1
|
Lee J, Lim J, Park S, Kim S, Park J. Morphologic Response in Femoral Cartilage During and After 40-Minute Treadmill Running. J Athl Train 2024; 59:906-914. [PMID: 39320951 PMCID: PMC11440817 DOI: 10.4085/1062-6050-0659.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
CONTEXT It is unclear whether the response in femoral cartilage to running at different intensities is different. OBJECTIVE To examine the acute patterns of deformation and recovery in femoral cartilage thickness during and after running at different speeds. DESIGN Crossover study. SETTING Laboratory. PATIENTS OR OTHER PARTICIPANTS A total of 17 healthy men (age = 23.9 ± 2.3 years, height = 173.1 ± 5.5 cm, mass = 73.9 ± 8.0 kg). INTERVENTION(S) Participants performed a 40-minute treadmill run at speeds of 7.5 and 8.5 km/h. MAIN OUTCOME MEASURE(S) Ultrasonographic images of femoral cartilage thickness (intercondylar, lateral condyle, and medial condyle) were obtained every 5 minutes during the experiment (40 minutes of running followed by a 60-minute recovery period) at each session. Data were analyzed using analysis of variance and Bonferroni- and Dunnett-adjusted post hoc t tests. To identify patterns of cartilage response, we extracted principal components (PCs) from the cartilage-thickness data using PC analysis, and PC scores were analyzed using t tests. RESULTS Regardless of time, femoral cartilage thicknesses were greater for the 8.5-km/h run than the 7.5-km/h run (intercondylar: F1,656 = 24.73, P < .001, effect size, 0.15; lateral condyle: F1,649 = 16.60, P < .001, effect size, 0.16; medial condyle: F1,649 = 16.55, P < .001, effect size, 0.12). We observed a time effect in intercondylar thickness (F20,656 = 2.15, P = .003), but the Dunnett-adjusted post hoc t test revealed that none of the time point values differed from the baseline value (P > .38 for all comparisons). Although the PC1 and PC2 captured the magnitudes of cartilage thickness and time shift (eg, earlier versus later response), respectively, t tests showed that the PC scores were not different between 7.5 and 8.5 km/h (intercondylar: P ≥ .32; lateral condyle: P ≥ .78; medial condyle: P ≥ .16). CONCLUSIONS Although the 40-minute treadmill run with different speeds produced different levels of fatigue, morphologic differences (<3%) in the femoral cartilage at both speeds seemed to be negligible.
Collapse
Affiliation(s)
- Jinwoo Lee
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| | - Junhyeong Lim
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| | - Sanghyup Park
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| | - Sojin Kim
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| | - Jihong Park
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| |
Collapse
|
2
|
Capuana E, Marino D, Di Gesù R, La Carrubba V, Brucato V, Tuan RS, Gottardi R. A High-Throughput Mechanical Activator for Cartilage Engineering Enables Rapid Screening of in vitro Response of Tissue Models to Physiological and Supra-Physiological Loads. Cells Tissues Organs 2023; 211:670-688. [PMID: 34261061 PMCID: PMC9843549 DOI: 10.1159/000514985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/02/2021] [Indexed: 01/25/2023] Open
Abstract
Articular cartilage is crucially influenced by loading during development, health, and disease. However, our knowledge of the mechanical conditions that promote engineered cartilage maturation or tissue repair is still incomplete. Current in vitro models that allow precise control of the local mechanical environment have been dramatically limited by very low throughput, usually just a few specimens per experiment. To overcome this constraint, we have developed a new device for the high throughput compressive loading of tissue constructs: the High Throughput Mechanical Activator for Cartilage Engineering (HiT-MACE), which allows the mechanoactivation of 6 times more samples than current technologies. With HiT-MACE we were able to apply cyclic loads in the physiological (e.g., equivalent to walking and normal daily activity) and supra-physiological range (e.g., injurious impacts or extensive overloading) to up to 24 samples in one single run. In this report, we compared the early response of cartilage to physiological and supra-physiological mechanical loading to the response to IL-1β exposure, a common but rudimentary in vitro model of cartilage osteoarthritis. Physiological loading rapidly upregulated gene expression of anabolic markers along the TGF-β1 pathway. Notably, TGF-β1 or serum was not included in the medium. Supra-physiological loading caused a mild catabolic response while IL-1β exposure drove a rapid anabolic shift. This aligns well with recent findings suggesting that overloading is a more realistic and biomimetic model of cartilage degeneration. Taken together, these findings showed that the application of HiT-MACE allowed the use of larger number of samples to generate higher volume of data to effectively explore cartilage mechanobiology, which will enable the design of more effective repair and rehabilitation strategies for degenerative cartilage pathologies.
Collapse
Affiliation(s)
- Elisa Capuana
- Department of Engineering, University of Palermo, Palermo, Italy,Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Davide Marino
- Department of Engineering, University of Palermo, Palermo, Italy,Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Roberto Di Gesù
- Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Children's Hospital of Philadelphia, and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA,Fondazione Ri.MED, Palermo, Italy
| | - Vincenzo La Carrubba
- Department of Engineering, University of Palermo, Palermo, Italy,INSTM, Palermo Research Unit, Palermo, Italy
| | - Valerio Brucato
- Department of Engineering, University of Palermo, Palermo, Italy
| | - Rocky S. Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Children's Hospital of Philadelphia, and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA,Fondazione Ri.MED, Palermo, Italy,*Riccardo Gottardi,
| |
Collapse
|
3
|
Korhonen RK, Eskelinen ASA, Orozco GA, Esrafilian A, Florea C, Tanska P. Multiscale In Silico Modeling of Cartilage Injuries. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:45-56. [PMID: 37052845 DOI: 10.1007/978-3-031-25588-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Injurious loading of the joint can be accompanied by articular cartilage damage and trigger inflammation. However, it is not well-known which mechanism controls further cartilage degradation, ultimately leading to post-traumatic osteoarthritis. For personalized prognostics, there should also be a method that can predict tissue alterations following joint and cartilage injury. This chapter gives an overview of experimental and computational methods to characterize and predict cartilage degradation following joint injury. Two mechanisms for cartilage degradation are proposed. In (1) biomechanically driven cartilage degradation, it is assumed that excessive levels of strain or stress of the fibrillar or non-fibrillar matrix lead to proteoglycan loss or collagen damage and degradation. In (2) biochemically driven cartilage degradation, it is assumed that diffusion of inflammatory cytokines leads to degradation of the extracellular matrix. When implementing these two mechanisms in a computational in silico modeling workflow, supplemented by in vitro and in vivo experiments, it is shown that biomechanically driven cartilage degradation is concentrated on the damage environment, while inflammation via synovial fluid affects all free cartilage surfaces. It is also proposed how the presented in silico modeling methodology may be used in the future for personalized prognostics and treatment planning of patients with a joint injury.
Collapse
Affiliation(s)
- Rami K Korhonen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland.
| | - Atte S A Eskelinen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Gustavo A Orozco
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Amir Esrafilian
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Cristina Florea
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Petri Tanska
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
4
|
Alizadeh Sardroud H, Wanlin T, Chen X, Eames BF. Cartilage Tissue Engineering Approaches Need to Assess Fibrocartilage When Hydrogel Constructs Are Mechanically Loaded. Front Bioeng Biotechnol 2022; 9:787538. [PMID: 35096790 PMCID: PMC8790514 DOI: 10.3389/fbioe.2021.787538] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/19/2022] Open
Abstract
Chondrocytes that are impregnated within hydrogel constructs sense applied mechanical force and can respond by expressing collagens, which are deposited into the extracellular matrix (ECM). The intention of most cartilage tissue engineering is to form hyaline cartilage, but if mechanical stimulation pushes the ratio of collagen type I (Col1) to collagen type II (Col2) in the ECM too high, then fibrocartilage can form instead. With a focus on Col1 and Col2 expression, the first part of this article reviews the latest studies on hyaline cartilage regeneration within hydrogel constructs that are subjected to compression forces (one of the major types of the forces within joints) in vitro. Since the mechanical loading conditions involving compression and other forces in joints are difficult to reproduce in vitro, implantation of hydrogel constructs in vivo is also reviewed, again with a focus on Col1 and Col2 production within the newly formed cartilage. Furthermore, mechanotransduction pathways that may be related to the expression of Col1 and Col2 within chondrocytes are reviewed and examined. Also, two recently-emerged, novel approaches of load-shielding and synchrotron radiation (SR)–based imaging techniques are discussed and highlighted for future applications to the regeneration of hyaline cartilage. Going forward, all cartilage tissue engineering experiments should assess thoroughly whether fibrocartilage or hyaline cartilage is formed.
Collapse
Affiliation(s)
- Hamed Alizadeh Sardroud
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- *Correspondence: Hamed Alizadeh Sardroud,
| | - Tasker Wanlin
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - B. Frank Eames
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
5
|
Henao-Murillo L, Pastrama MI, Ito K, van Donkelaar CC. The Relationship Between Proteoglycan Loss, Overloading-Induced Collagen Damage, and Cyclic Loading in Articular Cartilage. Cartilage 2021; 13:1501S-1512S. [PMID: 31729263 PMCID: PMC8721617 DOI: 10.1177/1947603519885005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE The interaction between proteoglycan loss and collagen damage in articular cartilage and the effect of mechanical loading on this interaction remain unknown. The aim of this study was to answer the following questions: (1) Is proteoglycan loss dependent on the amount of collagen damage and does it depend on whether this collagen damage is superficial or internal? (2) Does repeated loading further increase the already enhanced proteoglycan loss in cartilage with collagen damage? DESIGN Fifty-six bovine osteochondral plugs were equilibrated in phosphate-buffered saline for 24 hours, mechanically tested in compression for 8 hours, and kept in phosphate-buffered saline for another 48 hours. The mechanical tests included an overloading step to induce collagen damage, creep steps to determine tissue stiffness, and cyclic loading to induce convection. Proteoglycan release was measured before and after mechanical loading, as well as 48 hours post-loading. Collagen damage was scored histologically. RESULTS Histology revealed different collagen damage grades after the application of mechanical overloading. After 48 hours in phosphate-buffered saline postloading, proteoglycan loss increased linearly with the amount of total collagen damage and was dependent on the presence but not the amount of internal collagen damage. In samples without collagen damage, repeated loading also resulted in increased proteoglycan loss. However, repeated loading did not further enhance the proteoglycan loss induced by damaged collagen. CONCLUSION Proteoglycan loss is enhanced by collagen damage and it depends on the presence of internal collagen damage. Cyclic loading stimulates proteoglycan loss in healthy cartilage but does not lead to additional loss in cartilage with damaged collagen.
Collapse
Affiliation(s)
- Lorenza Henao-Murillo
- Orthopaedic Biomechanics, Department of
Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Noord
Brabant, the Netherlands,Department of Electronics and Industrial
Automation, Universidad Autónoma de Manizales, Manizales, Colombia
| | - Maria-Ioana Pastrama
- Orthopaedic Biomechanics, Department of
Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Noord
Brabant, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of
Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Noord
Brabant, the Netherlands
| | - Corrinus C. van Donkelaar
- Orthopaedic Biomechanics, Department of
Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Noord
Brabant, the Netherlands,Corrinus C. van Donkelaar, Orthopaedic
Biomechanics, Department of Biomedical Engineering, Eindhoven University of
Technology, Gemini-Zuid 1.106, P.O. Box 513, Eindhoven, Noord Brabant 5600 MB,
the Netherlands.
| |
Collapse
|
6
|
Sauerland K, Wolf A, Schudok M, Steinmeyer J. A novel model of a biomechanically induced osteoarthritis-like cartilage for pharmacological in vitro studies. J Cell Mol Med 2021; 25:11221-11231. [PMID: 34766430 PMCID: PMC8650028 DOI: 10.1111/jcmm.17044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/28/2021] [Accepted: 10/26/2021] [Indexed: 11/27/2022] Open
Abstract
Excessive pressure or overload induces and aggravates osteoarthritic changes in articular cartilage, but the underlying biomechanical forces are largely ignored in existing pharmacological in vitro models that are used to investigate drugs against osteoarthritis (OA). Here, we introduce a novel in vitro model to perform pathophysiological and pharmacological investigations, in which cartilage explants are subjected to intermittent cyclic pressure, and characterize its ability to mimic OA‐like tissue reactivity. Mechanical loading time‐dependently increased the biosynthesis, content and retention of fibronectin (Fn), whereas collagen metabolism remained unchanged. This protocol upregulated the production and release of proteoglycans (PGs). The release of PGs from explants was significantly inhibited by a matrix metalloproteinase (MMP) inhibitor, suggesting the involvement of such proteinases in the destruction of the model tissue, similar to what is observed in human OA cartilage. In conclusion, the metabolic alterations in our new biomechanical in vitro model are similar to those of early human OA cartilage, and our pharmacological prevalidation with an MMP‐inhibitor supports its value for further in vitro drug studies.
Collapse
Affiliation(s)
- Katrin Sauerland
- Institute for Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Amela Wolf
- Institute for Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Manfred Schudok
- R&D, Drug Metabolism & Pharmacokinetics, Sanofi-Aventis Deutschand GmbH, Frankfurt, Germany
| | - Juergen Steinmeyer
- Institute for Pharmacology and Toxicology, University of Bonn, Bonn, Germany.,Laboratory for Experimental Orthopaedics, Department of Orthopaedics, University of Giessen, Giessen, Germany
| |
Collapse
|
7
|
Otoo B, Li L, Hart DA, Herzog W. Development of a Porcine Model to Assess the Effect of In-Situ Knee Joint Loading On Site-Specific Cartilage Gene Expression. J Biomech Eng 2021; 144:1115048. [PMID: 34318319 DOI: 10.1115/1.4051922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Indexed: 11/08/2022]
Abstract
Cyclic mechanical loading of cartilage induces stresses and fluid flow which are thought to modulate chondrocyte metabolism. The uneven surface, plus the heterogeneity of cartilage within a joint, makes stress and fluid pressure distribution in the tissue non-uniform, and gene expression may vary at different sites as a function of load magnitude, frequency and time. In previous studies, cartilage explants were used for loading tests to investigate biological responses of the cartilage to mechanical loading. In contrast, we used loading tests on intact knee joints, to better reflect the loading conditions in a joint, and thus provide a more physiologically relevant mechanical environment. Gene expression levels in loaded samples for a selection of relevant genes were compared with those of the corresponding unloaded control samples to characterize potential differences. Furthermore, the effect of load magnitude and duration on gene expression levels were investigated. We observed differences in gene expression levels between samples from different sites in the same joint and between corresponding samples from the same site in loaded and unloaded joints. Consistent with previous findings, our results indicate that there is a critical upper and lower threshold of loading for triggering the expression of certain genes. Variations in gene expression levels may reflect the effect of local loading, topography and structure of the cartilage in an intact joint on the metabolic activity of the associated cells.
Collapse
Affiliation(s)
- Baaba Otoo
- Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive, Calgary, Alberta, Canada T2N 1N4
| | - LePing Li
- Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive, Calgary, Alberta, Canada T2N 1N4
| | - David A Hart
- McCaig Institute for Bone and Joint Health, Department of Surgery, University of Calgary, 2500 University Drive, Calgary, Alberta, Canada T2N 1N4; Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, 2500 University Drive, Calgary, Alberta, Canada T2N 1N4
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, 2500 University Drive, Calgary, Alberta, Canada T2N 1N4
| |
Collapse
|
8
|
Lower Limb Movement Pattern Differences Between Males and Females in Squatting and Kneeling. J Appl Biomech 2021; 37:204-214. [PMID: 33690162 DOI: 10.1123/jab.2020-0185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/15/2020] [Accepted: 12/09/2020] [Indexed: 11/18/2022]
Abstract
Movement pattern differences may contribute to differential injury or disease prevalence between individuals. The purpose of this study was to identify lower limb movement patterns in high knee flexion, a risk factor for knee osteoarthritis, and to investigate kinematic differences between males and females, as females typically develop knee osteoarthritis more commonly and severely than males. Lower extremity kinematic data were recorded from 110 participants completing 4 variations of squatting and kneeling. Principal component analysis was used to identify principal movements associated with the largest variability in the sample. Across the tasks, similar principal movements emerged at maximal flexion and during transitions. At maximal flexion, females achieved greater knee flexion, facilitated by a wider base of support, which may alter posterior and lateral tibiofemoral stress. Principal movements also detected differences in movement temporality between males and females. When these temporal differences occur due to alterations in movement velocity and/or acceleration, they may elicit changes in muscle activation and knee joint stress. Movement variability identified in the current study provides a framework for potential modifiable factors in high knee flexion, such as foot position, and suggests that kinematic differences between the sexes may contribute to differences in knee osteoarthritis progression.
Collapse
|
9
|
Takeda Y, Niki Y, Fukuhara Y, Fukuda Y, Udagawa K, Shimoda M, Kikuchi T, Kobayashi S, Harato K, Miyamoto T, Matsumoto M, Nakamura M. Compressive mechanical stress enhances susceptibility to interleukin-1 by increasing interleukin-1 receptor expression in 3D-cultured ATDC5 cells. BMC Musculoskelet Disord 2021; 22:238. [PMID: 33648469 PMCID: PMC7923672 DOI: 10.1186/s12891-021-04095-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 02/17/2021] [Indexed: 12/31/2022] Open
Abstract
Background Mechanical overload applied on the articular cartilage may play an important role in the pathogenesis of osteoarthritis. However, the mechanism of chondrocyte mechanotransduction is not fully understood. The purpose of this study was to assess the effects of compressive mechanical stress on interleukin-1 receptor (IL-1R) and matrix-degrading enzyme expression by three-dimensional (3D) cultured ATDC5 cells. In addition, the implications of transient receptor potential vanilloid 4 (TRPV4) channel regulation in promoting effects of compressive mechanical loading were elucidated. Methods ATDC5 cells were cultured in alginate beads with the growth medium containing insulin-transferrin-selenium and BMP-2 for 6 days. The cultured cell pellet was seeded in collagen scaffolds to produce 3D-cultured constructs. Cyclic compressive loading was applied on the 3D-cultured constructs at 0.5 Hz for 3 h. The mRNA expressions of a disintegrin and metalloproteinases with thrombospondin motifs 4 (ADAMTS4) and IL-1R were determined with or without compressive loading, and effects of TRPV4 agonist/antagonist on mRNA expressions were examined. Immunoreactivities of reactive oxygen species (ROS), TRPV4 and IL-1R were assessed in 3D-cultured ATDC5 cells. Results In 3D-cultured ATDC5 cells, ROS was induced by cyclic compressive loading stress. The mRNA expression levels of ADAMTS4 and IL-1R were increased by cyclic compressive loading, which was mostly prevented by pyrollidine dithiocarbamate. Small amounts of IL-1β upregulated ADAMTS4 and IL-1R mRNA expressions only when combined with compressive loading. TRPV4 agonist suppressed ADAMTS4 and IL-1R mRNA levels induced by the compressive loading, whereas TRPV4 antagonist enhanced these levels. Immunoreactivities to TRPV4 and IL-1R significantly increased in constructs with cyclic compressive loading. Conclusion Cyclic compressive loading induced mRNA expressions of ADAMTS4 and IL-1R through reactive oxygen species. TRPV4 regulated these mRNA expressions, but excessive compressive loading may impair TRPV4 regulation. These findings suggested that TRPV4 regulates the expression level of IL-1R and subsequent IL-1 signaling induced by cyclic compressive loading and participates in cartilage homeostasis. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-021-04095-x.
Collapse
Affiliation(s)
- Yuki Takeda
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yasuo Niki
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Yusuke Fukuhara
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yoshitsugu Fukuda
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuhiko Udagawa
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masayuki Shimoda
- Department of Pathology, School of Medicine, Keio University, Tokyo, Japan
| | - Toshiyuki Kikuchi
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shu Kobayashi
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kengo Harato
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takeshi Miyamoto
- Department of Orthopaedic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
10
|
Gamez C, Schneider-Wald B, Bieback K, Schuette A, Büttner S, Hafner M, Gretz N, Schwarz ML. Compression Bioreactor-Based Mechanical Loading Induces Mobilization of Human Bone Marrow-Derived Mesenchymal Stromal Cells into Collagen Scaffolds In Vitro. Int J Mol Sci 2020; 21:ijms21218249. [PMID: 33158020 PMCID: PMC7672606 DOI: 10.3390/ijms21218249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 11/16/2022] Open
Abstract
Articular cartilage (AC) is an avascular tissue composed of scattered chondrocytes embedded in a dense extracellular matrix, in which nourishment takes place via the synovial fluid at the surface. AC has a limited intrinsic healing capacity, and thus mainly surgical techniques have been used to relieve pain and improve function. Approaches to promote regeneration remain challenging. The microfracture (MF) approach targets the bone marrow (BM) as a source of factors and progenitor cells to heal chondral defects in situ by opening small holes in the subchondral bone. However, the original function of AC is not obtained yet. We hypothesize that mechanical stimulation can mobilize mesenchymal stromal cells (MSCs) from BM reservoirs upon MF of the subchondral bone. Thus, the aim of this study was to compare the counts of mobilized human BM-MSCs (hBM-MSCs) in alginate-laminin (alginate-Ln) or collagen-I (col-I) scaffolds upon intermittent mechanical loading. The mechanical set up within an established bioreactor consisted of 10% strain, 0.3 Hz, breaks of 10 s every 180 cycles for 24 h. Contrary to previous findings using porcine MSCs, no significant cell count was found for hBM-MSCs into alginate-Ln scaffolds upon mechanical stimulation (8 ± 5 viable cells/mm3 for loaded and 4 ± 2 viable cells/mm3 for unloaded alginate-Ln scaffolds). However, intermittent mechanical stimulation induced the mobilization of hBM-MSCs into col-I scaffolds 10-fold compared to the unloaded col-I controls (245 ± 42 viable cells/mm3 vs. 22 ± 6 viable cells/mm3, respectively; p-value < 0.0001). Cells that mobilized into the scaffolds by mechanical loading did not show morphological changes. This study confirmed that hBM-MSCs can be mobilized in vitro from a reservoir toward col-I but not alginate-Ln scaffolds upon intermittent mechanical loading, against gravity.
Collapse
Affiliation(s)
- Carolina Gamez
- Section for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (C.G.); (B.S.-W.); (A.S.)
| | - Barbara Schneider-Wald
- Section for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (C.G.); (B.S.-W.); (A.S.)
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden Württemberg—Hessen, 68167 Mannheim, Germany;
| | - Andy Schuette
- Section for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (C.G.); (B.S.-W.); (A.S.)
| | - Sylvia Büttner
- Department for Statistical Analysis, Faculty of Medicine Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany;
- Institute of Medical Technology, Heidelberg University & Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Norbert Gretz
- Medical Research Centre, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Markus L. Schwarz
- Section for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (C.G.); (B.S.-W.); (A.S.)
- Correspondence: ; Tel.: +49-621-383-4569
| |
Collapse
|
11
|
Gamez C, Schneider-Wald B, Schuette A, Mack M, Hauk L, Khan AUM, Gretz N, Stoffel M, Bieback K, Schwarz ML. Bioreactor for mobilization of mesenchymal stem/stromal cells into scaffolds under mechanical stimulation: Preliminary results. PLoS One 2020; 15:e0227553. [PMID: 31923210 PMCID: PMC6953860 DOI: 10.1371/journal.pone.0227553] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 12/20/2019] [Indexed: 11/18/2022] Open
Abstract
Introduction Articular cartilage (AC) is a viscoelastic tissue with a limited regenerative capability because of the lack of vasculature. Mechanical stimulation contributes to the homeostasis of functional AC since it promotes the delivery of nutrients, cytokines and growth factors between the distant chondrocytes. We hypothesized that biomechanical stimulation might enhance mobilization of endogenous mesenchymal stem/stromal cells (MSCs) from neighboring niches as the bone marrow. Aim This study aimed to introduce a bioreactor for inducing mobilization of MSCs from one compartment to another above by mechanical stimulation in vitro. Methods A novel mechanical system for evaluating mobilization of cells in a 3D context in vitro is presented. The system consists of a compression bioreactor able to induce loading on hydrogel-based scaffolds, custom-made software for settings management and data recording, and image based biological evaluation. Intermittent load was applied under a periodic regime with frequency of 0.3 Hz and unload phases of 10 seconds each 180 cycles over 24 hours. The mechanical stimulation acted on an alginate scaffold and a cell reservoir containing MSCs below it. The dynamic compression exerted amplitude of 200 μm as 10% strain regarding the original height of the scaffold. Results The bioreactor was able to stimulate the scaffolds and the cells for 24.4 (±1.7) hours, exerting compression with vertical displacements of 185.8 (±17.8) μm and a force-amplitude of 1.87 (±1.37; min 0.31, max 4.42) N. Our results suggest that continuous mechanical stimulation hampered the viability of the cells located at the cell reservoir when comparing to intermittent mechanical stimulation (34.4 ± 2.0% vs. 66.8 ± 5.9%, respectively). Functionalizing alginate scaffolds with laminin-521 (LN521) seemed to enhance the mobilization of cells from 48 (±21) to 194 (±39) cells/mm3 after applying intermittent mechanical loading. Conclusion The bioreactor presented here was able to provide mechanical stimulation that seemed to induce the mobilization of MSCs into LN521-alginate scaffolds under an intermittent loading regime.
Collapse
Affiliation(s)
- Carolina Gamez
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Barbara Schneider-Wald
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Andy Schuette
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Michael Mack
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Luisa Hauk
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Arif ul Maula Khan
- Medical Research Centre (ZMF), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Norbert Gretz
- Medical Research Centre (ZMF), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Marcus Stoffel
- Institute of General Mechanics, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, FlowCore Mannheim, German Red Cross Blood Service of Baden Württemberg-Hessen, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Markus L. Schwarz
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
- * E-mail:
| |
Collapse
|
12
|
Vaca-González JJ, Guevara JM, Moncayo MA, Castro-Abril H, Hata Y, Garzón-Alvarado DA. Biophysical Stimuli: A Review of Electrical and Mechanical Stimulation in Hyaline Cartilage. Cartilage 2019; 10:157-172. [PMID: 28933195 PMCID: PMC6425540 DOI: 10.1177/1947603517730637] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Hyaline cartilage degenerative pathologies induce morphologic and biomechanical changes resulting in cartilage tissue damage. In pursuit of therapeutic options, electrical and mechanical stimulation have been proposed for improving tissue engineering approaches for cartilage repair. The purpose of this review was to highlight the effect of electrical stimulation and mechanical stimuli in chondrocyte behavior. DESIGN Different information sources and the MEDLINE database were systematically revised to summarize the different contributions for the past 40 years. RESULTS It has been shown that electric stimulation may increase cell proliferation and stimulate the synthesis of molecules associated with the extracellular matrix of the articular cartilage, such as collagen type II, aggrecan and glycosaminoglycans, while mechanical loads trigger anabolic and catabolic responses in chondrocytes. CONCLUSION The biophysical stimuli can increase cell proliferation and stimulate molecules associated with hyaline cartilage extracellular matrix maintenance.
Collapse
Affiliation(s)
- Juan J. Vaca-González
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| | - Johana M. Guevara
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogota, Colombia
| | - Miguel A. Moncayo
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| | - Hector Castro-Abril
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| | - Yoshie Hata
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
| | - Diego A. Garzón-Alvarado
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| |
Collapse
|
13
|
Maximum shear strain-based algorithm can predict proteoglycan loss in damaged articular cartilage. Biomech Model Mechanobiol 2019; 18:753-778. [PMID: 30631999 DOI: 10.1007/s10237-018-01113-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/24/2018] [Indexed: 01/25/2023]
Abstract
Post-traumatic osteoarthritis (PTOA) is a common disease, where the mechanical integrity of articular cartilage is compromised. PTOA can be a result of chondral defects formed due to injurious loading. One of the first changes around defects is proteoglycan depletion. Since there are no methods to restore injured cartilage fully back to its healthy state, preventing the onset and progression of the disease is advisable. However, this is problematic if the disease progression cannot be predicted. Thus, we developed an algorithm to predict proteoglycan loss of injured cartilage by decreasing the fixed charge density (FCD) concentration. We tested several mechanisms based on the local strains or stresses in the tissue for the FCD loss. By choosing the degeneration threshold suggested for inducing chondrocyte apoptosis and cartilage matrix damage, the algorithm driven by the maximum shear strain showed the most substantial FCD losses around the lesion. This is consistent with experimental findings in the literature. We also observed that by using coordinate system-independent strain measures and selecting the degeneration threshold in an ad hoc manner, all the resulting FCD distributions would appear qualitatively similar, i.e., the greatest FCD losses are found at the tissue adjacent to the lesion. The proposed strain-based FCD degeneration algorithm shows a great potential for predicting the progression of PTOA via biomechanical stimuli. This could allow identification of high-risk defects with an increased risk of PTOA progression.
Collapse
|
14
|
Nickien M, Heuijerjans A, Ito K, van Donkelaar CC. Comparison between in vitro and in vivo cartilage overloading studies based on a systematic literature review. J Orthop Res 2018; 36:2076-2086. [PMID: 29644716 PMCID: PMC6120482 DOI: 10.1002/jor.23910] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 03/27/2018] [Indexed: 02/04/2023]
Abstract
Methodological differences between in vitro and in vivo studies on cartilage overloading complicate the comparison of outcomes. The rationale of the current review was to (i) identify consistencies and inconsistencies between in vitro and in vivo studies on mechanically-induced structural damage in articular cartilage, such that variables worth interesting to further explore using either one of these approaches can be identified; and (ii) suggest how the methodologies of both approaches may be adjusted to facilitate easier comparison and therewith stimulate translation of results between in vivo and in vitro studies. This study is anticipated to enhance our understanding of the development of osteoarthritis, and to reduce the number of in vivo studies. Generally, results of in vitro and in vivo studies are not contradicting. Both show subchondral bone damage and intact cartilage above a threshold value of impact energy. At lower loading rates, excessive loads may cause cartilage fissuring, decreased cell viability, collagen network de-structuring, decreased GAG content, an overall damage increase over time, and low ability to recover. This encourages further improvement of in vitro systems, to replace, reduce, and/or refine in vivo studies. However, differences in experimental set up and analyses complicate comparison of results. Ways to bridge the gap include (i) bringing in vitro set-ups closer to in vivo, for example, by aligning loading protocols and overlapping experimental timeframes; (ii) synchronizing analytical methods; and (iii) using computational models to translate conclusions from in vitro results to the in vivo environment and vice versa. © 2018 The Authors. Journal of Orthopaedic Research® Published by Wiley Periodicals, Inc. J Orthop Res 9999:1-11, 2018.
Collapse
Affiliation(s)
- Mieke Nickien
- Department of Biomedical Engineering, Orthopaedic BiomechanicsEindhoven University of TechnologyP.O. Box 513, 5600MBEindhovenThe Netherlands
| | - Ashley Heuijerjans
- Department of Biomedical Engineering, Orthopaedic BiomechanicsEindhoven University of TechnologyP.O. Box 513, 5600MBEindhovenThe Netherlands
| | - Keita Ito
- Department of Biomedical Engineering, Orthopaedic BiomechanicsEindhoven University of TechnologyP.O. Box 513, 5600MBEindhovenThe Netherlands
| | - Corrinus C. van Donkelaar
- Department of Biomedical Engineering, Orthopaedic BiomechanicsEindhoven University of TechnologyP.O. Box 513, 5600MBEindhovenThe Netherlands
| |
Collapse
|
15
|
Trevino RL, Pacione CA, Malfait AM, Chubinskaya S, Wimmer MA. Development of a Cartilage Shear-Damage Model to Investigate the Impact of Surface Injury on Chondrocytes and Extracellular Matrix Wear. Cartilage 2017; 8:444-455. [PMID: 28934882 PMCID: PMC5613899 DOI: 10.1177/1947603516681133] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Many i n vitro damage models investigate progression of cartilage degradation after a supraphysiologic, compressive impact at the surface and do not model shear-induced damage processes. Models also neglect the response to uninterrupted tribological stress after damage. It was hypothesized that shear-induced removal of the superficial zone would accelerate matrix degradation when damage was followed by continued load and articulation. Methods Bovine cartilage underwent a 5-day test. Shear-damaged samples experienced 2 days of damage induction with articulation against polyethylene and then continued articulation against cartilage (CoC), articulation against metal (MoC), or rest as free-swelling control (FSC). Surface-intact samples were randomized to CoC, MoC, or FSC for the entire 5-day test. Samples were evaluated for chondrocyte viability, GAG (glycosaminoglycan) release (matrix wear surrogate), and histological integrity. Results Shear induction wore away the superficial zone. Damaged samples began continued articulation with collagen matrix disruption and increased cell death compared to intact samples. In spite of the damaged surface, these samples did not exhibit higher GAG release than intact samples articulating against the same counterface ( P = 0.782), contrary to our hypothesis. Differences in GAG release were found to be due to tribological testing against metal ( P = 0.003). Conclusion Shear-induced damage lowers chondrocyte viability and affects extracellular matrix integrity. Continued motion of either cartilage or metal against damaged surfaces did not increase wear compared with intact samples. We conjecture that favorable reorganization of the surface collagen fibers during articulation protected the underlying matrix. This finding suggests a potential window for clinical interventions to slow matrix degradation after traumatic incidents.
Collapse
Affiliation(s)
- Robert L. Trevino
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Carol A. Pacione
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, Chicago, IL, USA
| | - Susan Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA
| | - Markus A. Wimmer
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
- Markus A. Wimmer, Department of Orthopedic Surgery, Rush University Medical Center, 1611 West Harrison Street, Chicago, IL 60612, USA.
| |
Collapse
|
16
|
Huwe LW, Sullan GK, Hu JC, Athanasiou KA. Using Costal Chondrocytes to Engineer Articular Cartilage with Applications of Passive Axial Compression and Bioactive Stimuli. Tissue Eng Part A 2017; 24:516-526. [PMID: 28683690 DOI: 10.1089/ten.tea.2017.0136] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Generating neocartilage with suitable mechanical integrity from a cell source that can circumvent chondrocyte scarcity is indispensable for articular cartilage regeneration strategies. Costal chondrocytes of the rib eliminate donor site morbidity in the articular joint, but it remains unclear how neocartilage formed from these cells responds to mechanical loading, especially if the intent is to use it in a load-bearing joint. In a series of three experiments, this study sought to determine efficacious parameters of passive axial compressive stimulation that would enable costal chondrocytes to synthesize mechanically robust cartilage. Experiment 1 determined a suitable time window for stimulation by its application during either the matrix synthesis phase, the maturation phase, or during both phases of the self-assembling process. The results showed that compressive stimulation at either time was effective in increasing instantaneous moduli by 92% and 87% in the synthesis and maturation phases, respectively. Compressive stimulation during both phases did not further improve properties beyond a one-time stimulation. The magnitude of passive axial compression was examined in Experiment 2 by applying 0, 3.3, 5.0, or 6.7 kPa stresses to the neocartilage. Unlike 6.7 kPa, both 3.3 and 5.0 kPa significantly increased neocartilage compressive properties by 42% and 48% over untreated controls, respectively. Experiment 3 examined how the passive axial compression regimen developed from the previous phases interacted with a bioactive regimen (transforming growth factor [TGF]-β1, chondroitinase ABC, and lysyl oxidase-like 2). Passive axial compression significantly improved the relaxation modulus compared with bioactive treatment alone. Furthermore, a combined treatment of compressive and bioactive stimulation improved the tensile properties of neocartilage 2.6-fold compared with untreated control. The ability to create robust articular cartilage from passaged costal chondrocytes through appropriate mechanical and bioactive stimuli will greatly extend the clinical applicability of tissue-engineered products to a wider patient population.
Collapse
Affiliation(s)
- Le W Huwe
- 1 Department of Biomedical Engineering, University of California , Davis, One Shields Avenue, Davis, California
| | - Gurdeep K Sullan
- 1 Department of Biomedical Engineering, University of California , Davis, One Shields Avenue, Davis, California
| | - Jerry C Hu
- 1 Department of Biomedical Engineering, University of California , Davis, One Shields Avenue, Davis, California
| | - Kyriacos A Athanasiou
- 1 Department of Biomedical Engineering, University of California , Davis, One Shields Avenue, Davis, California.,2 Department of Orthopaedic Surgery, University of California , Davis, One Shields Avenue, Davis, California
| |
Collapse
|
17
|
Trevino RL, Stoia J, Laurent MP, Pacione CA, Chubinskaya S, Wimmer MA. ESTABLISHING A LIVE CARTILAGE-ON-CARTILAGE INTERFACE FOR TRIBOLOGICAL TESTING. ACTA ACUST UNITED AC 2016; 9:1-11. [PMID: 29242820 DOI: 10.1016/j.biotri.2016.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Mechano-biochemical wear encompasses the tribological interplay between biological and mechanical mechanisms responsible for cartilage wear and degradation. The aim of this study was to develop and start validating a novel tribological testing system, which better resembles the natural joint environment through incorporating a live cartilage-on-cartilage articulating interface, joint specific kinematics, and the application of controlled mechanical stimuli for the measurement of biological responses in order to study the mechano-biochemical wear of cartilage. The study entailed two parts. In Part 1, the novel testing rig was used to compare two bearing systems: (a) cartilage articulating against cartilage (CoC) and (b) metal articulating against cartilage (MoC). The clinically relevant MoC, which is also a common tribological interface for evaluating cartilage wear, should produce more wear to agree with clinical observations. In Part II, the novel testing system was used to determine how wear is affected by tissue viability in live and dead CoC articulations. For both parts, bovine cartilage explants were harvested and tribologically tested for three consecutive days. Wear was defined as release of glycosaminoglycans into the media and as evaluation of the tissue structure. For Part I, we found that the live CoC articulation did not cause damage to the cartilage, to the extent of being comparable to the free swelling controls, whereas the MoC articulation caused decreased cell viability, extracellular matrix disruption, and increased wear when compared to CoC, and consistent with clinical data. These results provided confidence that this novel testing system will be adequate to screen new biomaterials for articulation against cartilage, such as in hemiarthroplasty. For Part II, the live and dead cartilage articulation yielded similar wear as determined by the release of proteoglycans and aggrecan fragments, suggesting that keeping the cartilage alive may not be essential for short term wear tests. However, the biosynthesis of glycosaminoglycans was significantly higher due to live CoC articulation than due to the corresponding live free swelling controls, indicating that articulation stimulated cell activity. Moving forward, the cell response to mechanical stimuli and the underlying mechano-biochemical wear mechanisms need to be further studied for a complete picture of tissue degradation.
Collapse
Affiliation(s)
- Robert L Trevino
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL
| | - Jonathan Stoia
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL
| | - Michel P Laurent
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL
| | - Carol A Pacione
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL
| | - Susan Chubinskaya
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL.,Department of Pediatrics, Rush University Medical Center, Chicago, IL
| | - Markus A Wimmer
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL.,Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL
| |
Collapse
|
18
|
Bar Oz M, Kumar A, Elayyan J, Reich E, Binyamin M, Kandel L, Liebergall M, Steinmeyer J, Lefebvre V, Dvir‐Ginzberg M. Acetylation reduces SOX9 nuclear entry and ACAN gene transactivation in human chondrocytes. Aging Cell 2016; 15:499-508. [PMID: 26910618 PMCID: PMC4854920 DOI: 10.1111/acel.12456] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2016] [Indexed: 12/13/2022] Open
Abstract
Changes in the content of aggrecan, an essential proteoglycan of articular cartilage, have been implicated in the pathophysiology of osteoarthritis (OA), a prevalent age-related, degenerative joint disease. Here, we examined the effect of SOX9 acetylation on ACAN transactivation in the context of osteoarthritis. Primary chondrocytes freshly isolated from degenerated OA cartilage displayed lower levels of ACAN mRNA and higher levels of acetylated SOX9 compared with cells from intact regions of OA cartilage. Degenerated OA cartilage presented chondrocyte clusters bearing diffused immunostaining for SOX9 compared with intact cartilage regions. Primary human chondrocytes freshly isolated from OA knee joints were cultured in monolayer or in three-dimensional alginate microbeads (3D). SOX9 was hypo-acetylated in 3D cultures and displayed enhanced binding to a -10 kb ACAN enhancer, a result consistent with higher ACAN mRNA levels than in monolayer cultures. It also co-immunoprecipitated with SIRT1, a major deacetylase responsible for SOX9 deacetylation. Finally, immunofluorescence assays revealed increased nuclear localization of SOX9 in primary chondrocytes treated with the NAD SIRT1 cofactor, than in cells treated with a SIRT1 inhibitor. Inhibition of importin β by importazole maintained SOX9 in the cytoplasm, even in the presence of NAD. Based on these data, we conclude that deacetylation promotes SOX9 nuclear translocation and hence its ability to activate ACAN.
Collapse
Affiliation(s)
- Michal Bar Oz
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Ashok Kumar
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Jinan Elayyan
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Eli Reich
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Milana Binyamin
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Leonid Kandel
- Joint Replacement and Reconstructive Surgery UnitOrthopaedic Surgery ComplexHadassah Mount Scopus HospitalJerusalemIsrael
| | - Meir Liebergall
- Joint Replacement and Reconstructive Surgery UnitOrthopaedic Surgery ComplexHadassah Mount Scopus HospitalJerusalemIsrael
| | - Juergen Steinmeyer
- Laboratory for Experimental OrthopaedicsDepartment of Orthopaedic SurgeryUniversity Hospital Giessen & Marburg GmbHGießenGermany
| | | | - Mona Dvir‐Ginzberg
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| |
Collapse
|
19
|
Hyaluronic Acid Suppresses the Expression of Metalloproteinases in Osteoarthritic Cartilage Stimulated Simultaneously by Interleukin 1β and Mechanical Load. PLoS One 2016; 11:e0150020. [PMID: 26934732 PMCID: PMC4774918 DOI: 10.1371/journal.pone.0150020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/08/2016] [Indexed: 12/05/2022] Open
Abstract
Purpose In patients with osteoarthritis (OA), intraarticular injection of hyaluronic acid (HA) frequently results in reduced pain and improved function for prolonged periods of time, i.e. more than 6 months. However, the mechanisms underlying these effects are not fully understood. Our underlying hypothesis is that HA modifies the enzymatic breakdown of joint tissues. Methods To test this hypothesis, we examined osteochondral cylinders from 12 OA patients. In a bioreactor, these samples were stimulated by interleukin 1β (Il1ß) (2 ng/ml) plus mechanical load (2.0 Mpa at 0.5 Hz horizontal and 0.1 Hz vertical rotation), thus the experimental setup recapitulated both catabolic and anabolic clues of the OA joint. Results Upon addition of HA at either 1 or 3 mg/ml, we observed a significant suppression of expression of metalloproteinase (MMP)-13. A more detailed analysis based on the Kellgren and Lawrence (K&L) OA grade, showed a much greater degree of suppression of MMP-13 expression in grade IV as compared to grade II OA. In contrast to the observed MMP-13 suppression, treatment with HA resulted in a suppression of MMP-1 expression only at 1 mg/ml HA, while MMP-2 expression was not significantly affected by either HA concentration. Conclusion Together, these data suggest that under concurrent catabolic and anabolic stimulation, HA exhibits a pronounced suppressive effect on MMP-13. In the long-run these findings may benefit the development of treatment strategies aimed at blocking tissue degradation in OA patients.
Collapse
|
20
|
Kaviani R, Londono I, Parent S, Moldovan F, Villemure I. Compressive mechanical modulation alters the viability of growth plate chondrocytes in vitro. J Orthop Res 2015; 33:1587-93. [PMID: 26019113 DOI: 10.1002/jor.22951] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/25/2015] [Indexed: 02/04/2023]
Abstract
The aim of this study was to investigate the effect of compressive modulation parameters (mode, magnitude, duration, as well as frequency and amplitude for cyclic modulation) on the viability of growth plate chondrocytes. Swine ulnar growth plate explants (n = 60) were randomly distributed among 10 groups: baseline (n = 1 × 6); culture control (n = 1 × 6); static (n = 3 × 6); and dynamic (n = 5 × 6). Static and dynamic samples were modulated in vitro using a bioreactor. Different compression magnitudes (0.1 MPa or 0.2 MPa), durations (12 h or 24 h), frequencies (0.1 Hz or 1.0 Hz), and amplitudes (30% or 100%) were investigated. Viability was assessed by automatic quantification of number of live/dead cells from confocal images of Live/Dead labeled tissues. Chondrocyte viability was found to be dependent on compression magnitude, duration, frequency, and amplitude in a way that increasing each parameter decreased viability in certain zones of growth plate. More specifically, proliferative and hypertrophic chondrocytes were found to be more sensitive to the applied compression. This study provides an in vitro protocol for studying the effects of compressive modulation on biomechanical and biological responses of growth plate explants, which will be useful in finding efficient and non-detrimental parameters for mechanical modulation of bone growth exploited in scoliosis fusionless treatments.
Collapse
Affiliation(s)
- Rosa Kaviani
- Mechanical Engineering Department, Biomedical Engineering Institute, Ecole Polytechnique of Montreal, Montreal, Quebec, Canada.,Sainte-Justine University Hospital Center, Montreal, Quebec, Canada
| | - Irene Londono
- Sainte-Justine University Hospital Center, Montreal, Quebec, Canada
| | - Stefan Parent
- Sainte-Justine University Hospital Center, Montreal, Quebec, Canada.,Department of Surgery, University of Montreal, Montreal, Quebec, Canada
| | - Florina Moldovan
- Sainte-Justine University Hospital Center, Montreal, Quebec, Canada.,Department of Stomatology, University of Montreal, Montreal, Canada
| | - Isabelle Villemure
- Mechanical Engineering Department, Biomedical Engineering Institute, Ecole Polytechnique of Montreal, Montreal, Quebec, Canada.,Sainte-Justine University Hospital Center, Montreal, Quebec, Canada
| |
Collapse
|
21
|
Bhattacharjee M, Coburn J, Centola M, Murab S, Barbero A, Kaplan DL, Martin I, Ghosh S. Tissue engineering strategies to study cartilage development, degeneration and regeneration. Adv Drug Deliv Rev 2015; 84:107-22. [PMID: 25174307 DOI: 10.1016/j.addr.2014.08.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/01/2014] [Accepted: 08/20/2014] [Indexed: 01/09/2023]
Abstract
Cartilage tissue engineering has primarily focused on the generation of grafts to repair cartilage defects due to traumatic injury and disease. However engineered cartilage tissues have also a strong scientific value as advanced 3D culture models. Here we first describe key aspects of embryonic chondrogenesis and possible cell sources/culture systems for in vitro cartilage generation. We then review how a tissue engineering approach has been and could be further exploited to investigate different aspects of cartilage development and degeneration. The generated knowledge is expected to inform new cartilage regeneration strategies, beyond a classical tissue engineering paradigm.
Collapse
|
22
|
Extracellular matrix integrity affects the mechanical behaviour of in-situ chondrocytes under compression. J Biomech 2014; 47:1004-13. [PMID: 24480705 DOI: 10.1016/j.jbiomech.2014.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 12/31/2013] [Accepted: 01/03/2014] [Indexed: 11/22/2022]
Abstract
Cartilage lesions change the microenvironment of cells and may accelerate cartilage degradation through catabolic responses from chondrocytes. In this study, we investigated the effects of structural integrity of the extracellular matrix (ECM) on chondrocytes by comparing the mechanics of cells surrounded by an intact ECM with cells close to a cartilage lesion using experimental and numerical methods. Experimentally, 15% nominal compression was applied to bovine cartilage tissues using a light-transmissible compression system. Target cells in the intact ECM and near lesions were imaged by dual-photon microscopy. Changes in cell morphology (N(cell)=32 for both ECM conditions) were quantified. A two-scale (tissue level and cell level) Finite Element (FE) model was also developed. A 15% nominal compression was applied to a non-linear, biphasic tissue model with the corresponding cell level models studied at different radial locations from the centre of the sample in the transient phase and at steady state. We studied the Green-Lagrange strains in the tissue and cells. Experimental and theoretical results indicated that cells near lesions deform less axially than chondrocytes in the intact ECM at steady state. However, cells near lesions experienced large tensile strains in the principal height direction, which are likely associated with non-uniform tissue radial bulging. Previous experiments showed that tensile strains of high magnitude cause an up-regulation of digestive enzyme gene expressions. Therefore, we propose that cartilage degradation near tissue lesions may be due to the large tensile strains in the principal height direction applied to cells, thus leading to an up-regulation of catabolic factors.
Collapse
|
23
|
Schadow S, Siebert HC, Lochnit G, Kordelle J, Rickert M, Steinmeyer J. Collagen metabolism of human osteoarthritic articular cartilage as modulated by bovine collagen hydrolysates. PLoS One 2013; 8:e53955. [PMID: 23342047 PMCID: PMC3546930 DOI: 10.1371/journal.pone.0053955] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 12/04/2012] [Indexed: 02/07/2023] Open
Abstract
Destruction of articular cartilage is a characteristic feature of osteoarthritis (OA). Collagen hydrolysates are mixtures of collagen peptides and have gained huge public attention as nutriceuticals used for prophylaxis of OA. Here, we evaluated for the first time whether different bovine collagen hydrolysate preparations indeed modulate the metabolism of collagen and proteoglycans from human OA cartilage explants and determined the chemical composition of oligopeptides representing collagen fragments. Using biophysical techniques, like MALDI-TOF-MS, AFM, and NMR, the molecular weight distribution and aggregation behavior of collagen hydrolysates from bovine origin (CH-Alpha®, Peptan™ B 5000, Peptan™ B 2000) were determined. To investigate the metabolism of human femoral OA cartilage, explants were obtained during knee replacement surgery. Collagen synthesis of explants as modulated by 0–10 mg/ml collagen hydrolysates was determined using a novel dual radiolabeling procedure. Proteoglycans, NO, PGE2, MMP-1, -3, -13, TIMP-1, collagen type II, and cell viability were determined in explant cultures. Groups of data were analyzed using ANOVA and the Friedman test (n = 5–12). The significance was set to p≤0.05. We found that collagen hydrolysates obtained from different sources varied with respect to the width of molecular weight distribution, average molecular weight, and aggregation behavior. None of the collagen hydrolysates tested stimulated the biosynthesis of collagen. Peptan™ B 5000 elevated NO and PGE2 levels significantly but had no effect on collagen or proteoglycan loss. All collagen hydrolysates tested proved not to be cytotoxic. Together, our data demonstrate for the first time that various collagen hydrolysates differ with respect to their chemical composition of collagen fragments as well as by their pharmacological efficacy on human chondrocytes. Our study underscores the importance that each collagen hydrolysate preparation should first demonstrate its pharmacological potential both in vitro and in vivo before being used for both regenerative medicine and prophylaxis of OA.
Collapse
Affiliation(s)
- Saskia Schadow
- Department of Orthopedics, University Hospital Giessen and Marburg, Giessen, Germany
| | | | - Günter Lochnit
- Department of Biochemistry, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jens Kordelle
- Agaplesion Evangelical Hospital Mittelhessen, Giessen, Germany
| | - Markus Rickert
- Department of Orthopedics, University Hospital Giessen and Marburg, Giessen, Germany
| | - Jürgen Steinmeyer
- Department of Orthopedics, University Hospital Giessen and Marburg, Giessen, Germany
- * E-mail:
| |
Collapse
|
24
|
Effects of unloading on knee articular cartilage T1rho and T2 magnetic resonance imaging relaxation times: a case series. J Orthop Sports Phys Ther 2012; 42:511-20. [PMID: 22402583 PMCID: PMC3673554 DOI: 10.2519/jospt.2012.3975] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
STUDY DESIGN Case series. BACKGROUND It has been shown in rodent and canine models that cartilage composition is significantly altered in response to long-term unloading. To date, however, no in vivo human studies have investigated this topic. The objective of this case series was to determine the influence of unloading and reloading on T1rho and T2 relaxation times of articular cartilage in healthy young joints. CASE DESCRIPTION Ten patients who required 6 to 8 weeks of non-weight bearing (NWB) for injuries affecting the distal lower extremity participated in the study. Quantitative T1rho and T2 imaging of the ipsilateral knee joint was performed at 3 time points: (1) prior to surgery (baseline), (2) immediately after a period of NWB (post-NWB), and (3) after 4 weeks of full weight bearing (post-FWB). Cartilage regions of interest were segmented and overlaid on T1rho and T2 relaxation time maps for quantification. Descriptive statistics are provided for all changes. OUTCOMES Increases of 5% to 10% in T1rho times of all femoral and tibial compartments were noted post-NWB. All values returned to near-baseline levels post-FWB. Increases in medial tibia T2 times were noted post-NWB and remained elevated post-FWB. The load-bearing regions showed the most significant changes in response to unloading, with increases of up to 12%. DISCUSSION The observation of a transient shift in relaxation times confirms that cartilage composition is subject to alterations based on loading conditions. These changes appear to be mostly related to proteoglycan content and more localized to the load-bearing regions. However, following 4 weeks of full weight bearing, relaxation times of nearly all regions had returned to baseline levels, demonstrating reversibility in compositional fluctuations. LEVEL OF EVIDENCE Therapy, level 4.
Collapse
|
25
|
Biomechanical influence of cartilage homeostasis in health and disease. ARTHRITIS 2011; 2011:979032. [PMID: 22046527 PMCID: PMC3196252 DOI: 10.1155/2011/979032] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/26/2011] [Indexed: 11/30/2022]
Abstract
There is an urgent demand for long term solutions to improve osteoarthritis treatments in the ageing population. There are drugs that control the pain but none that stop the progression of the disease in a safe and efficient way. Increased intervention efforts, augmented by early diagnosis and integrated biophysical therapies are therefore needed. Unfortunately, progress has been hampered due to the wide variety of experimental models which examine the effect of mechanical stimuli and inflammatory mediators on signal transduction pathways. Our understanding of the early mechanopathophysiology is poor, particularly the way in which mechanical stimuli influences cell function and regulates matrix synthesis. This makes it difficult to identify reliable targets and design new therapies. In addition, the effect of mechanical loading on matrix turnover is dependent on the nature of the mechanical stimulus. Accumulating evidence suggests that moderate mechanical loading helps to maintain cartilage integrity with a low turnover of matrix constituents. In contrast, nonphysiological mechanical signals are associated with increased cartilage damage and degenerative changes. This review will discuss the pathways regulated by compressive loading regimes and inflammatory signals in animal and in vitro 3D models. Identification of the chondroprotective pathways will reveal novel targets for osteoarthritis treatments.
Collapse
|
26
|
Moo EK, Osman NAA, Pingguan-Murphy B. The metabolic dynamics of cartilage explants over a long-term culture period. Clinics (Sao Paulo) 2011; 66:1431-6. [PMID: 21915496 PMCID: PMC3161224 DOI: 10.1590/s1807-59322011000800021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 04/05/2011] [Accepted: 04/24/2011] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Although previous studies have been performed on cartilage explant cultures, the generalized dynamics of cartilage metabolism after extraction from the host are still poorly understood due to differences in the experimental setups across studies, which in turn prevent building a complete picture. METHODS In this study, we investigated the response of cartilage to the trauma sustained during extraction and determined the time needed for the cartilage to stabilize. Explants were extracted aseptically from bovine metacarpal-phalangeal joints and cultured for up to 17 days. RESULTS The cell viability, cell number, proteoglycan content, and collagen content of the harvested explants were analyzed at 0, 2, 10, and 17 days after explantation. A high percentage of the cartilage explants were found to be viable. The cell density initially increased significantly but stabilized after two days. The proteoglycan content decreased gradually over time, but it did not decrease to a significant level due to leakage through the distorted peripheral collagen network and into the bathing medium. The collagen content remained stable for most of the culture period until it dropped abruptly on day 17. CONCLUSION Overall, the tested cartilage explants were sustainable over long-term culture. They were most stable from day 2 to day 10. The degradation of the collagen on day 17 did not reach diseased levels, but it indicated the potential of the cultures to develop into degenerated cartilage. These findings have implications for the application of cartilage explants in pathophysiological fields.
Collapse
Affiliation(s)
- E K Moo
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | | | | |
Collapse
|
27
|
Bar-Ziv Y, Beer Y, Ran Y, Benedict S, Halperin N. A treatment applying a biomechanical device to the feet of patients with knee osteoarthritis results in reduced pain and improved function: a prospective controlled study. BMC Musculoskelet Disord 2010; 11:179. [PMID: 20698991 PMCID: PMC2928172 DOI: 10.1186/1471-2474-11-179] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 08/10/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study examined the effect of treatment with a novel biomechanical device on the level of pain and function in patients with knee OA. METHODS Patients with bilateral knee OA were enrolled to active and control groups. Patients were evaluated at baseline, at 4 weeks and at the 8-week endpoint. A novel biomechanical device was individually calibrated to patients from the active group. Patients from the control group received an identical foot-worn platform without the biomechanical elements. Primary outcomes were the WOMAC Index and ALF assessments. RESULTS There were no baseline differences between the groups. At 8 weeks, the active group showed a mean improvement of 64.8% on the WOMAC pain scale, a mean improvement of 62.7% on the WOMAC function scale, and a mean improvement of 31.4% on the ALF scale. The control group demonstrated no improvement in the above parameters. Significant differences were found between the active and control groups in all the parameters of assessment. CONCLUSIONS The biomechanical device and treatment methodology is effective in significantly reducing pain and improving function in knee OA patients.The study is registered at clinicaltrials.gov, identifier NCT00457132, http://www.clinicaltrials.gov/ct/show/NCT00457132?order=1.
Collapse
Affiliation(s)
- Yaron Bar-Ziv
- Department of Orthopedic Surgery, Assaf Harofeh Medical Center, Zerifin, Israel
| | | | | | | | | |
Collapse
|
28
|
Different mechanical loading protocols influence serum cartilage oligomeric matrix protein levels in young healthy humans. Eur J Appl Physiol 2010; 110:651-7. [DOI: 10.1007/s00421-010-1529-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2010] [Indexed: 10/19/2022]
|
29
|
Steinmeyer J, Kordelle J, Stürz H. In vitro inhibition of aggrecanase activity by tetracyclines and proteoglycan loss from osteoarthritic human articular cartilage. J Orthop Res 2010; 28:828-33. [PMID: 20069635 DOI: 10.1002/jor.21026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Tetracyclines were reported to slow down the progression of cartilage damage both in an animal model of osteoarthritis (OA) and in humans. In search for the underlying mechanisms we examined whether tetracyclines possess an inhibitory potential on the activity of aggrecanases and inflammatory mediators and can thus prevent proteoglycan (PG) loss from human articular cartilage. In vitro activity of aggrecanase-1 and -2 was recorded in the presence of 1-100 microM tetracycline, minocycline, or doxycyline. Human knee articular cartilage explants were sorted according to the degree of OA and treated for 10 days with tetracycline derivatives in the presence of interleukin-1 (IL-1beta). Synthesis and loss of PGs, nitric oxide (NO), and prostaglandin E(2) (PGE(2)), as well as the viability were determined. Tetracyclines derivatives dose-dependently inhibited the activities of both aggrecanases in vitro, whereas no inhibitory effect of tetracyclines on any proteoglycanolytic activities within IL-1beta-treated human cartilage explants were found. Tetracyclines can significantly modulate NO and PGE(2) levels, but have no effect on PG synthesis and loss within the same human cartilage explant cultures. Altogether, our data show that tetracyclines have no inhibitory potential on any proteoglycanolytic activities within mild or moderately affected human OA cartilage at therapeutic achievable plasma levels.
Collapse
Affiliation(s)
- Jürgen Steinmeyer
- Department of Orthopaedic Surgery, University Hospital Giessen and Marburg GmbH, Giessen, Germany.
| | | | | |
Collapse
|
30
|
Gunja NJ, Athanasiou KA. Effects of hydrostatic pressure on leporine meniscus cell-seeded PLLA scaffolds. J Biomed Mater Res A 2010; 92:896-905. [PMID: 19283825 DOI: 10.1002/jbm.a.32451] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Hydrostatic pressure (HP) is an important component of the loading environment of the knee joint. Studies with articular chondrocytes and TMJ disc fibrochondrocytes have identified certain benefits of HP for tissue engineering purposes. However, similar studies with meniscus cells are lacking. Thus, in this experiment, the effects of applying 10 MPa of HP at three different frequencies (0, 0.1, and 1 Hz) to leporine meniscus cell-seeded PLLA scaffolds were examined. HP was applied once every 3 days for 1 h for a period of 28 days. Constructs were analyzed for cellular, biochemical, and biomechanical properties. At t = 4 weeks, total collagen/scaffold was found to be significantly higher in the 10 MPa, 0 Hz group when compared with other groups. This despite the fact that the cell numbers/scaffold were found to be lower in all HP groups when compared with the culture control. Additionally, the total GAG/scaffold, instantaneous modulus, and relaxation modulus were significantly increased in the 10 MPa, 0 Hz group when compared with the culture control. In summary, this experiment provides evidence for the benefit of a 10 MPa, 0 Hz stimulus, on both biochemical and biomechanical aspects, for the purposes of meniscus tissue engineering using PLLA scaffolds.
Collapse
Affiliation(s)
- Najmuddin J Gunja
- Department of Bioengineering, Rice University, Houston, Texas 77251, USA
| | | |
Collapse
|
31
|
Torzilli PA, Bhargava M, Park S, Chen CC. Mechanical load inhibits IL-1 induced matrix degradation in articular cartilage. Osteoarthritis Cartilage 2010; 18:97-105. [PMID: 19747586 PMCID: PMC2818235 DOI: 10.1016/j.joca.2009.07.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 07/17/2009] [Accepted: 06/22/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis is a disease process of cellular degradation of articular cartilage caused by mechanical loads and inflammatory cytokines. We studied the cellular response in native cartilage subjected to a mechanical load administered simultaneously with an inflammatory cytokine interleukin-1 (IL-1), hypothesizing that the combination of load and cytokine would result in accelerated extracellular matrix (ECM) degradation. METHODS Mature bovine articular cartilage was loaded for 3 days (stimulation) with 0.2 and 0.5 MPa stresses, with and without IL-1 (IL-1alpha, 10 ng/ml), followed by 3 days of no stimulation (recovery). Aggrecan and collagen loss were measured as well as aggrecan cleavage using monoclonal antibodies AF-28 and BC-3 for cleavage by aggrecanases (ADAMTS) and matrix metalloproteinases (MMPs), respectively. RESULTS Incubation with IL-1 caused aggrecan cleavage by aggrecanases and MMPs during the 3 days of stimulation. A load of 0.5 MPa inhibited the IL-1-induced aggrecan loss while no inhibition was found for the 0.2 MPa stress. There was no collagen loss during the treatments but upon load and IL-1 removal proteoglycan and collagen loss increased. Load itself under these conditions was found to have no effect when compared to the unloaded controls. CONCLUSIONS A mechanical load of sufficient magnitude can inhibit ECM degradation by chondrocytes when stimulated by IL-1. The molecular mechanisms involved in this process are not clear but probably involve altered mechanochemical signal transduction between the ECM and chondrocyte.
Collapse
Affiliation(s)
| | | | - Seonghun Park
- School of Mechanical Engineering Pusan National University Busan, Republic of Korea (South Korea)
| | | |
Collapse
|
32
|
Bougault C, Paumier A, Aubert-Foucher E, Mallein-Gerin F. Investigating conversion of mechanical force into biochemical signaling in three-dimensional chondrocyte cultures. Nat Protoc 2009; 4:928-38. [PMID: 19478808 DOI: 10.1038/nprot.2009.63] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The culture of chondrocytes embedded within agarose hydrogels maintains chondrocytic phenotype over extended periods and allows analysis of the chondrocyte response to mechanical forces. The mechanisms involved in the transduction of a mechanical stimulus to a physiological process are not completely deciphered. We present protocols to prepare and characterize constructs of murine chondrocytes and agarose (1 week pre-culture period), to analyze the effect of compression on mRNA level by RT-PCR (2-3 d), gene transcription by gene reporter assay (3 d) and phosphorylation state of signaling molecules by western blotting (3-4 d). The protocols can be carried out with a limited number of mouse embryos or newborns and this point is particularly important regarding genetically modified mice.
Collapse
Affiliation(s)
- Carole Bougault
- UMR5086, CNRS, IFR128, IBCP (Institut de Biologie et Chimie des Protéines), Université de Lyon, Lyon, France
| | | | | | | |
Collapse
|
33
|
Nam J, Rath B, Knobloch TJ, Lannutti JJ, Agarwal S. Novel electrospun scaffolds for the molecular analysis of chondrocytes under dynamic compression. Tissue Eng Part A 2009; 15:513-23. [PMID: 18694324 DOI: 10.1089/ten.tea.2007.0353] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mechanical training of engineered tissue constructs is believed necessary to improve regeneration of cartilaginous grafts. Nevertheless, molecular mechanisms underlying mechanical activation are not clear. This is partly due to unavailability of appropriate scaffolds allowing exposure of cells to dynamic compressive strains (DCS) in vitro while permitting subsequent molecular analyses. We demonstrate that three-dimensional macroporous electrospun poly(epsilon-caprolactone) scaffolds can be fabricated that are suitable for the functional and molecular analysis of dynamically loaded chondrocytes. These scaffolds encourage chondrocytic proliferation promoting expression of collagen type II, aggrecan, and Sox9 while retaining mechanical strength after prolonged dynamic compression. Further, they exhibit superior infiltration of exogenous agents into the cells and permit easy retrieval of cellular components postcompression to allow exploration of molecular mechanisms of DCS. Using these scaffolds, we observed that chondrocytes responded to DCS in a magnitude-dependent manner exhibiting antiinflammatory and proanabolic responses at low physiological magnitudes. Proinflammatory responses and decreased cellular viability were observed at hyperphysiological magnitudes. These scaffolds provide a means of unraveling the mechanotransduction-induced transcriptional and posttranslational activities involved in cartilage regeneration and repair.
Collapse
Affiliation(s)
- Jin Nam
- Biomechanics and Tissue Engineering Laboratory, College of Dentistry, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
34
|
Kim E, Guilak F, Haider MA. The dynamic mechanical environment of the chondrocyte: a biphasic finite element model of cell-matrix interactions under cyclic compressive loading. J Biomech Eng 2009; 130:061009. [PMID: 19045538 DOI: 10.1115/1.2978991] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cyclic mechanical loading of articular cartilage results in a complex biomechanical environment at the scale of the chondrocytes that strongly affects cellular metabolic activity. Under dynamic loading conditions, the quantitative relationships between macroscopic loading characteristics and solid and fluid mechanical variables in the local cellular environment are not well understood. In this study, an axisymmetric multiscale model of linear biphasic cell-matrix interactions in articular cartilage was developed to investigate the cellular microenvironment in an explant subjected to cyclic confined compressive loading. The model was based on the displacement-velocity-pressure (u-v-p) mixed-penalty weighted residual formulation of linear biphasic theory that was implemented in the COMSOL MULTIPHYSICS software package. The microscale cartilage environment was represented as a three-zone biphasic region consisting of a spherical chondrocyte with encapsulating pericellular matrix (PCM) that was embedded in a cylindrical extracellular matrix (ECM) subjected to cyclic confined compressive loading boundary conditions. Biphasic material properties for the chondrocyte and the PCM were chosen based on previous in vitro micropipette aspiration studies of cells or chondrons isolated from normal or osteoarthritic cartilage. Simulations performed at four loading frequencies in the range 0.01-1.0 Hz supported the hypothesized dual role of the PCM as both a protective layer for the cell and a mechanical transducer of strain. Time varying biphasic variables at the cellular scale were strongly dependent on relative magnitudes of the loading period, and the characteristic gel diffusion times for the ECM, the PCM, and the chondrocyte. The multiscale simulations also indicated that axial strain was significantly amplified in the range 0.01-1.0 Hz, with a decrease in amplification factor and frequency insensitivity at the higher frequencies. Simulations of matrix degradation due to osteoarthritis indicated that strain amplification factors were more significantly altered when loss of matrix stiffness was exclusive to the PCM. The findings of this study demonstrate the complex dependence of dynamic mechanics in the local cellular environment of cartilage on macroscopic loading features and material properties of the ECM and the chondron.
Collapse
Affiliation(s)
- Eunjung Kim
- Department of Mathematics, North Carolina State University, Raleigh, NC 27695, USA
| | | | | |
Collapse
|
35
|
Bougault C, Paumier A, Aubert-Foucher E, Mallein-Gerin F. Molecular analysis of chondrocytes cultured in agarose in response to dynamic compression. BMC Biotechnol 2008; 8:71. [PMID: 18793425 PMCID: PMC2556324 DOI: 10.1186/1472-6750-8-71] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Accepted: 09/15/2008] [Indexed: 12/02/2022] Open
Abstract
Background Articular cartilage is exposed to high mechanical loads under normal physiological conditions and articular chondrocytes regulate the composition of cartilaginous matrix, in response to mechanical signals. However, the intracellular pathways involved in mechanotransduction are still being defined. Using the well-characterized chondrocyte/agarose model system and dynamic compression, we report protocols for preparing and characterizing constructs of murine chondrocytes and agarose, and analyzing the effect of compression on steady-state level of mRNA by RT-PCR, gene transcription by gene reporter assay, and phosphorylation state of signalling molecules by Western-blotting. The mouse model is of particular interest because of the availability of a large choice of bio-molecular tools suitable to study it, as well as genetically modified mice. Results Chondrocytes cultured in agarose for one week were surrounded by a newly synthesized pericellular matrix, as revealed by immunohistochemistry prior to compression experiments. This observation indicates that this model system is suitable to study the role of matrix molecules and trans-membrane receptors in cellular responsiveness to mechanical stress. The chondrocyte/agarose constructs were then submitted to dynamic compression with FX-4000C™ Flexercell® Compression Plus™ System (Flexcell). After clearing proteins off agarose, Western-blotting analysis showed transient activation of Mitogen-activated protein kinases (MAPK) in response to dynamic compression. After assessment by capillary electrophoresis of the quality of RNA extracted from agarose, steady-state levels of mRNA expression was measured by real time PCR. We observed an up-regulation of cFos and cJun mRNA levels as a response to compression, in accordance with the mechanosensitive character observed for these two genes in other studies using cartilage explants submitted to compression. To explore further the biological response of mouse chondrocytes to the dynamic compression at the transcriptional level, we also developed an approach for monitoring changes in gene transcription in agarose culture by using reporter promoter constructs. A decrease in promoter activity of the gene coding for type II procollagen, the most abundant protein in cartilage, was observed in response to dynamic loading. Conclusion The protocols developed here offer the possibility to perform an integrated analysis of the molecular mechanisms of mechanotransduction in chondrocytes, at the gene and protein level.
Collapse
Affiliation(s)
- Carole Bougault
- UMR 5086, CNRS, Université de Lyon, IFR 128, IBCP, Institut de Biologie et Chimie des Protéines, 7 passage du Vercors F-69367 Lyon FRANCE.
| | | | | | | |
Collapse
|
36
|
Stephens EH, Chu CK, Grande-Allen KJ. Valve proteoglycan content and glycosaminoglycan fine structure are unique to microstructure, mechanical load and age: Relevance to an age-specific tissue-engineered heart valve. Acta Biomater 2008; 4:1148-60. [PMID: 18448399 PMCID: PMC10615646 DOI: 10.1016/j.actbio.2008.03.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Revised: 02/01/2008] [Accepted: 03/20/2008] [Indexed: 02/02/2023]
Abstract
This study characterized valve proteoglycan and glycosaminoglycan composition during development and aging. This knowledge is important for the development of age-specific tissue-engineered heart valves as well as treatments for age-specific valvulopathies. Aortic valves and mitral valves from first-third trimester, 6-week, 6-month and 6-year-old pigs were examined using immunohistochemistry for versican, biglycan, decorin and hyaluronan, as well as elastin and fibrillin. The fine structure of glycosaminoglycans was examined by fluorophore-assisted carbohydrate electrophoresis. Decorin expression was strongest in the 6-year-old valves, particularly in the aortic valve spongiosa. The quantity of iduronate was also highest in the 6-year-old valves. The central tensile-loading region of the anterior mitral leaflet demonstrated reduced glycosaminoglycan content, chain length and hydration and a larger fraction of 4-sulfated iduronate and lower fraction of 6-sulfation. With age, the anterior leaflet center showed a further increase in 4-sulfated iduronate and decrease in 6-sulfation. In contrast, the anterior leaflet free edge showed decreased iduronate and 4-sulfated glucuronate content with age. The young aortic valve was similar to the mitral valve free edge with a higher concentration of glycosaminoglycans and 6-rather than 4-sulfation, but aged to resemble the mitral anterior leaflet center, with an increase in 4-sulfated iduronate content and a decrease in the 6-sulfation fraction. Elastin and fibrillin often co-localized with the proteoglycans studied, but elastin co-localized most specifically with versican. In conclusion, composition and fine structure changes in valve proteoglycans and glycosaminoglycans with age are complex and distinct within valve type, histological layers and regions of different mechanical loading.
Collapse
Affiliation(s)
- Elizabeth H Stephens
- Department of Bioengineering, Rice University, P.O. Box 1892 - MS142, Houston, TX 77251-1892, USA
| | | | | |
Collapse
|
37
|
Niehoff A, Offermann M, Dargel J, Schmidt A, Brüggemann GP, Bloch W. Dynamic and static mechanical compression affects Akt phosphorylation in porcine patellofemoral joint cartilage. J Orthop Res 2008; 26:616-23. [PMID: 18050339 DOI: 10.1002/jor.20542] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Protein kinase B (Akt)-dependent signaling pathways induced by mechanical loading have been identified in a variety of tissue. However, there is no evidence for a potential regulation of Akt in cartilage mechanotransduction. This study was conducted in order to determine whether or not the Akt in chondrocytes is regulated by mechanical loading. Porcine patellofemoral joints were loaded in compression at 500 N for 150 s either dynamically at 12 Hz or 1 Hz or statically using a custom-designed loading frame. Left-sided knees served as intervention, right-sided as unloaded control. Cartilage samples were harvested at different time points after mechanical loading and the phosphorylation of Akt was analyzed immunohistochemically. A downregulation of Akt phosphorylation was seen in cartilage 300 s after mechanical loading whereas Akt phosphorylation remained unchanged in unloaded specimens. In addition, regulation of Akt appeared to change with the frequency of loading, presenting different patterns in Akt phosphorylation with static and dynamic loading. Variations in Akt phosphorylation were detected through different zones of cartilage. Overall, our findings indicate that Akt signaling in porcine patellofemoral joint cartilage is dependent upon frequency of loading, cartilage zone, and the time interval between loading and cartilage harvest. It may be concluded that Akt plays a role in cartilage mechanotransduction.
Collapse
Affiliation(s)
- Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Carl-Diem-Weg 6, 50933 Cologne, Germany.
| | | | | | | | | | | |
Collapse
|
38
|
Pharmacological enhancement of disc diffusion and differentiation of healthy, ageing and degenerated discs : Results from in-vivo serial post-contrast MRI studies in 365 human lumbar discs. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2008; 17:626-43. [PMID: 18357472 DOI: 10.1007/s00586-008-0645-6] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Accepted: 02/16/2008] [Indexed: 01/07/2023]
Abstract
Degenerative disc disease (DDD) is still a poorly understood phenomenon because of the lack of availability of precise definition of healthy, ageing and degenerated discs. Decreased nutrition is the final common pathway for DDD and the status of the endplate (EP) plays a crucial role in controlling the extent of diffusion, which is the only source of nutrition. The vascular channels in the subchondral plate have muscarinic receptors but the possibility of enhancing diffusion pharmacologically by dilation of these vessels has not been probed. Although it is well accepted that EP damage will affect diffusion and thereby nutrition, there is no described method to quantify the extent of EP damage. Precise definitions with an objective method of differentiating healthy, ageing and degenerated discs on the basis of anatomical integrity of the disc and physiological basis of altered nutrition will be useful. This information is an urgent necessity for better understanding of DDD and also strategizing prevention and treatment. Seven hundred and thirty endplates of 365 lumbar discs from 73 individuals (26 healthy volunteers and 47 patients) with age ranging from 10-64 years were evaluated by pre-contrast and 10 min, 2, 4, 6 and 12 h post contrast MRI after IV injection of 0.3 mmol/kg of Gadodiamide. End plates were classified according to the extent of damage into six grades and an incremental score was given for each category. A total endplate score (TEPS) was derived by adding the EP score of the two endplates for each concerned disc. The base line value (SI(base)) and the signal intensity at particular time periods were used to derive the enhancement percentage for each time period (Enhancement (%) = SI(tp) - SI(base)/SI(base) x 100). The enhancement percentage for each time period, the time for peak enhancement (T-max) and the time intensity curve (TIC) over 12 h were used to study and compare the diffusion characteristics. The differences in pattern of diffusion were obvious visually at 4 h which was categorized into five patterns-Pattern A representing normal diffusion to Pattern E representing a total abnormality in diffusion. Degeneration was classified according to Pfirrmann's grading and this was correlated to the TEPS and the alterations in diffusion patterns. The relationship of TEPS on the increase in DDD was evaluated by a logistic curve and the cut point for severe DDD was found by ROC curve. The influence of the variables of age, level, Modic changes, instability, annulus fibrosis defect (DEBIT), TEPS and diffusion patterns on DDD was analyzed by multiple and stepwise regression analysis. Oral nimodipine study: Additional forty lumbar end-plates from four young healthy volunteers were studied to document the effect of oral nimodipine. Pre-drug diffusion levels were studied by pre and post contrast MRI (0.3 mmol/kg of gadodiamide) at 10 min, 2, 4, 6, 12 and 24 h. Oral nimodipine was administered (30 mg QID) for 5 days and post-contrast MRI studies were performed similarly. Enhancement was calculated at vertebral body-VB; subchondral bone-SCB; Endplate Zone-EPZ and at superior and inferior peripheral nucleus pulposus-PNP and central nucleus pulposus-CNP, using appropriate cursors by a blinded investigator. Paired sample t test and area under curve (AUC) measurements were done.The incidence of disc degeneration had a significant correlation with increasing TEPS (Trend Chi-square, P < 0.01). Only one out of 83 (1.2%) disc had either Pfirrmann Grade IV or V when the score was 4 or below when compared to 34/190 (17.9%) for scores 5-7; 41 of 72 (56.9%) for scores 8-10 and 18 of 20 (90%) for scores 11 and 12 (P < 0.001 for all groups). Pearson's correlation between TEPS and DDD was statistically significant, irrespective of the level of disc or different age groups (r value was above 0.6 and P < 0.01 for all age groups). Logistic curve fit analysis and ROC curve analysis showed that the incidence of DDD increased abruptly when the TEPS crossed six. With a progressive increase of end plate damage, five different patterns of diffusion were visualized. Pattern D and E represented totally altered diffusion pattern questioning the application of biological method of treatment in such situations. Four types of time intensity curves (TIC) were noted which helped to differentiate between healthy, aged and degenerated discs. Multiple and stepwise regression analysis indicated that pattern of disc diffusion and TEPS to be the most significant factors influencing DDD, irrespective of age. Nimodipine increased the average signal intensity for all regions-by 7.6% for VB, 8% for SCB and EPZ and 11% for CNP at all time intervals (P < 0.01 for all cases). Although the increase was high at all time intervals, the maximum increase was at 2 h for VB, SCB and EPZ; 4 h for PNP and 12 h for CNP. It was also interesting that post-nimodipine, the peak signal intensity was attained early, was higher and maintained longer compared to pre-nimodipine values. Our study has helped to establish that EP damage as a crucial event leading to structural failure thereby precipitating DDD. An EP damage score has been devised which had a good correlation to DDD and discs with a score of six and above can be considered 'at risk' for severe DDD. New data on disc diffusion patterns were obtained which may help to differentiate healthy, ageing and degenerated discs in in-vivo conditions. This is also the first study to document an increase in diffusion of human lumbar discs by oral nimodipine and poses interesting possibility of pharmacological enhancement of lumbar disc nutrition.
Collapse
|
39
|
Static compression of single chondrocytes catabolically modifies single-cell gene expression. Biophys J 2007; 94:2412-22. [PMID: 18065463 DOI: 10.1529/biophysj.107.114207] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Previous work has established that mechanical forces can lead to quantifiable alterations in cell function. However, how forces change gene expression in a single cell and the mechanisms of force transmission to the nucleus are poorly understood. Here we demonstrate that the gene expression of proteins related to the extracellular matrix in single articular chondrocytes is modified by compressive forces in a dosage-dependent manner. Increasing force exposure catabolically shifts single-cell mRNA levels of aggrecan, collagen IIa, and tissue inhibitor of metalloproteinase-1. Cytohistochemistry reveals that the majority of strain experienced by the cell is also experienced by the nucleus, resulting in considerable changes in nuclear volume and structure. Transforming growth factor-beta1 and insulin-like growth factor-I offer mechanoprotection and recovery of gene expression of aggrecan and metalloproteinase-1. These results suggest that forces directly influence gene transcription and may do so by changing chromatin conformation.
Collapse
|
40
|
Sauerland K, Steinmeyer J. Intermittent mechanical loading of articular cartilage explants modulates chondroitin sulfate fine structure. Osteoarthritis Cartilage 2007; 15:1403-9. [PMID: 17574451 DOI: 10.1016/j.joca.2007.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Accepted: 05/01/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Alterations in the sulfation pattern of chondroitin sulfate (CS) chains of proteoglycans have been associated with aging and degeneration of articular cartilage. The purpose of the present study was to investigate systematically the effect of load amplitudes, frequencies and load durations of intermittently applied mechanical pressure on the sulfation of CS chains of cultured bovine articular cartilage explants. METHODS Using a sinusoidal waveform of 0.5 Hz frequency, cyclic compressive pressure of 0.1-1.0 MPa was applied for 10s followed by a period of unloading lasting 10-1000 s. These intermittent loading protocols were repeated for a total duration of 1-6 days. Newly synthesized as well as endogenous CS chains were isolated, depolymerized and subsequently quantitated after fractionation by high-performance anion-exchange chromatography. RESULTS Increasing the mechanical demands on cartilage explants by elevating either the duration or the frequency of loading can significantly alter the fine structure of newly synthesized CS in that less chains terminate on galNAc4,6S and, in that simultaneously the ratio of the internal disaccharides DeltaDi6S to DeltaDi4S is increased. Similar results were obtained with explants being slightly mechanically challenged by low magnitudes of loads. CONCLUSION Our data show for the first time that intermittent loading of articular cartilage explants can significantly alter the sulfation pattern of the terminal CS residues as well as of the internal disaccharides. Furthermore, our results indicate that explants possess a physiological window of stress in which they are able to produce also a normal extracellular matrix.
Collapse
Affiliation(s)
- K Sauerland
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University Clinics Giessen and Marburg GmbH, Paul-Meimberg-Strasse 3, 35385 Giessen, Germany
| | | |
Collapse
|
41
|
Zhou Y, Millward-Sadler SJ, Lin H, Robinson H, Goldring M, Salter DM, Nuki G. Evidence for JNK-dependent up-regulation of proteoglycan synthesis and for activation of JNK1 following cyclical mechanical stimulation in a human chondrocyte culture model. Osteoarthritis Cartilage 2007; 15:884-93. [PMID: 17408985 DOI: 10.1016/j.joca.2007.02.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2005] [Accepted: 02/04/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To examine the expression of mitogen-activated protein kinases (MAPKs) in human chondrocytes, to investigate whether selective activation of MAPKs is involved in up-regulation of proteoglycan (PG) synthesis following cyclical mechanical stimulation (MS), and to examine whether MS is associated with integrin-dependent or independent activation of MAPKs. METHODS The C-28/I2 and C-20/A4 human chondrocyte cell lines were mechanically stimulated in monolayer cell culture. PG synthesis was assessed by [(35)S]-sulphate incorporation in the presence and absence of the p38 inhibitor SB203580, and the extracellular-regulated kinase (ERK1/2) inhibitor PD98059. Kinase expression and activation were assessed by Western blotting using phosphorylation status-dependent and independent antibodies, and by kinase assays. The Jun N-terminal kinase (JNK) inhibitor SP600125 and the anti-beta(1) integrin (CD29) function-blocking antibody were used to assess JNK activation and integrin dependence, respectively. RESULTS Increased PG synthesis following 3 h of cyclic MS was abolished by pretreatment with 10 microM SB203580, but was not affected by 50 microM PD98059. The kinases p38, ERK1/ERK2 and JNKs were expressed in both stimulated and unstimulated cells. Phosphorylated p38 was detected at various time points following 0.5, 1, 2 and 3 h MS in C-28/I2, but not detected in C-20/A4 cell lines. Phosphorylation of ERK1 and ERK2 was not significantly affected by MS. Phosphorylation of the 54 and 46 kDa JNKs increased following 0.5, 1, 2 and 3 h of MS, and following CO(2) deprivation. MS-induced JNK phosphorylation was inhibited by SB203580 at concentrations > or =5 microM and activation of JNK1 following MS was blocked by SP600125 and partially inhibited by anti-CD29. CONCLUSIONS The data suggest JNK, rather than p38 or ERK dependent increases in PG synthesis, and selective, partially integrin-dependent, activation of JNK kinases in human chondrocyte cell lines following cyclical MS. JNK activation is also very sensitive to changes in CO(2)/pH in this chondrocyte culture model.
Collapse
Affiliation(s)
- Y Zhou
- University of Edinburgh, Osteoarticular Research Group, Queen's Medical Research Institute, Edinburgh, Scotland, UK
| | | | | | | | | | | | | |
Collapse
|
42
|
Vincent TL, McLean CJ, Full LE, Peston D, Saklatvala J. FGF-2 is bound to perlecan in the pericellular matrix of articular cartilage, where it acts as a chondrocyte mechanotransducer. Osteoarthritis Cartilage 2007; 15:752-63. [PMID: 17368052 DOI: 10.1016/j.joca.2007.01.021] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2006] [Accepted: 01/28/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We have shown previously that cutting or loading articular cartilage resulted in a fibroblast growth factor-2 (FGF-2) dependent activation of the extracellularly regulated kinase (ERK), and induction of a number of chondrocyte regulatory proteins including tissue inhibitor of metalloproteinase-1 and matrix metalloproteinases 1 and 3. An extracellular matrix-bound pool of FGF-2 was apparent, which could be liberated from the tissue by heparitinase (Vincent et al., Proc Natl Acad Sci U S A 2002;99(12):8259-64, Vincent et al., Arthritis Rheum 2004 Feb;50(2):526-33). Our objectives were to determine where FGF-2 was stored in articular cartilage, to which proteoglycan it was bound, and to elucidate its role in chondrocyte mechanotransduction. METHODS Immunohistochemistry and confocal microscopy were used to localise FGF-2 in the tissue. In vitro binding studies were performed using IASYS surface plasmon resonance. To study the role of pericellular FGF-2 in mechanotransduction cartilage explants or articular chondrocytes encapsulated in alginate were loaded using an in house loading rig. The loading response was assessed by the activation of ERK, in the presence or absence of a specific FGFR inhibitor. RESULTS Here we have identified perlecan as the heparan sulphate proteoglycan that sequesters FGF-2 in articular cartilage. Perlecan and FGF-2 co-localised within the type VI collagen-rich pericellular matrix of porcine and human articular cartilage. Chondrocytes encapsulated in alginate were able to accumulate pericellular perlecan and FGF-2 in culture, and deliver an FGF-dependent activation of ERK when loaded. CONCLUSION Loading-induced ERK activation was dependent upon the presence and concentration of pericellular FGF-2, suggesting a functional role for this matrix-bound growth factor in chondrocyte mechanotransduction.
Collapse
Affiliation(s)
- T L Vincent
- The Kennedy Institute of Rheumatology, Faculty of Medicine, Imperial College School of Science, Technology, and Medicine, London, UK.
| | | | | | | | | |
Collapse
|
43
|
Schulz RM, Bader A. Cartilage tissue engineering and bioreactor systems for the cultivation and stimulation of chondrocytes. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2007; 36:539-68. [PMID: 17318529 DOI: 10.1007/s00249-007-0139-1] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Revised: 01/23/2007] [Accepted: 01/29/2007] [Indexed: 12/14/2022]
Abstract
Damage to and degeneration of articular cartilage is a major health issue in industrialized nations. Articular cartilage has a particularly limited capacity for auto regeneration. At present, there is no established therapy for a sufficiently reliable and durable replacement of damaged articular cartilage. In this, as well as in other areas of regenerative medicine, tissue engineering methods are considered to be a promising therapeutic component. Nevertheless, there remain obstacles to the establishment of tissue-engineered cartilage as a part of the routine therapy for cartilage defects. One necessary aspect of potential tissue engineering-based therapies for cartilage damage that requires both elucidation and progress toward practical solutions is the reliable, cost effective cultivation of suitable tissue. Bioreactors and associated methods and equipment are the tools with which it is hoped that such a supply of tissue-engineered cartilage can be provided. The fact that in vivo adaptive physical stimulation influences chondrocyte function by affecting mechanotransduction leads to the development of specifically designed bioreactor devices that transmit forces like shear, hydrostatic pressure, compression, and combinations thereof to articular and artificial cartilage in vitro. This review summarizes the basic knowledge of chondrocyte biology and cartilage dynamics together with the exploration of the various biophysical principles of cause and effect that have been integrated into bioreactor systems for the cultivation and stimulation of chondrocytes.
Collapse
Affiliation(s)
- Ronny Maik Schulz
- Department of Cell Techniques and Applied Stem Cell Biology, Center of Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany.
| | | |
Collapse
|
44
|
Monfort J, Garcia-Giralt N, López-Armada MJ, Monllau JC, Bonilla A, Benito P, Blanco FJ. Decreased metalloproteinase production as a response to mechanical pressure in human cartilage: a mechanism for homeostatic regulation. Arthritis Res Ther 2007; 8:R149. [PMID: 16972994 PMCID: PMC1779454 DOI: 10.1186/ar2042] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2006] [Revised: 08/08/2006] [Accepted: 09/14/2006] [Indexed: 01/09/2023] Open
Abstract
Articular cartilage is optimised for bearing mechanical loads. Chondrocytes are the only cells present in mature cartilage and are responsible for the synthesis and integrity of the extracellular matrix. Appropriate joint loads stimulate chondrocytes to maintain healthy cartilage with a concrete protein composition according to loading demands. In contrast, inappropriate loads alter the composition of cartilage, leading to osteoarthritis (OA). Matrix metalloproteinases (MMPs) are involved in degradation of cartilage matrix components and have been implicated in OA, but their role in loading response is unclear. With this study, we aimed to elucidate the role of MMP-1 and MMP-3 in cartilage composition in response to mechanical load and to analyse the differences in aggrecan and type II collagen content in articular cartilage from maximum- and minimum-weight-bearing regions of human healthy and OA hips. In parallel, we analyse the apoptosis of chondrocytes in maximal and minimal load areas. Because human femoral heads are subjected to different loads at defined sites, both areas were obtained from the same hip and subsequently evaluated for differences in aggrecan, type II collagen, MMP-1, and MMP-3 content (enzyme-linked immunosorbent assay) and gene expression (real-time polymerase chain reaction) and for chondrocyte apoptosis (flow cytometry, bcl-2 Western blot, and mitochondrial membrane potential analysis). The results showed that the load reduced the MMP-1 and MMP-3 synthesis (p < 0.05) in healthy but not in OA cartilage. No significant differences between pressure areas were found for aggrecan and type II collagen gene expression levels. However, a trend toward significance, in the aggrecan/collagen II ratio, was found for healthy hips (p = 0.057) upon comparison of pressure areas (loaded areas > non-loaded areas). Moreover, compared with normal cartilage, OA cartilage showed a 10- to 20-fold lower ratio of aggrecan to type II collagen, suggesting that the balance between the major structural proteins is crucial to the integrity and function of the tissue. Alternatively, no differences in apoptosis levels between loading areas were found – evidence that mechanical load regulates cartilage matrix composition but does not affect chondrocyte viability. The results suggest that MMPs play a key role in regulating the balance of structural proteins of the articular cartilage matrix according to local mechanical demands.
Collapse
Affiliation(s)
- Jordi Monfort
- Unitat de recerca en fisiopatologia òssia i articular- Institut Municipal d'Investigació Mèdica (URFOA-IMIM), Hospital del Mar, Universitat Autònoma de Barcelona, Dr. Aiguader 80, 08003-Barcelona, Spain
| | - Natalia Garcia-Giralt
- Unitat de recerca en fisiopatologia òssia i articular- Institut Municipal d'Investigació Mèdica (URFOA-IMIM), Hospital del Mar, Universitat Autònoma de Barcelona, Dr. Aiguader 80, 08003-Barcelona, Spain
| | - María J López-Armada
- Osteoarticular and Aging Research Unit, Rheumatology Division, Biomedical Researcher Center, Complejo Hospitalario Universitario Juan Canalejo, Xubias 84, 15006 – A, Coruña, Spain
| | - Joan C Monllau
- Unitat de recerca en fisiopatologia òssia i articular- Institut Municipal d'Investigació Mèdica (URFOA-IMIM), Hospital del Mar, Universitat Autònoma de Barcelona, Dr. Aiguader 80, 08003-Barcelona, Spain
| | - Angeles Bonilla
- Osteoarticular and Aging Research Unit, Rheumatology Division, Biomedical Researcher Center, Complejo Hospitalario Universitario Juan Canalejo, Xubias 84, 15006 – A, Coruña, Spain
| | - Pere Benito
- Unitat de recerca en fisiopatologia òssia i articular- Institut Municipal d'Investigació Mèdica (URFOA-IMIM), Hospital del Mar, Universitat Autònoma de Barcelona, Dr. Aiguader 80, 08003-Barcelona, Spain
| | - Francisco J Blanco
- Osteoarticular and Aging Research Unit, Rheumatology Division, Biomedical Researcher Center, Complejo Hospitalario Universitario Juan Canalejo, Xubias 84, 15006 – A, Coruña, Spain
| |
Collapse
|
45
|
Stoddart MJ, Ettinger L, Häuselmann HJ. Enhanced matrix synthesis in de novo, scaffold free cartilage-like tissue subjected to compression and shear. Biotechnol Bioeng 2007; 95:1043-51. [PMID: 16804949 DOI: 10.1002/bit.21052] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Production of a de novo cartilage-like tissue construct is a goal for the repair of traumatic chondral defects. We aimed to enhance the matrix synthesis within a scaffold free, de novo cartilage-like tissue construct by way of mechanical load. A novel loading machine that enables the application of shear, as well as compression, was used to subject tissue engineered cartilage-like tissue to mechanical stress. The machine, which applies the load through a roller mechanism, can load up to 20 constructs with four different loading patterns simultaneously. The expression of mRNA encoding matrix products, and subsequent changes in matrix protein content, were analyzed after various loading regimes. The force applied to the immature tissue had a direct bearing on the short-term (first 4 h) response. A load of 0.5 N caused an increase in collagen II and aggrecan mRNA within an hour, with a peak at 2 h. This increased mRNA expression was translated into an increase of up to 60% in the glycosaminoglycan content of the optimally loaded constructs after 4 days of intermittent cyclical loading. Introducing pauses between load cycles reproducibly lead to an increase in GAG/DNA. In contrast, constant cyclical load, with no pause, lead to a decrease in the final glycosaminoglycan content compared with unloaded controls. Our data suggest that a protocol of mechanical stimulation, simulating in vivo conditions and involving shear and compression, may be a useful mechanism to enhance the properties of tissue engineered tissue prior to implantation.
Collapse
Affiliation(s)
- Martin James Stoddart
- Laboratory for Experimental Cartilage Research, Centre for Rheumatology and Bone Disease, Klinik Im Park, Zürich, Bellariastrasse 38, CH-8038 Zürich, Switzerland
| | | | | |
Collapse
|
46
|
Wolf A, Ackermann B, Steinmeyer J. Collagen synthesis of articular cartilage explants in response to frequency of cyclic mechanical loading. Cell Tissue Res 2006; 327:155-66. [PMID: 16941123 DOI: 10.1007/s00441-006-0251-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2006] [Accepted: 05/18/2006] [Indexed: 10/24/2022]
Abstract
Articular cartilage in vivo experiences the effects of both cell-regulatory proteins and mechanical forces. This study has addressed the hypothesis that the frequency of intermittently or continuously applied mechanical loads is a critical parameter in the regulation of chondrocyte collagen biosynthesis. Cyclic compressive pressure was applied intermittently to bovine articular cartilage explants by using a sinusoidal waveform of 0.1-1.0 Hz frequency with a peak stress of 0.5 MPa for a period of 5-20 s followed by a load-free period of 10-1,000 s. These loading protocols were repeated for a total duration of 6 days. In separate experiments, cyclic loading was continuously applied by using a sinusoidal waveform of 0.001-0.5 Hz frequency and a peak stress of 1.0 MPa for a period of 3 days. Unloaded cartilage discs of the same condyle were cultured in identically constructed loading chambers and served as controls. We report quantitative data showing that (1) no correlation exists between the relative rate of collagen synthesis expressed as the proportion of newly synthesized collagen among newly made proteins and either the frequency of intermittently or continuously applied loads or the overall time cartilage is actively loaded, and (2) individual protocols of intermittently applied loads can reduce the relative rate of collagen synthesis and increase the water content, whereas (3) continuously applied cyclic loads always suppress the relative rate of collagen synthesis compared with that of unloaded control specimens. The results provide further experimental evidence that collagen metabolism is difficult to manipulate by mechanical stimuli. This is physiologically important for the maintainance of the material properties of collagen in view of the heavy mechanical demands made upon it. Moreover, the unaltered or reduced collagen synthesis of cartilage explants might reflect more closely the metabolism of normal or early human osteoarthritic cartilage.
Collapse
Affiliation(s)
- Amela Wolf
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University Clinics Giessen and Marburg, Paul-Meimberg-Strasse 3, 35385 Giessen, Germany
| | | | | |
Collapse
|
47
|
Aufderheide AC, Athanasiou KA. A Direct Compression Stimulator for Articular Cartilage and Meniscal Explants. Ann Biomed Eng 2006; 34:1463-74. [PMID: 16897420 DOI: 10.1007/s10439-006-9157-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 06/22/2006] [Indexed: 11/29/2022]
Abstract
This paper describes the development and use of a direct compression stimulator for culturing explants from the meniscus of the knee and articular cartilage. Following design and fabrication of the instrument along with its data acquisition system, the function of the machine was verified by both mechanical means and tissue effect. The loading chamber can hold up to 45 5 mm diameter samples. While designed to stimulate samples up to 4 mm thick, axial displacements as little as 0.127 microm are within the theoretical capacity of the stimulator. In gene expression studies, collagen II and aggrecan expression were examined in explants from articular cartilage as well as medial and lateral menisci subjected to dynamic stimulation and static compression. These results were then compared to free swelling samples. It was found that static compression to cut thickness down-regulated aggrecan and collagen II expression in articular cartilage explants compared to free swelling controls by 94% and 90%, respectively. The application of a dynamic, intermittent, 2% oscillation around the cut thickness returned expression levels to those of free swelling controls at 4 h but not at 76 h. In medial meniscus samples, dynamic compression up-regulated aggrecan expression by 108%, but not collagen II expression, at 4 and 76 h compared to static controls. No difference in gene expression was observed for lateral meniscal explants. Thus, effects of direct compression seen in articular cartilage may not necessarily translate to the knee meniscus. The design of this stimulator will allow a variety of tissues and loading regimens to be examined. It is hoped that regimens can be found that not only return samples to the production levels of free swelling controls, but also surpass them in terms of gene expression, protein synthesis, and functional properties.
Collapse
Affiliation(s)
- Adam C Aufderheide
- Department of Bioengineering, Rice University, Houston, Texas 77251, USA
| | | |
Collapse
|
48
|
Fitzgerald JB, Jin M, Grodzinsky AJ. Shear and Compression Differentially Regulate Clusters of Functionally Related Temporal Transcription Patterns in Cartilage Tissue. J Biol Chem 2006; 281:24095-103. [PMID: 16782710 DOI: 10.1074/jbc.m510858200] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chondrocytes are subjected to a variety of biophysical forces and flows during physiological joint loading, including mechanical deformation, fluid flow, hydrostatic pressure, and streaming potentials; however, the role of these physical stimuli in regulating chondrocyte behavior is still being elucidated. To isolate the effects of these forces, we subjected intact cartilage explants to 1-24 h of continuous dynamic compression or dynamic shear loading at 0.1 Hz. We then measured the transcription levels of 25 genes known to be involved in cartilage homeostasis using real-time PCR and compared the gene expression profiles obtained from dynamic compression, dynamic shear, and our recent results on static compression amplitude and duration. Using clustering analysis, we determined that transcripts for proteins with similar function had correlated responses to loading. However, the temporal expression patterns were strongly dependent on the type of loading applied. Most matrix proteins were up-regulated by 24 h of dynamic compression or dynamic shear, but down-regulated by 24 h of 50% static compression, suggesting that cyclic matrix deformation is a key stimulator of matrix protein expression. Most matrix proteases were up-regulated by 24 h under all loading types. Transcription factors c-Fos and c-Jun maximally responded within 1 h to all loading types. Pre-incubating cartilage explants with either a chelator of intracellular calcium or an inhibitor of the cyclic AMP pathway demonstrated the involvement of both pathways in transcription induced by dynamic loading.
Collapse
Affiliation(s)
- Jonathan B Fitzgerald
- Biological Engineering Division, Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
49
|
Wu JZ, Herzog W. Analysis of the mechanical behavior of chondrocytes in unconfined compression tests for cyclic loading. J Biomech 2006; 39:603-16. [PMID: 16439231 DOI: 10.1016/j.jbiomech.2005.01.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2004] [Accepted: 01/16/2005] [Indexed: 10/25/2022]
Abstract
Experimental evidence indicates that the biosynthetic activity of chondrocytes is associated with the mechanical environment. For example, excessive, repetitive loading has been found to induce cell death, morphological and cellular damage, as seen in degenerative joint disease, while cyclic, physiological-like loading has been found to trigger a partial recovery of morphological and ultrastructural aspects in osteoarthritic human articular chondrocytes. Mechanical stimuli are believed to influence the biosynthetic activity via the deformation of cells. However, the in situ deformation of chondrocytes for cyclic loading conditions has not been investigated experimentally or theoretically. The purpose of the present study was to simulate the mechanical response of chondrocytes to cyclic loading in unconfined compression tests using a finite element model. The material properties of chondrocytes and extracellular matrix were considered to be biphasic. The time-histories of the shape and volume variations of chondrocytes at three locations (i.e., surface, center, and bottom) within the cartilage were predicted for static and cyclic loading conditions at two frequencies (0.02 and 0.1 Hz) and two amplitudes (0.1 and 0.2 MPa). Our results show that cells at different depths within the cartilage deform differently during cyclic loading, and that the depth dependence of cell deformation is influenced by the amplitude of the cyclic loading. Cell deformations under cyclic loading of 0.02 Hz were found to be similar to those at 0.1 Hz. We conclude from the simulation results that, in homogeneous cartilage layers, cell deformations are location-dependent, and further are affected by load magnitude. In physiological conditions, the mechanical environment of cells are even more complex due to the anisotropy, depth-dependent inhomogeneity, and tension-compression non-linearity of the cartilage matrix. Therefore, it is feasible to speculate that biosynthetic responses of chondrocytes to cyclic loading depend on cell location and load magnitude.
Collapse
Affiliation(s)
- John Z Wu
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26505, USA.
| | | |
Collapse
|
50
|
Huser CAM, Davies ME. Validation of an in vitro single-impact load model of the initiation of osteoarthritis-like changes in articular cartilage. J Orthop Res 2006; 24:725-32. [PMID: 16514652 DOI: 10.1002/jor.20111] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The objective of this study was the development and characterization of an in vitro model of the initiation of traumatic osteoarthritis (OA). Articular cartilage was obtained from seven healthy horses and from four horses diagnosed with OA. Cartilage disks were subjected to a single-impact load (500 g from 25, 50, or 100 mm) using a simple drop-tower device and cultured in vitro for up to 20 days. Cartilage sections were examined histologically to observe surface damage and proteoglycan loss. Percentage cell death was determined using TUNEL, release of glycosaminoglycans (GAG) to the medium was measured using the DMMB assay, and percentage weight gain calculated. Following a single-impact load and subsequent culture in vitro, articular cartilage explants demonstrated characteristic surface damage, proteoglycan loss, and chondrocyte death. This closely resembled degenerative changes observed in OA cartilage samples. A kinetic study showed that these degenerative changes (increased weight gain, GAG release into the medium, and chondrocyte death) were initiated within 48 h following impact and increased with recovery time in culture. These parameters were proportional to impact height, that is, impact energy. In conclusion, articular cartilage disks subjected to a single-impact load followed by 48 h of recovery time in culture in vitro developed traumatic OA-like changes. These changes can be quantified and compared, making the in vitro single-impact load model a useful tool for the elucidation of the early molecular pathways involved in the process leading from trauma to cartilage degeneration.
Collapse
Affiliation(s)
- Camille A M Huser
- Comparative Orthopaedics Research Group, Department of Veterinary Medicine, University of Cambridge, Cambridge, CB3 0ES, United Kingdom.
| | | |
Collapse
|