms for promoting cartilage regeneration.
Collapse
Affiliation(s)
- Sedat Dogru
- College of Engineering, Boston University, Boston, MA, United States
| | - Gabriela M Alba
- College of Engineering, Boston University, Boston, MA, United States
| | - Kirk C Pierce
- College of Engineering, Boston University, Boston, MA, United States
| | - Tianbai Wang
- College of Engineering, Boston University, Boston, MA, United States
| | | | - Michael B Albro
- College of Engineering, Boston University, Boston, MA, United States.
| |
Collapse
|
4
|
Costa V, Terrando S, Bellavia D, Salvatore C, Alessandro R, Giavaresi G. MiR203a-3p as a potential biomarker for synovial pathology associated with osteoarthritis: a pilot study. J Orthop Surg Res 2024; 19:746. [PMID: 39533317 PMCID: PMC11558974 DOI: 10.1186/s13018-024-05237-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative musculoskeletal disease that significantly impacts the quality of life. Currently, no validated biomarkers for early detection of OA are defined. The possibility of discovering OA biomarkers is the focus of this study. METHODS Human primary OA synovial cells (SVs), isolated from discarded joint tissue of patients with Kellgren & Lawrence score (KL) < 3, (mild/moderate) and KL ≥ 3 (severe), were characterized by FACS analysis. Through qRT-PCR and ELISA assays the inflammation, fibrosis status and the different miRNAs expression has been investigated. The role of miR-203a-3p and its precursors were evaluated through gain and loss of function study, IL-1β synoviocytes treatments and methylation analysis of miR203a promoter. The qRT-PCR analysis of miR203a-3p and pre-miR203a on plasma (isolated 24 h before surgery, 3 days and 1 month after surgery) and synovial fluid (recovered during the surgery) were done to support our in vitro data. RESULTS MiR203a-3p expression is inversely correlated with the aggressiveness of OA, modulating the expression of epithelial to mesenchymal transition (EMT) and pro-inflammatory factors, as well as regulating the expression of secreted protein acidic and rich in cysteine (SPARC) mRNA. Methylation analysis of the miR203a promoter and SVs IL-1β treatment's highlighted the impact of inflammation on miR203a-3p and pre-miR203a expression; as confirmed by both miRNAs detection in biological fluids derived from patients with severe OA. CONCLUSION Our preliminary results suggest that miR-203a-3p might be a potential candidate for staging OA progression and a new protective/predictive biomarker for synovial OA degeneration. Further studies are needed to validate its potential impact on OA.
Collapse
Affiliation(s)
- Viviana Costa
- Scienze E Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Silvio Terrando
- Ortopedia Generale, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| | - Daniele Bellavia
- Scienze E Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano, 1/10, 40136, Bologna, Italy.
| | - Caruccio Salvatore
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Section of Cellular Biology, University of Palermo, 90133, Palermo, Italy
| | - Riccardo Alessandro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Biology and Genetics, University of Palermo, 90133, Palermo, Italy
- Istituto Per La Ricerca E L'Innovazione Biomedica (IRIB-CNR), 90133, Palermo, Italy
| | - Gianluca Giavaresi
- Scienze E Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano, 1/10, 40136, Bologna, Italy
| |
Collapse
|
5
|
Khattar P, Ulmner M, Häbel H, Lund B, Sugars RV. Synovial Matrix Remodeling and Inflammatory Profile in Disc Displacement of the Temporomandibular Joint: An Observational Case-Control Study. Int J Dent 2024; 2024:2450066. [PMID: 39329157 PMCID: PMC11424871 DOI: 10.1155/2024/2450066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/07/2024] [Accepted: 08/14/2024] [Indexed: 09/28/2024] Open
Abstract
Background: Pain-related temporomandibular joint disorders (TMJD) are a major public health problem, including the diagnoses of disc displacement (DD) with and without reduction (DDwR/DDwoR). Objectives: The study aimed to examine the matrix remodeling and the inflammatory profile in synovial tissues of patients with TMJ-DD, with a view to understand the pathophysiology, and to contribute to the development of tissue-based diagnostic criteria. Methods: This laboratory-based observational case-control study included 30 synovial tissue samples obtained from 30 patients, diagnosed with delayed (DO) or sudden (SO) onset of DDwoR, which were compared against the reference patient material, DDwR (n = 10/diagnosis group). Tissue samples were investigated histologically and via quantitative immunohistochemistry for a panel of antibodies targeted against extracellular matrix proteins and inflammatory markers. The data were analyzed using a generalized linear model with a gamma family distribution (p < 0.05). Results: Quantification of immunostaining revealed significant differences in the distribution of collagen type III (DO, p < 0.001), lumican (DO, p < 0.05), matrix metalloproteinase-2 (DO, p < 0.05), CD4 T-helper cells (DO, p < 0.01; SO, p < 0.001), and CD68 monocytic immune cells (both SO and DO, p < 0.001) in DDwoR groups compared to the reference patient material, DDwR. Conclusions: The observations confirmed differences in matrix remodeling and an increase in local inflammatory activity in the DDwoR diagnosis compared to the reference patient material, DDwR. The study highlighted the importance of synovial tissue characterization to unite micropathology and clinical findings, leading to more reliable diagnostic tools.
Collapse
Affiliation(s)
- Pallavi Khattar
- Department of Dental MedicineKarolinska Institutet, Stockholm 171 77, Sweden
| | - Mattias Ulmner
- Department of Dental MedicineKarolinska Institutet, Stockholm 171 77, Sweden
- Medical Unit of Plastic Surgery and Oral and Maxillofacial SurgeryKarolinska University Hospital, Stockholm 171 76, Sweden
| | - Henrike Häbel
- Department of Learning, Informatics, Management and EthicsKarolinska Institutet, Stockholm 171 77, Sweden
| | - Bodil Lund
- Department of Dental MedicineKarolinska Institutet, Stockholm 171 77, Sweden
- Medical Unit of Plastic Surgery and Oral and Maxillofacial SurgeryKarolinska University Hospital, Stockholm 171 76, Sweden
| | - Rachael V. Sugars
- Department of Dental MedicineKarolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
6
|
Ladner YD, Stoddart MJ. Tribological loading of cartilage and chondrogenic cells. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100088. [PMID: 38389863 PMCID: PMC10881303 DOI: 10.1016/j.bbiosy.2024.100088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/06/2023] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
Novel cartilage regeneration therapies often look promising in-vitro but fail when implanted in vivo. One of the possible reasons for this discrepancy is the simplified, static in-vitro chondrogenesis models typically used. Complex mechanical stimulation plays a key role in physiological cartilage and chondrogenic cell metabolism, including the development of cartilage structure, yet it is routinely lacking during in-vitro studies. Multiaxial load bioreactors are becoming more widespread and offer advantages over more simple loading devices. Within this article, we highlight some of the important findings from in-vitro assays and key aspects relating to tribological loading of cartilage and chondrogenic cells.
Collapse
Affiliation(s)
- Yann D Ladner
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos Platz, Switzerland
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos Platz, Switzerland
| |
Collapse
|
7
|
St Amant J, Michaud J, Hinds D, Coyle M, Pozzi A, Clark AL. Depleting transforming growth factor beta receptor 2 signalling in the cartilage of itga1-null mice attenuates spontaneous knee osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100399. [PMID: 37649532 PMCID: PMC10462827 DOI: 10.1016/j.ocarto.2023.100399] [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: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Objectives Integrin α1β1 protects against osteoarthritis (OA) when it is upregulated in the superficial zone of cartilage in the early stages of disease. However, the mechanism behind this protection is unknown. Integrin α1β1 moderates transforming growth factor β receptor II (TGFBR2) signalling, a critical regulator of chondrocyte anabolic activity. To this end, mice lacking integrin α1β1 have increased baseline activation of TGFBR2 signalling and overall fibrosis. The purpose of this study was to evaluate the interplay between integrin α1β1 and TGFBR2 in the development of spontaneous OA. We hypothesized that dampening TGFBR2 signalling in the cartilage of itga1-null mice would attenuate OA. Methods Behavioural and histological manifestations of spontaneous knee OA were measured at 4, 8, 12 and 16 months in mice with and without a ubiquitous itga1 deletion and with and without a tamoxifen-induced cartilage specific TGFBR2 depletion. Results Knee cartilage degeneration, collateral ligament ossification and pain responses increased with age. Itga1-null mice with intact TGFBR2 signalling developed earlier and more severe OA compared to controls. In agreement with our hypothesis, depleting TGFBR2 signalling in the cartilage of itga1-null mice attenuated OA progression. Conclusion Intact TGFBR2 signalling drives early and worse knee OA in itga1-null mice. This result supports the hypothesis that the increased expression of integrin α1β1 by superficial zone chondrocytes early in OA development dampens TGFBR2 signalling and thus protects against degeneration.
Collapse
Affiliation(s)
- Jennifer St Amant
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Jana Michaud
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Daniel Hinds
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Madison Coyle
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Andrea L. Clark
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
8
|
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
|
9
|
Wang T, Dogru S, Dai Z, Kim SY, Vickers NA, Albro MB. Physiologic Doses of TGF-β Improve the Composition of Engineered Articular Cartilage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559554. [PMID: 37808691 PMCID: PMC10557735 DOI: 10.1101/2023.09.27.559554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
For cartilage regeneration applications, transforming growth factor beta (TGF-β) is conventionally administered at highly supraphysiologic doses (10-10,000 ng/mL) in an attempt to cue cells to fabricate neocartilage that matches the composition, structure, and functional properties of native hyaline cartilage. While supraphysiologic doses enhance ECM biosynthesis, they are also associated with inducing detrimental tissue features, such as fibrocartilage matrix deposition, pathologic-like chondrocyte clustering, and tissue swelling. Here we investigate the hypothesis that moderated TGF-β doses (0.1-1 ng/mL), akin to those present during physiological cartilage development, can improve neocartilage composition. Variable doses of media-supplemented TGF-β were administered to a model system of reduced-size cylindrical constructs (Ø2-Ø3 mm), which mitigate the TGF-β spatial gradients observed in conventional-size constructs (Ø4-Ø6 mm), allowing for a novel assessment of the intrinsic effect of TGF-β doses on macroscale neocartilage properties and composition. The administration of physiologic TGF-β to reduced-size constructs yields neocartilage with native-matched sGAG content and mechanical properties while providing a more hyaline cartilage-like composition, marked by: 1) reduced fibrocartilage-associated type I collagen, 2) 77% reduction in the fraction of cells present in a clustered morphology, and 3) 45% reduction in the degree of tissue swelling. Physiologic TGF-β appears to achieve an important balance of promoting requisite ECM biosynthesis, while mitigating hyaline cartilage compositional deficits. These results can guide the development of novel physiologic TGF-β-delivering scaffolds to improve the regeneration clinical-sized neocartilage tissues.
Collapse
|
10
|
Sarkovich S, Issa PP, Longanecker A, Martin D, Redondo K, McTernan P, Simkin J, Marrero L. Minoxidil weakens newly synthesized collagen in fibrotic synoviocytes from osteoarthritis patients. J Exp Orthop 2023; 10:84. [PMID: 37605092 PMCID: PMC10441905 DOI: 10.1186/s40634-023-00650-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023] Open
Abstract
PURPOSE Synovial fibrosis (SFb) formation and turnover attributable to knee osteoarthritis (KOA) can impart painful stiffness and persist following arthroplasty. To supplement joint conditioning aimed at maximizing peri-operative function, we evaluated the antifibrotic effect of Minoxidil (MXD) on formation of pyridinoline (Pyd) cross-links catalyzed by Plod2-encoded lysyl hydroxylase (LH)2b that strengthen newly synthesized type-I collagen (COL1) in fibroblastic synovial cells (FSCs) from KOA patients. MXD was predicted to decrease Pyd without significant alterations to Col1a1 transcription by FSCs stimulated with transforming growth factor (TGF)β1. METHODS Synovium from 10 KOA patients grouped by SFb severity was preserved for picrosirius and LH2b histology or culture. Protein and RNA were purified from fibrotic FSCs after 8 days with or without 0.5 µM MXD and/or 4 ng/mL of TGFβ1. COL1 and Pyd protein concentrations from ELISA and expression of Col1a1, Acta2, and Plod2 genes by qPCR were compared by parametric tests with α = 0.05. RESULTS Histological LH2b expression corresponded to SFb severity. MXD attenuated COL1 output in KOA FSCs but only in the absence of TGFβ1 and consistently decreased Pyd under all conditions with significant downregulation of Plod2 but minimal alterations to Col1a1 and Acta2 transcripts. CONCLUSIONS MXD is an attractive candidate for local antifibrotic pharmacotherapy for SFb by compromising the integrity of newly formed fibrous deposits by FSCs during KOA and following arthroplasty. Targeted antifibrotic supplementation could improve physical therapy and arthroscopic lysis strategies aimed at breaking down joint scarring. However, the effect of MXD on other joint-specific TGFβ1-mediated processes or non-fibrotic components requires further investigation.
Collapse
Affiliation(s)
- Stefan Sarkovich
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, 2021 Perdido St., Center for Advanced Learning and Simulation, 7th floor, New Orleans, LA, 70112, USA
| | - Peter P Issa
- School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier St., Lions Building, 5th floor, New Orleans, LA, 70112, USA
| | - Andrew Longanecker
- School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier St., Lions Building, 5th floor, New Orleans, LA, 70112, USA
| | - Davis Martin
- School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier St., Lions Building, 5th floor, New Orleans, LA, 70112, USA
| | - Kaitlyn Redondo
- Morphology and Imaging Core, Louisiana State University Health Sciences Center, 533 Bolivar St., Clinical Sciences Research Building, 5th floor, New Orleans, LA, 70112, USA
| | - Patrick McTernan
- Department of Physiology, Louisiana State University Health Sciences Center, 533 Bolivar St., Clinical Sciences Research Building, 4th floor, New Orleans, LA, 70112, USA
| | - Jennifer Simkin
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, 2021 Perdido St., Center for Advanced Learning and Simulation, 7th floor, New Orleans, LA, 70112, USA
| | - Luis Marrero
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, 2021 Perdido St., Center for Advanced Learning and Simulation, 7th floor, New Orleans, LA, 70112, USA.
- School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier St., Lions Building, 5th floor, New Orleans, LA, 70112, USA.
- Morphology and Imaging Core, Louisiana State University Health Sciences Center, 533 Bolivar St., Clinical Sciences Research Building, 5th floor, New Orleans, LA, 70112, USA.
| |
Collapse
|
11
|
Fani N, Peshkova M, Bikmulina P, Golroo R, Timashev P, Vosough M. Fabricating the cartilage: recent achievements. Cytotechnology 2023; 75:269-292. [PMID: 37389132 PMCID: PMC10299965 DOI: 10.1007/s10616-023-00582-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 05/09/2023] [Indexed: 07/01/2023] Open
Abstract
This review aims to describe the most recent achievements and provide an insight into cartilage engineering and strategies to restore the cartilage defects. Here, we discuss cell types, biomaterials, and biochemical factors applied to form cartilage tissue equivalents and update the status of fabrication techniques, which are used at all stages of engineering the cartilage. The actualized concept to improve the cartilage tissue restoration is based on applying personalized products fabricated using a full cycle platform: a bioprinter, a bioink consisted of ECM-embedded autologous cell aggregates, and a bioreactor. Moreover, in situ platforms can help to skip some steps and enable adjusting the newly formed tissue in the place during the operation. Only some achievements described have passed first stages of clinical translation; nevertheless, the number of their preclinical and clinical trials is expected to grow in the nearest future.
Collapse
Affiliation(s)
- Nesa Fani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Polina Bikmulina
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, Moscow, Russia
| | - Reihaneh Golroo
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
12
|
Dogru S, Dai Z, Alba GM, Simone NJ, Albro MB. Computational and experimental characterizations of the spatiotemporal activity and functional role of TGF-β in the synovial joint. J Biomech 2023; 156:111673. [PMID: 37364394 DOI: 10.1016/j.jbiomech.2023.111673] [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: 08/31/2022] [Revised: 03/21/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023]
Abstract
TGF-β is a prominent anabolic signaling molecule associated with synovial joint health. Recent work has uncovered mechanochemical mechanisms that activate the latent form of TGF-β (LTGF-β) in the synovial joint-synovial fluid (SF) shearing or cartilage compression-pointing to mechanobiological phenomena, whereby enhanced TGF-β activity occurs during joint stimulation. Here, we implement computational and experimental models to better understand the role of mechanochemical-activated TGF-β (aTGF-β) in regulating the functional biosynthetic activities of synovial joint tissues. Reaction-diffusion models describe the pronounced role of extracellular chemical reactions-load-induced activation, reversible ECM-binding, and cell-mediated internalization-in modulating the spatiotemporal distribution of aTGF-β in joint tissues. Of note, aTGF-β from SF shearing predominantly acts on cells in peripheral tissue regions (superficial zone [SZ] chondrocytes and synoviocytes) and aTGF-β from cartilage compression acts on chondrocytes through all cartilage layers. Further, ECM reversible binding sites in cartilage act to modulate the temporal delivery of aTGF-β to cells, creating a dynamic where short durations of joint activity give rise to extended periods of aTGF-β exposure at moderated doses. Ex vivo tissue models were subsequently utilized to characterize the influence of physiologic aTGF-β activity regimens in regulating functional biosynthetic activities. Physiologic exposure regimens of aTGF-β in SF induce strong 4-fold to 9-fold enhancements in the secretion rate of the synovial biolubricant, PRG4, from SZ cartilage and synovium explants. Further, aTGF-β inhibition in cartilage over 1-month culture leads to a pronounced loss of GAG content (30-35% decrease) and tissue softening (60-65% EY reduction). Overall, this work advances a novel perspective on the regulation of TGF-β in the synovial joint and its role in maintaining synovial joint health.
Collapse
Affiliation(s)
- Sedat Dogru
- Department of Mechanical Engineering, Boston University, United States
| | - Zhonghao Dai
- Department of Biomedical Engineering, Boston University, United States
| | - Gabriela M Alba
- Department of Biomedical Engineering, Boston University, United States
| | - Nicholas J Simone
- Department of Biomedical Engineering, Boston University, United States
| | - Michael B Albro
- Department of Mechanical Engineering, Boston University, United States; Department of Biomedical Engineering, Boston University, United States; Division of Materials Science & Engineering, Boston University, United States.
| |
Collapse
|
13
|
Rahman MM, Watton PN, Neu CP, Pierce DM. A chemo-mechano-biological modeling framework for cartilage evolving in health, disease, injury, and treatment. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 231:107419. [PMID: 36842346 DOI: 10.1016/j.cmpb.2023.107419] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND AND OBJECTIVE Osteoarthritis (OA) is a pervasive and debilitating disease, wherein degeneration of cartilage features prominently. Despite extensive research, we do not yet understand the cause or progression of OA. Studies show biochemical, mechanical, and biological factors affect cartilage health. Mechanical loads influence synthesis of biochemical constituents which build and/or break down cartilage, and which in turn affect mechanical loads. OA-associated biochemical profiles activate cellular activity that disrupts homeostasis. To understand the complex interplay among mechanical stimuli, biochemical signaling, and cartilage function requires integrating vast research on experimental mechanics and mechanobiology-a task approachable only with computational models. At present, mechanical models of cartilage generally lack chemo-biological effects, and biochemical models lack coupled mechanics, let alone interactions over time. METHODS We establish a first-of-its kind virtual cartilage: a modeling framework that considers time-dependent, chemo-mechano-biologically induced turnover of key constituents resulting from biochemical, mechanical, and/or biological activity. We include the "minimally essential" yet complex chemical and mechanobiological mechanisms. Our 3-D framework integrates a constitutive model for the mechanics of cartilage with a novel model of homeostatic adaptation by chondrocytes to pathological mechanical stimuli, and a new application of anisotropic growth (loss) to simulate degradation clinically observed as cartilage thinning. RESULTS Using a single set of representative parameters, our simulations of immobilizing and overloading successfully captured loss of cartilage quantified experimentally. Simulations of immobilizing, overloading, and injuring cartilage predicted dose-dependent recovery of cartilage when treated with suramin, a proposed therapeutic for OA. The modeling framework prompted us to add growth factors to the suramin treatment, which predicted even better recovery. CONCLUSIONS Our flexible framework is a first step toward computational investigations of how cartilage and chondrocytes mechanically and biochemically evolve in degeneration of OA and respond to pharmacological therapies. Our framework will enable future studies to link physical activity and resulting mechanical stimuli to progression of OA and loss of cartilage function, facilitating new fundamental understanding of the complex progression of OA and elucidating new perspectives on causes, treatments, and possible preventions.
Collapse
Affiliation(s)
| | - Paul N Watton
- Department of Computer Science & Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK; Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, PA, USA
| | - Corey P Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - David M Pierce
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
14
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
15
|
Bailey KN, Alliston T. At the Crux of Joint Crosstalk: TGFβ Signaling in the Synovial Joint. Curr Rheumatol Rep 2022; 24:184-197. [PMID: 35499698 PMCID: PMC9184360 DOI: 10.1007/s11926-022-01074-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW The effect of the transforming growth factor beta (TGFβ) signaling pathway on joint homeostasis is tissue-specific, non-linear, and context-dependent, representing a unique complexity in targeting TGFβ signaling in joint disease. Here we discuss the variety of mechanisms that TGFβ signaling employs in the synovial joint to maintain healthy joint crosstalk and the ways in which aberrant TGFβ signaling can result in joint degeneration. RECENT FINDINGS Osteoarthritis (OA) epitomizes a condition of disordered joint crosstalk in which multiple joint tissues degenerate leading to overall joint deterioration. Synovial joint tissues, such as subchondral bone, articular cartilage, and synovium, as well as mesenchymal stem cells, each demonstrate aberrant TGFβ signaling during joint disease, whether by excessive or suppressed signaling, imbalance of canonical and non-canonical signaling, a perturbed mechanical microenvironment, or a distorted response to TGFβ signaling during aging. The synovial joint relies upon a sophisticated alliance among each joint tissue to maintain joint homeostasis. The TGFβ signaling pathway is a key regulator of the health of individual joint tissues, and the subsequent interaction among these different joint tissues, also known as joint crosstalk. Dissecting the sophisticated function of TGFβ signaling in the synovial joint is key to therapeutically interrogating the pathway to optimize overall joint health.
Collapse
Affiliation(s)
- Karsyn N Bailey
- Department of Orthopaedic Surgery, University of California San Francisco, 513 Parnassus Avenue, CA, 94143, San Francisco, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, 513 Parnassus Avenue, CA, 94143, San Francisco, USA.
| |
Collapse
|
16
|
Optimization of loading protocols for tissue engineering experiments. Sci Rep 2022; 12:5094. [PMID: 35332169 PMCID: PMC8948220 DOI: 10.1038/s41598-022-08849-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/14/2022] [Indexed: 11/08/2022] Open
Abstract
Tissue engineering (TE) combines cells and biomaterials to treat orthopedic pathologies. Maturation of de novo tissue is highly dependent on local mechanical environments. Mechanical stimulation influences stem cell differentiation, however, the role of various mechanical loads remains unclear. While bioreactors simplify the complexity of the human body, the potential combination of mechanical loads that can be applied make it difficult to assess how different factors interact. Human bone marrow-derived mesenchymal stromal cells were seeded within a fibrin-polyurethane scaffold and exposed to joint-mimicking motion. We applied a full factorial design of experiment to investigate the effect that the interaction between different mechanical loading parameters has on biological markers. Additionally, we employed planned contrasts to analyze differences between loading protocols and a linear mixed model with donor as random effect. Our approach enables screening of multiple mechanical loading combinations and identification of significant interactions that could not have been studied using classical mechanobiology studies. This is useful to screen the effect of various loading protocols and could also be used for TE experiments with small sample sizes and further combinatorial medication studies.
Collapse
|
17
|
Han AJ, Alexander LC, Huebner JL, Reed AB, Kraus VB. Increase in Free and Total Plasma TGF-β1 Following Physical Activity. Cartilage 2021; 13:1741S-1748S. [PMID: 32340467 PMCID: PMC8808803 DOI: 10.1177/1947603520916523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To evaluate effects of physical activity and food consumption on plasma concentrations of free and total transforming growth factor beta-1 (TGF-β1), beta-2 (TGF-β2), and beta-3 (TGF-β3) in individuals with knee osteoarthritis (OA). METHODS Participants (n = 40 in 2 cohorts of 20; mean age 70 years) with radiographic knee OA were admitted overnight for serial blood sampling. Cohorts 1 and 2 assessed the impacts of food intake and activity, respectively, on TGF-β concentrations. Cohort 1 blood draws included 2 hours postprandial the evening of day 1 (T3), fasting before rising on day 2 (T0), nonfasting 1 hour after rising (T1B), and 4 hours after rising (T2). Cohort 2 blood draws included T3, T0, fasting 1 hour after rising and performing activities of daily living (T1A), and nonfasting 2 hours after rising (T1B). By sandwich ELISAs, we quantified plasma free and total TGF-β1 concentrations in all samples, and plasma total TGF-β2 and TGF-β3 in cohort 2. RESULTS Free TGF-β1 represented a small fraction of the total systemic concentration (mean 0.026%). In cohort 2, free and total TGF-β1 and total TGF-β2 concentration significantly increased in fasting samples collected after an hour (T1A) of activities of daily living (free TGF-β1: P = 0.006; total TGF-β1: P < 0.001; total TGF-β2: P = 0.001). Total TGF-β3 increased nonsignificantly following activity (P = 0.590) and decreased (P = 0.035) after food consumption while resting (T1B). CONCLUSIONS Increased plasma concentrations of TGF-β with physical activity suggests activity should be standardized prior to TGF-β1 analyses.
Collapse
Affiliation(s)
- Ashley J. Han
- Department of Medicine, Duke Molecular
Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Louie C. Alexander
- Department of Medicine, Duke Molecular
Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Janet L. Huebner
- Department of Medicine, Duke Molecular
Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Alexander B. Reed
- Department of Medicine, Duke Molecular
Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Virginia B. Kraus
- Department of Medicine, Duke Molecular
Physiology Institute, Duke University School of Medicine, Durham, NC, USA,Department of Medicine, Division of
Rheumatology, Duke University School of Medicine, Durham, NC, USA,Virginia B. Kraus, Department of Medicine,
Duke Molecular Physiology Institute, Duke University School of Medicine, PO Box
104775, Carmichael Building, 300 North Duke Street, Durham, NC 27701, USA.
| |
Collapse
|
18
|
Neefjes M, Housmans BAC, van Beuningen HM, Vitters EL, van den Akker GGH, Welting TJM, van Caam APM, van der Kraan PM. Prediction of the Effect of the Osteoarthritic Joint Microenvironment on Cartilage Repair. Tissue Eng Part A 2021; 28:27-37. [PMID: 34039008 DOI: 10.1089/ten.tea.2021.0051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Osteoarthritis (OA) is characterized by progressive articular cartilage loss. Human mesenchymal stromal cells (MSCs) can be used for cartilage repair therapies based on their potential to differentiate into chondrocytes. However, the joint microenvironment is a major determinant of the success of MSC-based cartilage formation. Currently, there is no tool that is able to predict the effect of a patient's OA joint microenvironment on MSC-based cartilage formation. Our goal was to develop a molecular tool that can predict this effect before the start of cartilage repair therapies. Six different promoter reporters (hIL6, hIL8, hADAMTS5, hWISP1, hMMP13, and hADAM28) were generated and evaluated in an immortalized human articular chondrocyte for their responsiveness to an osteoarthritic microenvironment by stimulation with OA synovium-conditioned medium (OAs-cm) obtained from 32 different knee OA patients. To study the effect of this OA microenvironment on MSC-based cartilage formation, MSCs were cultured in a three-dimensional pellet culture model, while stimulated with OAs-cm. Cartilage formation was assessed histologically and by quantifying sulfated glycosaminoglycan (sGAG) production. We confirmed that OAs-cm of different patients had significantly different effects on sGAG production. In addition, significant correlations were obtained between the effect of the OAs-cm on cartilage formation and promoter reporter outcome. Furthermore, we validated the predictive value of measuring two promoter reporters with an independent cohort of OAs-cm and the effect of 87.5% of the OAs-cm on MSC-based cartilage formation could be predicted. Together, we developed a novel tool to predict the effect of the OA joint microenvironment on MSC-based cartilage formation. This is an important first step toward personalized cartilage repair strategies for OA patients.
Collapse
Affiliation(s)
- Margot Neefjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bas A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Henk M van Beuningen
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elly L Vitters
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Guus G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Arjan P M van Caam
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
19
|
Monteiro DA, Dole NS, Campos JL, Kaya S, Schurman CA, Belair CD, Alliston T. Fluid shear stress generates a unique signaling response by activating multiple TGFβ family type I receptors in osteocytes. FASEB J 2021; 35:e21263. [PMID: 33570811 PMCID: PMC7888383 DOI: 10.1096/fj.202001998r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/11/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022]
Abstract
Bone is a dynamic tissue that constantly adapts to changing mechanical demands. The transforming growth factor beta (TGFβ) signaling pathway plays several important roles in maintaining skeletal homeostasis by both coupling the bone‐forming and bone‐resorbing activities of osteoblasts and osteoclasts and by playing a causal role in the anabolic response of bone to applied loads. However, the extent to which the TGFβ signaling pathway in osteocytes is directly regulated by fluid shear stress (FSS) is unknown, despite work suggesting that fluid flow along canaliculi is a dominant physical cue sensed by osteocytes following bone compression. To investigate the effects of FSS on TGFβ signaling in osteocytes, we stimulated osteocytic OCY454 cells cultured within a microfluidic platform with FSS. We find that FSS rapidly upregulates Smad2/3 phosphorylation and TGFβ target gene expression, even in the absence of added TGFβ. Indeed, relative to treatment with TGFβ, FSS induced a larger increase in levels of pSmad2/3 and Serpine1 that persisted even in the presence of a TGFβ receptor type I inhibitor. Our results show that FSS stimulation rapidly induces phosphorylation of multiple TGFβ family R‐Smads by stimulating multimerization and concurrently activating several TGFβ and BMP type I receptors, in a manner that requires the activity of the corresponding ligand. While the individual roles of the TGFβ and BMP signaling pathways in bone mechanotransduction remain unclear, these results implicate that FSS activates both pathways to generate a downstream response that differs from that achieved by either ligand alone.
Collapse
Affiliation(s)
- David A Monteiro
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Neha S Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - J Luke Campos
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Charles A Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Cassandra D Belair
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA.,Department of Urology, University of California, San Francisco, CA, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA.,The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| |
Collapse
|
20
|
Chijimatsu R, Miwa S, Okamura G, Miyahara J, Tachibana N, Ishikura H, Higuchi J, Maenohara Y, Tsuji S, Sameshima S, Takagi K, Nakazato K, Kawaguchi K, Yamagami R, Inui H, Taketomi S, Tanaka S, Saito T. Divergence in chondrogenic potential between in vitro and in vivo of adipose- and synovial-stem cells from mouse and human. Stem Cell Res Ther 2021; 12:405. [PMID: 34266496 PMCID: PMC8281654 DOI: 10.1186/s13287-021-02485-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Somatic stem cell transplantation has been performed for cartilage injury, but the reparative mechanisms are still conflicting. The chondrogenic potential of stem cells are thought as promising features for cartilage therapy; however, the correlation between their potential for chondrogenesis in vitro and in vivo remains undefined. The purpose of this study was to investigate the intrinsic chondrogenic condition depends on cell types and explore an indicator to select useful stem cells for cartilage regeneration. METHODS The chondrogenic potential of two different stem cell types derived from adipose tissue (ASCs) and synovium (SSCs) of mice and humans was assessed using bone morphogenic protein-2 (BMP2) and transforming growth factor-β1 (TGFβ1). Their in vivo chondrogenic potential was validated through transplantation into a mouse osteochondral defect model. RESULTS All cell types showed apparent chondrogenesis under the combination of BMP2 and TGFβ1 in vitro, as assessed by the formation of proteoglycan- and type 2 collagen (COL2)-rich tissues. However, our results vastly differed with those observed following single stimulation among species and cell types; apparent chondrogenesis of mouse SSCs was observed with supplementation of BMP2 or TGFβ1, whereas chondrogenesis of mouse ASCs and human SSCs was observed with supplementation of BMP2 not TGFβ1. Human ASCs showed no obvious chondrogenesis following single stimulation. Mouse SSCs showed the formation of hyaline-like cartilage which had less fibrous components (COL1/3) with supplementation of TGFβ1. However, human cells developed COL1/3+ tissues with all treatments. Transcriptomic analysis for TGFβ receptors and ligands of cells prior to chondrogenic induction did not indicate their distinct reactivity to the TGFβ1 or BMP2. In the transplanted site in vivo, mouse SSCs formed hyaline-like cartilage (proteoglycan+/COL2+/COL1-/COL3-) but other cell types mainly formed COL1/3-positive fibrous tissues in line with in vitro reactivity to TGFβ1. CONCLUSION Optimal chondrogenic factors driving chondrogenesis from somatic stem cells are intrinsically distinct among cell types and species. Among them, the response to TGFβ1 may possibly represent the fate of stem cells when locally transplanted into cartilage defects.
Collapse
Affiliation(s)
- Ryota Chijimatsu
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Satoshi Miwa
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Junya Miyahara
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naohiro Tachibana
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hisatoshi Ishikura
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junya Higuchi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuji Maenohara
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Shin Sameshima
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kentaro Takagi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keiu Nakazato
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kohei Kawaguchi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryota Yamagami
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Inui
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shuji Taketomi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
21
|
He Y, Yocum L, Alexander PG, Jurczak MJ, Lin H. Urolithin A Protects Chondrocytes From Mechanical Overloading-Induced Injuries. Front Pharmacol 2021; 12:703847. [PMID: 34220525 PMCID: PMC8245698 DOI: 10.3389/fphar.2021.703847] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/04/2021] [Indexed: 01/10/2023] Open
Abstract
Physiological mechanical stimulation has been shown to promote chondrogenesis, but excessive mechanical loading results in cartilage degradation. Currently, the underlying mechanotransduction pathways in the context of physiological and injurious loading are not fully understood. In this study, we aim to identify the critical factors that dictate chondrocyte response to mechanical overloading, as well as to develop therapeutics that protect chondrocytes from mechanical injuries. Specifically, human chondrocytes were loaded in hyaluronic hydrogel and then subjected to dynamic compressive loading under 5% (DL-5% group) or 25% strain (DL-25% group). Compared to static culture and DL-5%, DL-25% reduced cartilage matrix formation from chondrocytes, which was accompanied by the increased senescence level, as revealed by higher expression of p21, p53, and senescence-associated beta-galactosidase (SA-β-Gal). Interestingly, mitophagy was suppressed by DL-25%, suggesting a possible role for the restoration mitophagy in reducing cartilage degeneration with mechanical overloading. Next, we treated the mechanically overloaded samples (DL-25%) with Urolithin A (UA), a natural metabolite previously shown to enhance mitophagy in other cell types. qRT-PCR, histology, and immunostaining results confirmed that UA treatment significantly increased the quantity and quality of cartilage matrix deposition. Interestingly, UA also suppressed the senescence level induced by mechanical overloading, demonstrating its senomorphic potential. Mechanistic analysis confirmed that UA functioned partially by enhancing mitophagy. In summary, our results show that mechanical overloading results in cartilage degradation partially through the impairment of mitophagy. This study also identifies UA's novel use as a compound that can protect chondrocytes from mechanical injuries, supporting high-quality cartilage formation/maintenance.
Collapse
Affiliation(s)
- Yuchen He
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Lauren Yocum
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Peter G Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
22
|
Xu L, Li Y. A Molecular Cascade Underlying Articular Cartilage Degeneration. Curr Drug Targets 2021; 21:838-848. [PMID: 32056522 DOI: 10.2174/1389450121666200214121323] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/11/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022]
Abstract
Preserving of articular cartilage is an effective way to protect synovial joints from becoming osteoarthritic (OA) joints. Understanding of the molecular basis of articular cartilage degeneration will provide valuable information in the effort to develop cartilage preserving drugs. There are currently no disease-modifying OA drugs (DMOADs) available to prevent articular cartilage destruction during the development of OA. Current drug treatments for OA focus on the reduction of joint pain, swelling, and inflammation at advanced stages of the disease. However, based on discoveries from several independent research laboratories and our laboratory in the past 15 to 20 years, we believe that we have a functional molecular understanding of articular cartilage degeneration. In this review article, we present and discuss experimental evidence to demonstrate a sequential chain of the molecular events underlying articular cartilage degeneration, which consists of transforming growth factor beta 1, high-temperature requirement A1 (a serine protease), discoidin domain receptor 2 (a cell surface receptor tyrosine kinase for native fibrillar collagens), and matrix metalloproteinase 13 (an extracellularmatrix degrading enzyme). If, as we strongly suspect, this molecular pathway is responsible for the initiation and acceleration of articular cartilage degeneration, which eventually leads to progressive joint failure, then these molecules may be ideal therapeutic targets for the development of DMOADs.
Collapse
Affiliation(s)
- Lin Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02115 & Faculty of Medicine, Harvard Medical School 25 Shattuck St. Boston, MA 02115, United States
| | - Yefu Li
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02115 & Faculty of Medicine, Harvard Medical School 25 Shattuck St. Boston, MA 02115, United States
| |
Collapse
|
23
|
Lee AJ, Mahoney CM, Cai CC, Ichinose R, Stefani RM, Marra KG, Ateshian GA, Shah RP, Vunjak-Novakovic G, Hung CT. Sustained Delivery of SB-431542, a Type I Transforming Growth Factor Beta-1 Receptor Inhibitor, to Prevent Arthrofibrosis. Tissue Eng Part A 2021; 27:1411-1421. [PMID: 33752445 DOI: 10.1089/ten.tea.2021.0029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Fibrosis of the knee is a common disorder resulting from an aberrant wound healing response and is characterized by extracellular matrix deposition, joint contraction, and scar tissue formation. The principal regulator of the fibrotic cascade is transforming growth factor beta-1 (TGF-β1), a factor that induces rapid proliferation and differentiation of resident fibroblasts. In this study, we demonstrate successful inhibition of TGF-β1-driven myofibroblastic differentiation in human fibroblast-like synoviocytes using a small molecule TGF-β1 receptor inhibitor, SB-431542. We also demonstrate successful encapsulation of SB-431542 in poly(D,L-lactide-co-glycolide) (PLGA) as a potential prophylactic treatment for arthrofibrosis and characterize drug release and bioactivity in a three-dimensional collagen gel contraction assay. We assessed the effects of TGF-β1 and SB-431542 on cell proliferation and viability in monolayer cultures. Opposing dose-dependent trends were observed in cell proliferation, which increased in TGF-β1-treated cultures and decreased in SB-431542-treated cultures relative to control (p < 0.05). SB-431542 was not cytotoxic at the concentrations studied (0-50 μM) and inhibited TGF-β1-induced collagen gel contraction in a dose-dependent manner. Specifically, TGF-β1-treated gels contracted to 18% ± 1% of their initial surface area, while gels treated with TGF-β1 and ≥10 μM SB-431542 showed no evidence of contraction (p < 0.0001). Upon removal of the compound, all gels contracted to control levels after 44 h in culture, necessitating sustained delivery for prolonged inhibition. To this end, SB-431542 was encapsulated in PLGA microspheres (SBMS) that had an average diameter of 87.5 ± 24 μm and a loading capacity of 4.3 μg SB-431542 per milligram of SBMS. Functional assessment of SBMS revealed sustained inhibition of TGF-β1-induced gel contraction as well as hallmark features of myofibroblastic differentiation, including α-smooth muscle actin expression and connective tissue growth factor production. These results suggest that SB-431542 may be used to counter TGF-β1-driven events in the fibrotic cascade in the knee cartilage.
Collapse
Affiliation(s)
- Andy J Lee
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Christopher M Mahoney
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Charles C Cai
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Rika Ichinose
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Robert M Stefani
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, New York, New York, USA.,Department of Mechanical Engineering, Columbia University, New York, New York, USA
| | - Roshan P Shah
- Department of Orthopaedic Surgery, and Columbia University, New York, New York, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, New York, USA.,Department of Medicine, Columbia University, New York, New York, USA
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, New York, New York, USA.,Department of Orthopaedic Surgery, and Columbia University, New York, New York, USA
| |
Collapse
|
24
|
Wiegertjes R, Thielen NGM, van Caam APM, van Laar M, van Beuningen HM, Koenders MI, van Lent PLEM, van der Kraan PM, van de Loo FAJ, Blaney Davidson EN. Increased IL-6 receptor expression and signaling in ageing cartilage can be explained by loss of TGF-β-mediated IL-6 receptor suppression. Osteoarthritis Cartilage 2021; 29:773-782. [PMID: 33617971 DOI: 10.1016/j.joca.2021.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 12/12/2020] [Accepted: 01/06/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) development is strongly associated with ageing, possibly due to age-related changes in transforming growth factor-β (TGF-β) signaling in cartilage. Recently, we showed that TGF-β suppresses interleukin (IL)-6 receptor (IL-6R) expression in chondrocytes. As IL-6 is involved in cartilage degeneration, we hypothesized that age-related loss of TGF-β signaling results in increased IL-6R expression and signaling in ageing cartilage. DESIGN Bovine articular cartilage was collected and immediately processed to study age-related changes in IL-6R expression using qPCR and IHC (age-range: 0.5-14 years). Moreover, cartilage from young and aged cows was stimulated with rhIL-6 and/or rhTGF-β1 to measure IL-6-induced p-STAT3 using Western blot. Expression of STAT3-responsive genes was analyzed using qPCR. RESULTS Expression of IL-6 receptor (bIL-6R) significantly increased in cartilage upon ageing (slope: 0.32, 95%CI: 0.20-0.45), while expression of glycoprotein 130 (bGP130) was unaffected. Cartilage stimulation with IL-6 showed increased induction of p-STAT3 upon ageing (slope: 0.14, 95%CI: 0.08-0.20). Furthermore, IL-6-mediated induction of STAT3-responsive genes like bSOCS3 and bMMP3 was increased in aged compared to young cartilage. Interestingly, the ability of TGF-β to suppress bIL6R expression in young cartilage was lost upon ageing (slope: 0.21, 95%CI: 0.13-0.30). Concurrently, an age-related loss in TGF-β-mediated suppression of IL-6-induced p-STAT3 and bSOCS3 expression was observed. CONCLUSIONS Ageing results in enhanced IL-6R expression and subsequent IL-6-induced p-STAT3 signaling in articular cartilage. This is likely caused by age-related loss of protective TGF-β signaling, resulting in loss of TGF-β-mediated IL-6R suppression. Because of the detrimental role of IL-6 in cartilage, this mechanism may be involved in age-related OA development.
Collapse
Affiliation(s)
- R Wiegertjes
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - N G M Thielen
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A P M van Caam
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M van Laar
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - H M van Beuningen
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M I Koenders
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - P L E M van Lent
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - P M van der Kraan
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - F A J van de Loo
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - E N Blaney Davidson
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
25
|
Cherifi C, Monteagudo S, Lories RJ. Promising targets for therapy of osteoarthritis: a review on the Wnt and TGF-β signalling pathways. Ther Adv Musculoskelet Dis 2021; 13:1759720X211006959. [PMID: 33948125 PMCID: PMC8053758 DOI: 10.1177/1759720x211006959] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/10/2021] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic joint disorder worldwide, with a high personal burden for the patients and an important socio-economic impact. Current therapies are largely limited to pain management and rehabilitation and exercise strategies. For advanced cases, joint replacement surgery may be the only option. Hence, there is an enormous need for the development of effective and safe disease-modifying anti-OA drugs. A strong focus in OA research has been on the identification and role of molecular signalling pathways that contribute to the balance between anabolism and catabolism in the articular cartilage. In this context, most insights have been gained in understanding the roles of the transforming growth factor-beta (TGF-β) and the Wingless-type (Wnt) signalling cascades. The emerging picture demonstrates a high degree of complexity with context-dependent events. TGF-β appears to protect cartilage under healthy conditions, but shifts in its receptor use and subsequent downstream signalling may be deleterious in aged individuals or in damaged cartilage. Likewise, low levels of Wnt activity appear important to sustain chondrocyte viability but excessive activation is associated with progressive joint damage. Emerging clinical data suggest some potential for the use of sprifermin, a recombinant forms of fibroblast growth factor 18, a distant TGF-β superfamily member, and for lorecivivint, a Wnt pathway modulator.
Collapse
Affiliation(s)
- Chahrazad Cherifi
- Department of Development and Regeneration, KU Leuven, Skeletal Biology and Engineering Research Centre, Leuven, Belgium
| | - Silvia Monteagudo
- Department of Development and Regeneration, KU Leuven, Skeletal Biology and Engineering Research Centre, Leuven, Belgium
| | - Rik J Lories
- Department of Development and Regeneration, KU Leuven, Skeletal Biology and Engineering Research Centre, Box 813 O&N, Herestraat 49, Leuven 3000, Belgium; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
26
|
He Y, Makarczyk MJ, Lin H. Role of mitochondria in mediating chondrocyte response to mechanical stimuli. Life Sci 2020; 263:118602. [PMID: 33086121 PMCID: PMC7736591 DOI: 10.1016/j.lfs.2020.118602] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/22/2020] [Accepted: 10/11/2020] [Indexed: 12/21/2022]
Abstract
As the most common form of arthritis, osteoarthritis (OA) has become a major cause of severe joint pain, physical disability, and quality of life impairment in the affected population. To date, precise pathogenesis of OA has not been fully clarified, which leads to significant obstacles in developing efficacious treatments such as failures in finding disease-modifying OA drugs (DMOADs) in the last decades. Given that diarthrodial joints primarily display the weight-bearing and movement-supporting function, it is not surprising that mechanical stress represents one of the major risk factors for OA. However, the inner connection between mechanical stress and OA onset/progression has yet to be explored. Mitochondrion, a widespread organelle involved in complex biological regulation processes such as adenosine triphosphate (ATP) synthesis and cellular metabolism, is believed to have a controlling role in the survival and function implement of chondrocytes, the singular cell type within cartilage. Mitochondrial dysfunction has also been observed in osteoarthritic chondrocytes. In this review, we systemically summarize mitochondrial alterations in chondrocytes during OA progression and discuss our recent progress in understanding the potential role of mitochondria in mediating mechanical stress-associated osteoarthritic alterations of chondrocytes. In particular, we propose the potential signaling pathways that may regulate this process, which provide new views and therapeutic targets for the prevention and treatment of mechanical stress-associated OA.
Collapse
Affiliation(s)
- Yuchen He
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Meagan J Makarczyk
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
27
|
Zhao X, Meng F, Hu S, Yang Z, Huang H, Pang R, Wen X, Kang Y, Zhang Z. The Synovium Attenuates Cartilage Degeneration in KOA through Activation of the Smad2/3-Runx1 Cascade and Chondrogenesis-related miRNAs. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:832-845. [PMID: 33230479 PMCID: PMC7658376 DOI: 10.1016/j.omtn.2020.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022]
Abstract
Knee osteoarthritis (KOA) is a highly prevalent disabling joint disease in aged people. Progressive cartilage degradation is the hallmark of KOA, but its deeper mechanism remains unclear. Substantial evidence indicates the importance of the synovium for joint homeostasis. The present study aimed to determine whether the synovium regulates cartilage metabolism through chondrogenesis-related microRNAs (miRNAs) in the KOA microenvironment. Clinical sample testing and in vitro cell experiments screened out miR-455 and miR-210 as effective miRNAs. The levels of both were significantly reduced in KOA cartilage but increased in KOA synovial fluid compared with controls. We further revealed that transforming growth factor β1 (TGF-β1) can significantly upregulate miR-455 and miR-210 expression in synoviocytes. The upregulated miRNAs can be secreted into the extracellular environment and prevent cartilage degeneration. Through bioinformatics and in vitro experiments, we found that Runx1 can bind to the promoter regions of miR-455 and miR-210 and enhance their transcription in TGF-β1-treated synoviocytes. Collectively, our findings demonstrate a protective effect of the synovium against cartilage degeneration mediated by chondrogenesis-related miRNAs, which suggests that Runx1 is a potential target for KOA therapy.
Collapse
Affiliation(s)
- Xiaoyi Zhao
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong 510080, PR China
| | - Fangang Meng
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong 510080, PR China
| | - Shu Hu
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
| | - Zibo Yang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong 510080, PR China
| | - Hao Huang
- Department of Laboratory Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Rui Pang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, PR China
| | - Xingzhao Wen
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong 510080, PR China
| | - Yan Kang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong 510080, PR China
| | - Zhiqi Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong 510080, PR China
| |
Collapse
|
28
|
Harnessing Mechanosensation in Next Generation Cardiovascular Tissue Engineering. Biomolecules 2020; 10:biom10101419. [PMID: 33036467 PMCID: PMC7599461 DOI: 10.3390/biom10101419] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022] Open
Abstract
The ability of the cells to sense mechanical cues is an integral component of ”social” cell behavior inside tissues with a complex architecture. Through ”mechanosensation” cells are in fact able to decrypt motion, geometries and physical information of surrounding cells and extracellular matrices by activating intracellular pathways converging onto gene expression circuitries controlling cell and tissue homeostasis. Additionally, only recently cell mechanosensation has been integrated systematically as a crucial element in tissue pathophysiology. In the present review, we highlight some of the current efforts to assess the relevance of mechanical sensing into pathology modeling and manufacturing criteria for a next generation of cardiovascular tissue implants.
Collapse
|
29
|
Carballo CB, Coelho TRP, de Holanda Afonso RC, Faria JCDO, Alves T, Monte SM, Ventura Matioszek GM, Moura-Neto V, de Brito JM. Osteoarthritic Synovial Fluid and TGF-β1 Induce Interleukin-18 in Articular Chondrocytes. Cartilage 2020; 11:385-394. [PMID: 30146893 PMCID: PMC7298592 DOI: 10.1177/1947603518796149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Synovial fluid (SF) plays an important role in the maintenance of articular cartilage. SF is a dynamic reservoir of proteins derived from cartilage and synovial tissue; thus, its composition may serve as a biomarker that reflects the health and pathophysiological condition of the joint. The purpose of the current study was to evaluate the osteoarthritic synovial fluid (OASF) and transforming growth factor-β1 (TGF-β1) activity in articular chondrocytes catabolic and inflammatory responses. DESIGN Chondrocytes were seeded at passage 2 and cultured for 72 hours under different conditions. Human chondrocytes were subjected to OASF while rat chondrocytes were subjected to either healthy synovial fluid (rSF) or TGF-β1 and then assigned for cell viability analysis. In addition, the effects of OASF and TGF-β1 on chondrocytes metalloprotease (MMP)-3 and MMP-13 and interleukin-18 (IL-18) expression were evaluated by immunocytochemistry, ELISA, and reverse transcriptase-polymerase chain reaction. RESULTS SF from osteoarthritic patients significantly induced MMP-3, MMP-13, and IL-18 receptor expression in chondrocytes. To put in evidence the inflammatory activity of OASF, healthy chondrocytes from rat were cultured with TGF-β1. In the presence of TGF-β1 these cells started to express MMP-3, MMP-13, and IL-18 genes and attached to each other forming a chondrocyte aggregated structure. Healthy SF was able to maintain a typical monolayer of rounded chondrocytes with no inflammatory response. CONCLUSION In summary, these observations demonstrated that TGF-β1, one of the components of OASF, has a dual effect, acting in chondrocyte maintenance and also inducing inflammatory and catabolic properties of these cells.
Collapse
Affiliation(s)
- Camila B. Carballo
- Programa de Pós-graduação em Anatomia
Patológica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,Orthopaedic Soft Tissue Research
Program, Hospital for Special Surgery, New York, NY, USA
| | - Thiago R. P. Coelho
- Programa de Pós-graduação em Anatomia
Patológica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Tercia Alves
- Instituto de Ciências Biomédicas,
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Samylla M. Monte
- Instituto de Ciências Biomédicas,
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Vivaldo Moura-Neto
- Instituto de Ciências Biomédicas,
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - José M. de Brito
- Instituto de Ciências Biomédicas,
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,José M. de Brito, Universidade Federal do
Rio de Janeiro, Instituto de Ciências Biomédicas, Av. Carlos Chagas Filho 373,
Bloco F2-01, Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
30
|
Johnstone B, Stoddart MJ, Im GI. Multi-Disciplinary Approaches for Cell-Based Cartilage Regeneration. J Orthop Res 2020; 38:463-472. [PMID: 31478253 DOI: 10.1002/jor.24458] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 08/23/2019] [Indexed: 02/04/2023]
Abstract
Articular cartilage does not regenerate in adults. A lot of time and resources have been dedicated to cartilage regeneration research. The current understanding suggests that multi-disciplinary approach including biologic, genetic, and mechanical stimulations may be needed for cell-based cartilage regeneration. This review summarizes contents of a workshop sponsored by International Combined Orthopaedic Societies during the 2019 annual meeting of the Orthopaedic Research Society held in Austin, Texas. Three approaches for cell-based cartilage regeneration were introduced, including cellular basis of chondrogenesis, gene-enhanced cartilage regeneration, and physical modulation to divert stem cells to chondrogenic cell fate. While the complicated nature of cartilage regeneration has not allowed us to achieve successful regeneration of hyaline articular cartilage so far, the utilization of multi-disciplinary approaches in various fields of biomedical engineering will provide means to achieve this goal faster. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:463-472, 2020.
Collapse
Affiliation(s)
- Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, Oregon
| | | | - Gun-Il Im
- Integrative Research Institute for Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| |
Collapse
|
31
|
Dennis JE, Splawn T, Kean TJ. High-Throughput, Temporal and Dose Dependent, Effect of Vitamins and Minerals on Chondrogenesis. Front Cell Dev Biol 2020; 8:92. [PMID: 32161755 PMCID: PMC7053227 DOI: 10.3389/fcell.2020.00092] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Tissue engineered hyaline cartilage is plagued by poor mechanical properties largely due to inadequate type II collagen expression. Of note, commonly used defined chondrogenic media lack 14 vitamins and minerals, some of which are implicated in chondrogenesis. Type II collagen promoter-driven Gaussia luciferase was transfected into ATDC5 cells to create a chondrogenic cell with a secreted-reporter. The reporter cells were used in an aggregate-based chondrogenic culture model to develop a high-throughput analytic platform. This high-throughput platform was used to assess the effect of vitamins and minerals, alone and in combination with TGFβ1, on COL2A1 promoter-driven expression. Significant combinatorial effects between vitamins, minerals, and TGFβ1 in terms of COL2A1 promoter-driven expression and metabolism were discovered. An “optimal” continual supplement of copper and vitamin K in the presence of TGFβ1 gave a 2.5-fold increase in COL2A1 promoter-driven expression over TGFβ1 supplemented media alone in ATDC5 cells.
Collapse
Affiliation(s)
- James E Dennis
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Taylor Splawn
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Thomas J Kean
- Biionix Cluster, Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
32
|
Thielen NGM, van der Kraan PM, van Caam APM. TGFβ/BMP Signaling Pathway in Cartilage Homeostasis. Cells 2019; 8:cells8090969. [PMID: 31450621 PMCID: PMC6769927 DOI: 10.3390/cells8090969] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 01/15/2023] Open
Abstract
Cartilage homeostasis is governed by articular chondrocytes via their ability to modulate extracellular matrix production and degradation. In turn, chondrocyte activity is regulated by growth factors such as those of the transforming growth factor β (TGFβ) family. Members of this family include the TGFβs, bone morphogenetic proteins (BMPs), and growth and differentiation factors (GDFs). Signaling by this protein family uniquely activates SMAD-dependent signaling and transcription but also activates SMAD-independent signaling via MAPKs such as ERK and TAK1. This review will address the pivotal role of the TGFβ family in cartilage biology by listing several TGFβ family members and describing their signaling and importance for cartilage maintenance. In addition, it is discussed how (pathological) processes such as aging, mechanical stress, and inflammation contribute to altered TGFβ family signaling, leading to disturbed cartilage metabolism and disease.
Collapse
Affiliation(s)
- Nathalie G M Thielen
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Arjan P M van Caam
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands.
| |
Collapse
|
33
|
Glatt V, Evans CH, Stoddart MJ. Regenerative rehabilitation: The role of mechanotransduction in orthopaedic regenerative medicine. J Orthop Res 2019; 37:1263-1269. [PMID: 30561813 PMCID: PMC6546504 DOI: 10.1002/jor.24205] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/28/2018] [Indexed: 02/04/2023]
Abstract
Regenerative rehabilitation is an emerging area of investigation that seeks to integrate regenerative medicine with rehabilitation medicine. It is based on the realization that combining these two areas of medicine at an early stage of treatment will produce a better clinical outcome than the traditional linear approach of first administering the elements of regeneration followed, after a delay, by rehabilitation. Indeed, in certain settings, a case can be made for initiating rehabilitation protocols before starting regenerative intervention. This review summarizes the contents of a workshop held during the 2018 annual meeting of the Orthopaedic Research Society. It introduced the concept of regenerative rehabilitation and then provided two orthopaedic examples drawn from the domains of cartilage repair and bone healing. Rehabilitation medicine can supply a variety of physical stimuli, including electrical stimulation, thermal stimulation and mechanical stimulation. Of these, mechanical stimulation has the most obvious relevance to orthopaedics. The mechano-responsiveness of cartilage and bone has been known for a long time, but is poorly understood and has led to only limited clinical application. Improved bioreactor designs that allow multi-axial loading enable new insights into the responsiveness of chondrocytes and chondroprogenitor cells to specific types of load, especially shear. Recent studies on the mechanobiology of bone healing show that modulating the mechanical environment of an experimental osseous lesion by a process of "Reverse Dynamization" soon after injury considerably enhances healing. Future studies are needed to probe the molecular mechanisms responsible for these phenomena and to translate these findings into clinical practice. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1263-1269, 2019.
Collapse
Affiliation(s)
- Vaida Glatt
- Department of Orthopaedic Surgery, University of Texas Health Science Center, San Antonio, Texas
| | | | | |
Collapse
|
34
|
Armiento AR, Alini M, Stoddart MJ. Articular fibrocartilage - Why does hyaline cartilage fail to repair? Adv Drug Deliv Rev 2019; 146:289-305. [PMID: 30605736 DOI: 10.1016/j.addr.2018.12.015] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/07/2018] [Accepted: 12/27/2018] [Indexed: 12/12/2022]
Abstract
Once damaged, articular cartilage has a limited potential to repair. Clinically, a repair tissue is formed, yet, it is often mechanically inferior fibrocartilage. The use of monolayer expanded versus naïve cells may explain one of the biggest discrepancies in mesenchymal stromal/stem cell (MSC) based cartilage regeneration. Namely, studies utilizing monolayer expanded MSCs, as indicated by numerous in vitro studies, report as a main limitation the induction of type X collagen and hypertrophy, a phenotype associated with endochondral bone formation. However, marrow stimulation and transfer studies report a mechanically inferior collagen I/II fibrocartilage as the main outcome. Therefore, this review will highlight the collagen species produced during the different therapeutic approaches. New developments in scaffold design and delivery of therapeutic molecules will be described. Potential future directions towards clinical translation will be discussed. New delivery mechanisms are being developed and they offer new hope in targeted therapeutic delivery.
Collapse
Affiliation(s)
| | - Mauro Alini
- AO Research Institute Davos, 7270 Davos Platz, Switzerland.
| | | |
Collapse
|
35
|
Niemelä TM, Tulamo RM, Carmona JU, López C. Evaluation of the effect of experimentally induced cartilage defect and intra-articular hyaluronan on synovial fluid biomarkers in intercarpal joints of horses. Acta Vet Scand 2019; 61:24. [PMID: 31146775 PMCID: PMC6543688 DOI: 10.1186/s13028-019-0460-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 05/27/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Inflammatory and degenerative activity inside the joint can be studied in vivo by analysis of synovial fluid biomarkers. In addition to pro-inflammatory mediators, several anabolic and anti-inflammatory substances are produced during the disease process. They counteract the catabolic effects of the pro-inflammatory cytokines and thus diminish the cartilage damage. The response of synovial fluid biomarkers after intra-articular hyaluronan injection, alone or in combination with other substances, has been examined only in a few equine studies. The effects of hyaluronan on some pro-inflammatory mediators, such as prostaglandin E2, have been documented but especially the effects on synovial fluid anti-inflammatory mediators are less studied. In animal models hyaluronan has been demonstrated to reduce pain via protecting nociceptive nerve endings and by blocking pain receptor channels. However, the results obtained for pain-relief of human osteoarthritis are contradictory. The aim of the study was to measure the synovial fluid IL-1ra, PDGF-BB, TGF-β1 and TNF-α concentrations before and after surgically induced cartilage defect, and following intra-articular hyaluronan injection in horses. Eight Standardbred horses underwent bilateral arthroscopic surgeries of their intercarpal joints under general anaesthesia, and cartilage defect was created on the dorsal edge of the third carpal bone of one randomly selected intercarpal joint of each horse. Five days post-surgery, one randomly selected intercarpal joint was injected intra-articular with 3 mL HA (20 mg/mL). RESULTS Operation type had no significant effect on the synovial fluid IL-1ra, PDGF-BB, TGF-β1 and TNF-α concentrations but compared with baseline, synovial fluid IL-1ra and TNF-α concentrations increased. Intra-articular hyaluronan had no significant effect on the biomarker concentrations but a trend of mild improvement in the clinical signs of intra-articular inflammation was seen. CONCLUSIONS Creation of the cartilage defect and sham-operation lead to an increase of synovial fluid IL-1ra and TNF-α concentrations but changes in concentrations of anabolic growth factors TGF-β1 and PDGF-BB could not be documented 5 days after the arthroscopy. Intra-articular hyaluronan was well tolerated. Further research is needed to document possible treatment effects of intra-articular hyaluronan on the synovial fluid biomarkers of inflammation and cartilage metabolism.
Collapse
Affiliation(s)
- Tytti Maaria Niemelä
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, 00014 Helsinki, Finland
| | - Riitta-Mari Tulamo
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, 00014 Helsinki, Finland
| | - Jorge Uriel Carmona
- Grupo de Investigación Terapia Regenerativa, Departamento de Salud Animal, Universidad de Caldas, Calle 65 No26-10, Manizales, Caldas, Colombia
| | - Catalina López
- Grupo de Investigación Terapia Regenerativa, Departamento de Salud Animal, Universidad de Caldas, Calle 65 No26-10, Manizales, Caldas, Colombia
| |
Collapse
|
36
|
Smit Y, Marais HJ, Thompson PN, Mahne AT, Goddard A. Clinical findings, synovial fluid cytology and growth factor concentrations after intra-articular use of a platelet-rich product in horses with osteoarthritis. J S Afr Vet Assoc 2019; 90:e1-e9. [PMID: 31170778 PMCID: PMC6556911 DOI: 10.4102/jsava.v90i0.1721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 02/04/2019] [Accepted: 02/19/2019] [Indexed: 01/06/2023] Open
Abstract
Osteoarthritis is a common cause of lameness in horses, resulting in poor performance. Intra-articular platelet-rich plasma can deliver a collection of bioactive molecules, such as autologous growth factors and proteins involved in the quality of tissue repair. Horses (n=5) with osteoarthritis affecting antebrachiocarpal, middle carpal or metacarpophalangeal joints, and horses (n=5) without osteoarthritis of the corresponding joints (radiographically free of osteoarthritis), were used for the production of platelet-rich plasma which was subsequently injected into selected joints. Clinical and synovial fluid changes after intra-articular injection of platelet-rich plasma as well as synovial platelet-derived growth factor-BB and transforming growth factor-beta 1 concentration changes were evaluated in these joints and compared between normal joints and joints with osteoarthritis. A gravity filtration system produced a moderately concentrated platelet-rich plasma, representing a 4.7-fold increase in baseline platelet concentration. The synovial effusion score was significantly different between the control joints and joints with osteoarthritis on Day 0 with a higher score in the group with osteoarthritis. Within the control group, the synovial effusion score was significantly higher on Days 1 and 2 compared to Day 0. For both groups, the synovial fluid nucleated cell count, predominantly intact neutrophils, was significantly increased on Days 1 and 2, with no significant difference between groups. The mean synovial platelet-derived growth factor-BB and transforming growth factor-beta 1 concentrations were increased for both groups but significantly lowered in the group with osteoarthritis on Day 1 compared to normal joints. Concentrations for platelet-derived growth factor-BB remained unchanged on Day 5, compared to Day 1, with no significant difference between groups. In conclusion, intra-articular treatment with platelet-rich plasma resulted in increased synovial growth factor concentrations in joints but with lower concentrations in joints with osteoarthritis. A transient inflammatory reaction was seen both clinically as an increase in synovial effusion and cytologically in both normal joints and joints with osteoarthritis.
Collapse
Affiliation(s)
- Yolandi Smit
- Department of Companion Animal Clinical Studies, University of Pretoria, Onderstepoort.
| | | | | | | | | |
Collapse
|
37
|
Chen PH, Tang T, Liu C, Wang B, Mian M, Oka C, Baquerizo M, Li Y, Xu L. High-Temperature Requirement A1 Protease as a Rate-Limiting Factor in the Development of Osteoarthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1423-1434. [PMID: 31051168 DOI: 10.1016/j.ajpath.2019.03.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/22/2019] [Accepted: 03/19/2019] [Indexed: 02/05/2023]
Abstract
Preserving the mature articular cartilage of joints is a critical focus in the prevention and treatment of osteoarthritis. We determined whether the genetic inactivation of high-temperature requirement A1 (HtrA1) can significantly attenuate the degradation of articular or condylar cartilage. Two types of mouse models of osteoarthritis were used, a spontaneous mutant mouse model [type XI collagen-haploinsufficient (Col11a1+/-) mice] and two post-traumatic mouse models [destabilization of the medial meniscus (DMM) on the knee and a partial discectomy (PDE) on the temporomandibular joint]. Three different groups of mice were generated: i) HtrA1 was genetically deleted from Col11a1+/- mice (HtrA1-/-;Col11a1+/-), ii) HtrA1-deficient mice (HtrA1-/-) were subjected to DMM, and iii) HtrA1-/- mice were subjected to PDE. Knee and temporomandibular joints from the mice were characterized for evidence of cartilage degeneration. The degradation of articular or condylar cartilage was significantly delayed in HtrA1-/-;Col11a1+/- mice and HtrA1-/- mice after DMM or PDE. The amount of collagen type VI was significantly higher in the articular cartilage in HtrA1-/-;Col11a1+/- mice, compared with that in Col11a1+/- mice. The genetic removal of HtrA1 may delay the degradation of articular or condylar cartilage in mice.
Collapse
Affiliation(s)
- Peter H Chen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Tian Tang
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenlu Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Beiyu Wang
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Michelle Mian
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Chio Oka
- Division of Gene Function in Animals, Nara Institute of Science and Technology, Ikoma, Japan
| | - Maria Baquerizo
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Yefu Li
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; Faculty of Medicine, Harvard Medical School, Boston, Massachusetts.
| | - Lin Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; Faculty of Medicine, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
38
|
Oscillatory shear potentiates latent TGF-β1 activation more than steady shear as demonstrated by a novel force generator. Sci Rep 2019; 9:6065. [PMID: 30988341 PMCID: PMC6465594 DOI: 10.1038/s41598-019-42302-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/28/2019] [Indexed: 11/20/2022] Open
Abstract
Cardiovascular mechanical stresses trigger physiological and pathological cellular reactions including secretion of Transforming Growth Factor β1 ubiquitously in a latent form (LTGF-β1). While complex shear stresses can activate LTGF-β1, the mechanisms underlying LTGF-β1 activation remain unclear. We hypothesized that different types of shear stress differentially activate LTGF-β1. We designed a custom-built cone-and-plate device to generate steady shear (SS) forces, which are physiologic, or oscillatory shear (OSS) forces characteristic of pathologic states, by abruptly changing rotation directions. We then measured LTGF-β1 activation in platelet releasates. We modeled and measured flow profile changes between SS and OSS by computational fluid dynamics (CFD) simulations. We found a spike in shear rate during abrupt changes in rotation direction. OSS activated TGF-β1 levels significantly more than SS at all shear rates. OSS altered oxidation of free thiols to form more high molecular weight protein complex(es) than SS, a potential mechanism of shear-dependent LTGF-β1 activation. Increasing viscosity in platelet releasates produced higher shear stress and higher LTGF-β1 activation. OSS-generated active TGF-β1 stimulated higher pSmad2 signaling and endothelial to mesenchymal transition (EndoMT)-related genes PAI-1, collagen, and periostin expression in endothelial cells. Overall, our data suggest variable TGF-β1 activation and signaling occurs with competing blood flow patterns in the vasculature to generate complex shear stress, which activates higher levels of TGF-β1 to drive vascular remodeling.
Collapse
|
39
|
Hodgson D, Rowan AD, Falciani F, Proctor CJ. Systems biology reveals how altered TGFβ signalling with age reduces protection against pro-inflammatory stimuli. PLoS Comput Biol 2019; 15:e1006685. [PMID: 30677026 PMCID: PMC6363221 DOI: 10.1371/journal.pcbi.1006685] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 02/05/2019] [Accepted: 11/26/2018] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative condition caused by dysregulation of multiple molecular signalling pathways. Such dysregulation results in damage to cartilage, a smooth and protective tissue that enables low friction articulation of synovial joints. Matrix metalloproteinases (MMPs), especially MMP-13, are key enzymes in the cleavage of type II collagen which is a vital component for cartilage integrity. Transforming growth factor beta (TGFβ) can protect against pro-inflammatory cytokine-mediated MMP expression. With age there is a change in the ratio of two TGFβ type I receptors (Alk1/Alk5), a shift that results in TGFβ losing its protective role in cartilage homeostasis. Instead, TGFβ promotes cartilage degradation which correlates with the spontaneous development of OA in murine models. However, the mechanism by which TGFβ protects against pro-inflammatory responses and how this changes with age has not been extensively studied. As TGFβ signalling is complex, we used systems biology to combine experimental and computational outputs to examine how the system changes with age. Experiments showed that the repressive effect of TGFβ on chondrocytes treated with a pro-inflammatory stimulus required Alk5. Computational modelling revealed two independent mechanisms were needed to explain the crosstalk between TGFβ and pro-inflammatory signalling pathways. A novel meta-analysis of microarray data from OA patient tissue was used to create a Cytoscape network representative of human OA and revealed the importance of inflammation. Combining the modelled genes with the microarray network provided a global overview into the crosstalk between the different signalling pathways involved in OA development. Our results provide further insights into the mechanisms that cause TGFβ signalling to change from a protective to a detrimental pathway in cartilage with ageing. Moreover, such a systems biology approach may enable restoration of the protective role of TGFβ as a potential therapy to prevent age-related loss of cartilage and the development of OA.
Collapse
Affiliation(s)
- David Hodgson
- Institute of Cellular Medicine, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
| | - Andrew D. Rowan
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Francesco Falciani
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Carole J. Proctor
- Institute of Cellular Medicine, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Sarem M, Otto O, Tanaka S, Shastri VP. Cell number in mesenchymal stem cell aggregates dictates cell stiffness and chondrogenesis. Stem Cell Res Ther 2019; 10:10. [PMID: 30630531 PMCID: PMC6329065 DOI: 10.1186/s13287-018-1103-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/03/2018] [Accepted: 12/09/2018] [Indexed: 01/09/2023] Open
Abstract
Background Although mesenchymal stem/stromal cell (MSC) chondrogenic differentiation has been thoroughly investigated, the rudiments for enhancing chondrogenesis have remained largely dependent on external cues. Focus to date has been on extrinsic variables such as soluble signals, culture conditions (bioreactors), and mechanical stimulation. However, the role of intrinsic mechanisms of MSC programming-based mechanobiology remains to be explored. Since aggregation of MSCs, a prerequisite for chondrogenesis, generates tension within the cell agglomerate, we inquired if the initial number of cells forming the aggregate (aggregate cell number (ACN)) can impact chondrogenesis. Methods Aggregates of varying ACN were formed using well-established centrifugation approach. Progression of chondrogenic differentiation in the aggregates was assessed over 3 weeks in presence and absence of transforming growth factor-beta 1 (TGF-β1). Mechanical properties of the cells were characterized using high-throughput real-time deformability cytometry (RT-DC), and gene expression was analyzed using Affymetrix gene array. Expression of molecular markers linked to chondrogenesis was assessed using western blot and immunofluorescence. Results Reducing ACN from 500 k to 70 k lead to activation and acceleration of the chondrogenic differentiation, independent of soluble chondro-inductive factors, which involves changes to β-catenin-dependent TCF/LEF transcriptional activity and expression of anti-apoptotic protein survivin. RT-DC analysis revealed that stiffness and size of cells within aggregates are modulated by ACN. A direct correlation between progression of chondrogenesis and emergence of stiffer cell phenotype was found. Affymetrix gene array analysis revealed a downregulation of genes associated with lipid synthesis and regulation, which could account for observed changes in cell stiffness. Immunofluorescence and western blot analysis revealed that increasing ACN upregulates the expression of lipid raft protein caveolin-1, a β-catenin binding partner, and downregulates the expression of N-cadherin. As a demonstration of the relevance of these findings in MSC-based strategies for skeletal repair, it is shown that implanting aggregates within collagenous matrix not only decreases the necessity for high cell numbers but also leads to marked improvement in the quality of the deposited tissue. Conclusions This study presents a simple and donor-independent strategy to enhance the efficiency of MSC chondrogenic differentiation and identifies changes in cell mechanics coincident with MSC chondrogenesis with potential translational applications. Electronic supplementary material The online version of this article (10.1186/s13287-018-1103-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melika Sarem
- Institute for Macromolecular Chemistry, University of Freiburg, Stefan-Meier Str.31, 79104, Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany.,Helmholtz Virtual Institute on Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513, Teltow, Germany
| | - Oliver Otto
- Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University of Greifswald, Fleischmannstr. 42-44, 17489, Greifswald, Germany
| | - Simon Tanaka
- Computational Biology Group, D-BSSE, ETH Zürich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - V Prasad Shastri
- Institute for Macromolecular Chemistry, University of Freiburg, Stefan-Meier Str.31, 79104, Freiburg, Germany. .,BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany. .,Helmholtz Virtual Institute on Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513, Teltow, Germany.
| |
Collapse
|
41
|
|
42
|
Remes Lenicov F, Paletta AL, Gonzalez Prinz M, Varese A, Pavillet CE, Lopez Malizia Á, Sabatté J, Geffner JR, Ceballos A. Prostaglandin E2 Antagonizes TGF-β Actions During the Differentiation of Monocytes Into Dendritic Cells. Front Immunol 2018; 9:1441. [PMID: 29988364 PMCID: PMC6023975 DOI: 10.3389/fimmu.2018.01441] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/11/2018] [Indexed: 12/27/2022] Open
Abstract
Inflammatory dendritic cells (DCs) are a distinct subset of DCs that derive from circulating monocytes infiltrating injured tissues. Monocytes can differentiate into DCs with different functional signatures, depending on the presence of environment stimuli. Among these stimuli, transforming growth factor-beta (TGF-β) and prostaglandin E2 (PGE2) have been shown to modulate the differentiation of monocytes into DCs with different phenotypes and functional profiles. In fact, both mediators lead to contrasting outcomes regarding the production of inflammatory and anti-inflammatory cytokines. Previously, we have shown that human semen, which contains high concentrations of PGE2, promoted the differentiation of DCs into a tolerogenic profile through a mechanism dependent on signaling by E-prostanoid receptors 2 and 4. Notably, this effect was induced despite the huge concentration of TGF-β present in semen, suggesting that PGE2 overrides the influence exerted by TGF-β. No previous studies have analyzed the joint actions induced by PGE2 and TGF-β on the function of monocytes or DCs. Here, we analyzed the phenotype and functional profile of monocyte-derived DCs differentiated in the presence of TGF-β and PGE2. DC differentiation guided by TGF-β alone enhanced the expression of CD1a and abrogated LPS-induced expression of IL-10, while differentiation in the presence of PGE2 impaired CD1a expression, preserved CD14 expression, abrogated IL-12 and IL-23 production, stimulated IL-10 production, and promoted the expansion of FoxP3+ regulatory T cells in a mixed lymphocyte reaction. Interestingly, DCs differentiated in the presence of TGF-β and PGE2 showed a phenotype and functional profile closely resembling those induced by PGE2 alone. Finally, we found that PGE2 inhibited TGF-β signaling through an action exerted by EP2 and EP4 receptors coupled to cyclic AMP increase and protein kinase A activity. These results indicate that PGE2 suppresses the influence exerted by TGF-β during DC differentiation, imprinting a tolerogenic signature. High concentrations of TGF-β and PGE2 are usually found in infectious, autoimmune, and neoplastic diseases. Our observations suggest that in these scenarios PGE2 might play a mandatory role in the acquisition of a regulatory profile by DCs.
Collapse
Affiliation(s)
- Federico Remes Lenicov
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana Luz Paletta
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melina Gonzalez Prinz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Augusto Varese
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Clara E Pavillet
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Álvaro Lopez Malizia
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Sabatté
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jorge Raul Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana Ceballos
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
43
|
Krajewska-Włodarczyk M, Owczarczyk-Saczonek A, Placek W, Osowski A, Wojtkiewicz J. Articular Cartilage Aging-Potential Regenerative Capacities of Cell Manipulation and Stem Cell Therapy. Int J Mol Sci 2018; 19:E623. [PMID: 29470431 PMCID: PMC5855845 DOI: 10.3390/ijms19020623] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 02/11/2018] [Accepted: 02/16/2018] [Indexed: 12/13/2022] Open
Abstract
Changes in articular cartilage during the aging process are a stage of natural changes in the human body. Old age is the major risk factor for osteoarthritis but the disease does not have to be an inevitable consequence of aging. Chondrocytes are particularly prone to developing age-related changes. Changes in articular cartilage that take place in the course of aging include the acquisition of the senescence-associated secretory phenotype by chondrocytes, a decrease in the sensitivity of chondrocytes to growth factors, a destructive effect of chronic production of reactive oxygen species and the accumulation of the glycation end products. All of these factors affect the mechanical properties of articular cartilage. A better understanding of the underlying mechanisms in the process of articular cartilage aging may help to create new therapies aimed at slowing or inhibiting age-related modifications of articular cartilage. This paper presents the causes and consequences of cellular aging of chondrocytes and the biological therapeutic outlook for the regeneration of age-related changes of articular cartilage.
Collapse
Affiliation(s)
- Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, Municipal Hospital in Olsztyn, 10-900 Olsztyn, Poland.
- Department of Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Waldemar Placek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Adam Osowski
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Laboratory for Regenerative Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| |
Collapse
|
44
|
Liao Z, Xing Z, Chen Y, Deng Z, Wu D, Zhao L. [Intra-articular injection of ascorbic acid/ferric chloride relieves cartilage degradation in rats with osteoarthritis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:62-68. [PMID: 33177019 DOI: 10.3969/j.issn.1673-4254.2018.01.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To assess the effect of ascorbic acid/ferric chloride (AA/FeCl3) in attenuating cartilage damage in rats with osteoarthritis. METHODS Thirty adult male Wistar rats with surgically induced osteoarthritis were randomized into 2 groups for treatment with intra-articular injection of saline (control group) or AA/FeCl3 mixture (AA group) once a week starting from the third week after the operation. At 6, 9, and 12 weeks after the operation, 5 rats from each group were sacrificed for observing subchondral bone changes on X-ray films and evaluation of cartilage degeneration in the right knee joints using safranin-O/Fast green staining and a modified OARSI scoring system. The degradation of the cartilage matrix was observed by immunohistochemical staining for type Ⅱ collagen. RESULTS X-ray examination in saline control group revealed the presence of osteophytes and narrowing of the joint space at 9 weeks, and the joint line disappeared at 12 weeks after the surgery; only slight irregularity of the articular surface was observed in the AA group at 9 and 12 weeks. OARSI scores were significantly lower in AA group than in the control group at 9 weeks (18.67±0.67 vs 12.17±2.75; P < 0.05) and 12 weeks (20.11±1.84 vs 13.77± 0.40; P < 0.05) but not at 6 weeks after the surgery. The content of type 2 collagen in AA group was significantly higher than that in the control group at 6 weeks (0.36±0.039 vs 0.49±0.029; P < 0.05) and 9 weeks after the surgery (0.25±0.041 vs 0.38±0.040; P < 0.05). CONCLUSIONS Early intra-articular injection of AA/FeCl3 can effectively delay the progression of post-traumatic osteoarthritis in rats.
Collapse
Affiliation(s)
- Zhenting Liao
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhenquan Xing
- Department of Spinal Surgery, Sanya People's Hospital, Sanya 572000, China
| | - Yufan Chen
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhonghao Deng
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Desheng Wu
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Liang Zhao
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
45
|
Jacobsen MM, Tokareva OS, Ebrahimi D, Huang W, Ling S, Dinjaski N, Li D, Simon M, Staii C, Buehler MJ, Kaplan DL, Wong JY. Effect of Terminal Modification on the Molecular Assembly and Mechanical Properties of Protein-Based Block Copolymers. Macromol Biosci 2017; 17:10.1002/mabi.201700095. [PMID: 28665510 PMCID: PMC5600892 DOI: 10.1002/mabi.201700095] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/03/2017] [Indexed: 01/13/2023]
Abstract
Accurate prediction and validation of the assembly of bioinspired peptide sequences into fibers with defined mechanical characteristics would aid significantly in designing and creating materials with desired properties. This process may also be utilized to provide insight into how the molecular architecture of many natural protein fibers is assembled. In this work, computational modeling and experimentation are used in tandem to determine how peptide terminal modification affects a fiber-forming core domain. Modeling shows that increased terminal molecular weight and hydrophilicity improve peptide chain alignment under shearing conditions and promote consolidation of semicrystalline domains. Mechanical analysis shows acute improvements to strength and elasticity, but significantly reduced extensibility and overall toughness. These results highlight an important entropic function that terminal domains of fiber-forming peptides exhibit as chain alignment promoters, which ultimately has notable consequences on the mechanical behavior of the final fiber products.
Collapse
Affiliation(s)
- Matthew M Jacobsen
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Olena S Tokareva
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Davoud Ebrahimi
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Wenwen Huang
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Shengjie Ling
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Nina Dinjaski
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - David Li
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Marc Simon
- Department of Physics and Astronomy, Center for Nanoscopic Physics, Tufts University, Medford, MA, 02155, USA
| | - Cristian Staii
- Department of Physics and Astronomy, Center for Nanoscopic Physics, Tufts University, Medford, MA, 02155, USA
| | - Markus J Buehler
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Joyce Y Wong
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Division of Materials Science and Engineering, Boston University, Boston, MA, 02215, USA
| |
Collapse
|
46
|
Estell EG, Murphy LA, Silverstein AM, Tan AR, Shah RP, Ateshian GA, Hung CT. Fibroblast-like synoviocyte mechanosensitivity to fluid shear is modulated by interleukin-1α. J Biomech 2017; 60:91-99. [PMID: 28716465 PMCID: PMC5788292 DOI: 10.1016/j.jbiomech.2017.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/10/2017] [Accepted: 06/13/2017] [Indexed: 11/29/2022]
Abstract
Fibroblast-like synoviocytes (FLS) reside in the synovial membrane of diarthrodial joints and are exposed to a dynamic fluid environment that presents both physical and chemical stimuli. The ability of FLS to sense and respond to these stimuli plays a key role in their normal function, and is implicated in the alterations to function that occur in osteoarthritis (OA). The present work characterizes the response of FLS to fluid flow-induced shear stress via real-time calcium imaging, and tests the hypothesis that this response is modulated by interleukin-1α (IL-1α), a cytokine elevated in OA. FLS demonstrated a robust calcium signaling response to fluid shear that was dose dependent upon stress level and required both external and internal calcium sources. Preconditioning with 10ng/mL IL-1α for 24h heightened this shear stress response by significantly increasing the percent of responding cells and peak magnitude, while significantly decreasing the time for a peak to occur. Intercellular communication via gap junctions was found to account for a portion of the FLS population response in normal conditions, and was significantly increased by IL-1α preconditioning. IL-1α was also found to significantly increase average length and incidence of the primary cilium, an organelle commonly implicated in shear mechanosensing. These findings suggest that the elevated levels of IL-1α found in the OA environment heighten FLS sensitivity to fluid shear by altering both intercellular communication and individual cell sensitivity, which could affect downstream functions and contribute to progression of the disease state.
Collapse
Affiliation(s)
- Eben G Estell
- Columbia University, Department of Biomedical Engineering, New York, NY, United States
| | - Lance A Murphy
- Columbia University, Department of Biomedical Engineering, New York, NY, United States
| | - Amy M Silverstein
- Columbia University, Department of Biomedical Engineering, New York, NY, United States
| | - Andrea R Tan
- Columbia University, Department of Biomedical Engineering, New York, NY, United States
| | - Roshan P Shah
- Columbia University, Department of Orthopedic Surgery, New York, NY, United States
| | - Gerard A Ateshian
- Columbia University, Department of Biomedical Engineering, New York, NY, United States
| | - Clark T Hung
- Columbia University, Department of Biomedical Engineering, New York, NY, United States.
| |
Collapse
|
47
|
Yuan X, Wei Y, Villasante A, Ng JJD, Arkonac DE, Chao PHG, Vunjak-Novakovic G. Stem cell delivery in tissue-specific hydrogel enabled meniscal repair in an orthotopic rat model. Biomaterials 2017; 132:59-71. [PMID: 28407495 PMCID: PMC5473162 DOI: 10.1016/j.biomaterials.2017.04.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/28/2017] [Accepted: 04/03/2017] [Indexed: 01/21/2023]
Abstract
Interest in non-invasive injectable therapies has rapidly risen due to their excellent safety profile and ease of use in clinical settings. Injectable hydrogels can be derived from the extracellular matrix (ECM) of specific tissues to provide a biomimetic environment for cell delivery and enable seamless regeneration of tissue defects. We investigated the in situ delivery of human mesenchymal stem cells (hMSCs) in decellularized meniscus ECM hydrogel to a meniscal defect in a nude rat model. First, decellularized meniscus ECM hydrogel retained tissue-specific proteoglycans and collagens, and significantly upregulated expression of fibrochondrogenic markers by hMSCs versus collagen hydrogel alone in vitro. The meniscus ECM hydrogel in turn supported delivery of hMSCs for integrative repair of a full-thickness defect model in meniscal explants after in vitro culture and in vivo subcutaneous implantation. When applied to an orthotopic model of meniscal injury in nude rat, hMSCs in meniscus ECM hydrogel were retained out to eight weeks post-injection, contributing to tissue regeneration and protection from joint space narrowing, pathologic mineralization, and osteoarthritis development, as evidenced by macroscopic and microscopic image analysis. Based on these findings, we propose the use of tissue-specific meniscus ECM-derived hydrogel for the delivery of therapeutic hMSCs to treat meniscal injury.
Collapse
Affiliation(s)
- Xiaoning Yuan
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yiyong Wei
- Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Aránzazu Villasante
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Johnathan J D Ng
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Derya E Arkonac
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Pen-Hsiu Grace Chao
- Institute of Biomedical Engineering, School of Medicine and School of Engineering, National Taiwan University, Taipei, Taiwan
| | | |
Collapse
|
48
|
Szychlinska MA, Stoddart MJ, D'Amora U, Ambrosio L, Alini M, Musumeci G. Mesenchymal Stem Cell-Based Cartilage Regeneration Approach and Cell Senescence: Can We Manipulate Cell Aging and Function? TISSUE ENGINEERING PART B-REVIEWS 2017; 23:529-539. [PMID: 28514935 DOI: 10.1089/ten.teb.2017.0083] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aging is the most prominent risk factor triggering several degenerative diseases, such as osteoarthritis (OA). Due to its poor self-healing capacity, once injured cartilage needs to be reestablished. This process might be approached through resorting to cell-based therapies and/or tissue engineering. Human mesenchymal stem cells (MSCs) represent a promising approach due to their chondrogenic differentiation potential. Presently, in vitro chondrogenic differentiation of MSCs is limited by two main reasons as follows: aging of MSCs, which determines the loss of cell proliferative and differentiation capacity and MSC-derived chondrocyte hypertrophic differentiation, which limits the use of these cells in cartilage tissue regeneration approach. The effect of aging on MSCs is fundamental for stem cell-based therapy development, especially in older subjects. In the present review we focus on homeostasis alterations occurring in MSC-derived chondrocytes during in vitro aging. Moreover, we deal with potential cell aging regulation approaches, such as cell stimulation through telomerase activators, mechanical strain, and epigenetic regulation. Future investigations in this field might provide new insights into innovative strategies for cartilage regeneration and potentially inspire novel therapeutic approaches for OA treatment.
Collapse
Affiliation(s)
- Marta A Szychlinska
- 1 Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Catania, Italy
| | - Martin J Stoddart
- 2 Musculoskeletal Regeneration, AO Research Institute Davos , Davos Platz, Switzerland
| | - Ugo D'Amora
- 3 Institute of Polymers , Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Luigi Ambrosio
- 3 Institute of Polymers , Composites and Biomaterials, National Research Council of Italy, Naples, Italy .,4 Department of Chemical Science and Materials Technology, National Research Council of Italy , Rome, Italy
| | - Mauro Alini
- 2 Musculoskeletal Regeneration, AO Research Institute Davos , Davos Platz, Switzerland
| | - Giuseppe Musumeci
- 1 Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Catania, Italy .,5 Department of Health, Institut des Etudes Universitaries , UniPoliSI, Veyras, Switzerland
| |
Collapse
|
49
|
Scholtes S, Krämer E, Weisser M, Roth W, Luginbühl R, Grossner T, Richter W. Global chondrocyte gene expression after a single anabolic loading period: Time evolution and re-inducibility of mechano-responses. J Cell Physiol 2017; 233:699-711. [PMID: 28369921 DOI: 10.1002/jcp.25933] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 03/24/2017] [Indexed: 11/09/2022]
Abstract
Aim of this study was a genome-wide identification of mechano-regulated genes and candidate pathways in human chondrocytes subjected to a single anabolic loading episode and characterization of time evolution and re-inducibility of the response. Osteochondral constructs consisting of a chondrocyte-seeded collagen-scaffold connected to β-tricalcium-phosphate were pre-cultured for 35 days and subjected to dynamic compression (25% strain, 1 Hz, 9 × 10 min over 3 hr) before microarray-profiling was performed. Proteoglycan synthesis was determined by 35 S-sulfate-incorporation over 24 hr. Cell viability and hardness of constructs were unaltered by dynamic compression while proteoglycan synthesis was significantly stimulated (1.45-fold, p = 0.016). Among 115 significantly regulated genes, 114 were up-regulated, 48 of them ≥ twofold. AP-1-relevant transcription factors FOSB and FOS strongly increased in line with elevated ERK1/2-phosphorylation and rising MAP3K4 expression. Expression of proteoglycan-synthesizing enzymes CHSY1 and GALNT4 was load-responsive as were factors associated with the MAPK-, TGF-β-, calcium-, retinoic-acid-, Wnt-, and Notch-signaling pathway which were significantly upregulated SOX9, and BMP6 levels rose significantly also after multiple loading episodes at daily intervals even at the 14th cycle with no indication for desensitation. Canonical pSmad2/3 and pSmad1/5/9-signaling showed no consistent regulation. This study associates novel genes with mechanoregulation in chondrocytes, raising SOX9 protein levels with anabolic loading and suggests that more pathways than so far anticipated apparently work together in a complex network of stimulators and feedback-regulators. Upregulation of mechanosensitive indicators extending differentially into the resting time provides crucial knowledge to maximize cartilage matrix deposition for the generation of high-level cartilage replacement tissue.
Collapse
Affiliation(s)
- Simone Scholtes
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | - Elisabeth Krämer
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | - Melanie Weisser
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | - Wolfgang Roth
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | | | - Tobias Grossner
- Department of Orthopaedic and Trauma Surgery, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Wiltrud Richter
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| |
Collapse
|
50
|
Schneider MC, Barnes CA, Bryant SJ. Characterization of the chondrocyte secretome in photoclickable poly(ethylene glycol) hydrogels. Biotechnol Bioeng 2017; 114:2096-2108. [PMID: 28436002 DOI: 10.1002/bit.26320] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/28/2016] [Accepted: 04/17/2017] [Indexed: 12/30/2022]
Abstract
Poly(ethylene glycol) (PEG) hydrogels are highly tunable platforms that are promising cell delivery vehicles for chondrocytes and cartilage tissue engineering. In addition to characterizing the type of extracellular matrix (ECM) that forms, understanding the types of proteins that are secreted by encapsulated cells may be important. Thus, the objectives for this study were to characterize the secretome of chondrocytes encapsulated in PEG hydrogels and determine whether the secretome varies as a function of hydrogel stiffness and culture condition. Bovine chondrocytes were encapsulated in photoclickable PEG hydrogels with a compressive modulus of 8 and 46 kPa and cultured under free swelling or dynamic compressive loading conditions. Cartilage ECM deposition was assessed by biochemical assays and immunohistochemistry. The conditioned medium was analyzed by liquid chromatography-tandem mass spectrometry. Chondrocytes maintained their phenotype within the hydrogels and deposited cartilage-specific ECM that increased over time and included aggrecan and collagens II and VI. Analysis of the secretome revealed a total of 64 proteins, which were largely similar among all experimental conditions. The identified proteins have diverse functions such as biological regulation, response to stress, and collagen fibril organization. Notably, many of the proteins important to the assembly of a collagen-rich cartilage ECM were identified and included collagen types II(α1), VI (α1, α2, and α3), IX (α1), XI (α1 and α2), and biglycan. In addition, many of the other identified proteins have been reported to be present within cell-secreted exosomes. In summary, chondrocytes encapsulated within photoclickable PEG hydrogels secrete many types of proteins that diffuse out of the hydrogel and which have diverse functions, but which are largely preserved across different hydrogel culture environments. Biotechnol. Bioeng. 2017;114: 2096-2108. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Margaret C Schneider
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Campus Box 596, Boulder 80309, Colorado.,Biofrontiers Institute, University of Colorado, Boulder, Colorado
| | | | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Campus Box 596, Boulder 80309, Colorado.,Biofrontiers Institute, University of Colorado, Boulder, Colorado.,Material Science and Engineering Program, University of Colorado, Boulder, Colorado
| |
Collapse
|