1
|
Hamada M, Eskelinen ASA, Florea C, Mikkonen S, Nieminen P, Grodzinsky AJ, Tanska P, Korhonen RK. Loss of collagen content is localized near cartilage lesions on the day of injurious loading and intensified on day 12. J Orthop Res 2024. [PMID: 39312444 DOI: 10.1002/jor.25975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024]
Abstract
Joint injury can lead to articular cartilage damage, excessive inflammation, and post-traumatic osteoarthritis (PTOA). Collagen is an essential component for cartilage function, yet current literature has limited understanding of how biochemical and biomechanical factors contribute to collagen loss in injured cartilage. Our aim was to investigate spatially dependent changes in collagen content and collagen integrity of injured cartilage, with an explant model of early-stage PTOA. We subjected calf knee cartilage explants to combinations of injurious loading (INJ), interleukin-1α-challenge (IL) and physiological cyclic loading (CL). Using Fourier transform infrared microspectroscopy, collagen content (Amide I band) and collagen integrity (Amide II/1338 cm-1 ratio) were estimated on days 0 and 12 post-injury. We found that INJ led to lower collagen content near lesions compared to intact regions on day 0 (p < 0.001). On day 12, near-lesion collagen content was lower compared to day 0 (p < 0.05). Additionally, on day 12, INJ, IL, and INJ + IL groups exhibited lower collagen content along most of tissue depth compared to free-swelling control group (p < 0.05). CL groups showed higher collagen content along most of tissue depth compared to corresponding groups without CL (p < 0.05). Immunohistochemical analysis revealed higher MMP-1 and MMP-3 staining intensities localized within cell lacunae in INJ group compared to CTRL group on day 0. Our results suggest that INJ causes rapid loss of collagen content near lesions, which is intensified on day 12. Additionally, CL could mitigate the loss of collagen content at intact regions after 12 days.
Collapse
Affiliation(s)
- Moustafa Hamada
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Atte S A Eskelinen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Cristina Florea
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Santtu Mikkonen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Petteri Nieminen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Alan J Grodzinsky
- Departments of Biological Engineering, Electrical Engineering and Computer Science, and Mechanical Engineering, Massachusetts Institute of Technology, Massachusetts Avenue, Cambridge, Massachusetts, USA
| | - Petri Tanska
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Rami K Korhonen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
2
|
Ayala S, Matan SO, Delco ML, Fortier LA, Cohen I, Bonassar LJ. Degradation of lubricating molecules in synovial fluid alters chondrocyte sensitivity to shear strain. J Orthop Res 2024. [PMID: 39182184 DOI: 10.1002/jor.25960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/01/2024] [Accepted: 08/03/2024] [Indexed: 08/27/2024]
Abstract
Articular joints facilitate motion and transfer loads to underlying bone through a combination of cartilage tissue and synovial fluid, which together generate a low-friction contact surface. Traumatic injury delivered to cartilage and the surrounding joint capsule causes secretion of proinflammatory cytokines by chondrocytes and the synovium, triggering cartilage matrix breakdown and impairing the ability of synovial fluid to lubricate the joint. Once these inflammatory processes become chronic, posttraumatic osteoarthritis (PTOA) development begins. However, the exact mechanism by which negative alterations to synovial fluid leads to PTOA pathogenesis is not fully understood. We hypothesize that removing the lubricating macromolecules from synovial fluid alters the relationship between mechanical loads and subsequent chondrocyte behavior in injured cartilage. To test this hypothesis, we utilized an ex vivo model of PTOA that involves subjecting cartilage explants to a single rapid impact followed by continuous articulation within a lubricating bath of either healthy synovial fluid, phosphate-buffered saline (PBS), synovial fluid treated with hyaluronidase, or synovial fluid treated with trypsin. These treatments degrade the main macromolecules attributed with providing synovial fluid with its lubricating properties; hyaluronic acid and lubricin. Explants were then bisected and fluorescently stained to assess global and depth-dependent cell death, caspase activity, and mitochondrial depolarization. Explants were tested via confocal elastography to determine the local shear strain profile generated in each lubricant. These results show that degrading hyaluronic acid or lubricin in synovial fluid significantly increases middle zone chondrocyte damage and shear strain loading magnitudes, while also altering chondrocyte sensitivity to loading.
Collapse
Affiliation(s)
- Steven Ayala
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Salman O Matan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Michelle L Delco
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Lisa A Fortier
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Itai Cohen
- Department of Physics, Cornell University, Ithaca, New York, USA
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
3
|
Rai MF, Collins KH, Lang A, Maerz T, Geurts J, Ruiz-Romero C, June RK, Ramos Y, Rice SJ, Ali SA, Pastrello C, Jurisica I, Thomas Appleton C, Rockel JS, Kapoor M. Three decades of advancements in osteoarthritis research: insights from transcriptomic, proteomic, and metabolomic studies. Osteoarthritis Cartilage 2024; 32:385-397. [PMID: 38049029 DOI: 10.1016/j.joca.2023.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a complex disease involving contributions from both local joint tissues and systemic sources. Patient characteristics, encompassing sociodemographic and clinical variables, are intricately linked with OA rendering its understanding challenging. Technological advancements have allowed for a comprehensive analysis of transcripts, proteomes and metabolomes in OA tissues/fluids through omic analyses. The objective of this review is to highlight the advancements achieved by omic studies in enhancing our understanding of OA pathogenesis over the last three decades. DESIGN We conducted an extensive literature search focusing on transcriptomics, proteomics and metabolomics within the context of OA. Specifically, we explore how these technologies have identified individual transcripts, proteins, and metabolites, as well as distinctive endotype signatures from various body tissues or fluids of OA patients, including insights at the single-cell level, to advance our understanding of this highly complex disease. RESULTS Omic studies reveal the description of numerous individual molecules and molecular patterns within OA-associated tissues and fluids. This includes the identification of specific cell (sub)types and associated pathways that contribute to disease mechanisms. However, there remains a necessity to further advance these technologies to delineate the spatial organization of cellular subtypes and molecular patterns within OA-afflicted tissues. CONCLUSIONS Leveraging a multi-omics approach that integrates datasets from diverse molecular detection technologies, combined with patients' clinical and sociodemographic features, and molecular and regulatory networks, holds promise for identifying unique patient endophenotypes. This holistic approach can illuminate the heterogeneity among OA patients and, in turn, facilitate the development of tailored therapeutic interventions.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Annemarie Lang
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jeroen Geurts
- Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Cristina Ruiz-Romero
- Grupo de Investigación de Reumatología (GIR), Unidad de Proteómica, INIBIC -Hospital Universitario A Coruña, SERGAS, Spain
| | - Ronald K June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, USA
| | - Yolande Ramos
- Dept. Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Sarah J Rice
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Shabana Amanda Ali
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Chiara Pastrello
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada; Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON, Canada
| | - C Thomas Appleton
- Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Jason S Rockel
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada.
| |
Collapse
|
4
|
Walton BL, Shattuck-Brandt R, Hamann CA, Tung VW, Colazo JM, Brand DD, Hasty KA, Duvall CL, Brunger JM. A programmable arthritis-specific receptor for guided articular cartilage regenerative medicine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578281. [PMID: 38352576 PMCID: PMC10862827 DOI: 10.1101/2024.01.31.578281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Objective Investigational cell therapies have been developed as disease-modifying agents for the treatment of osteoarthritis (OA), including those that inducibly respond to inflammatory factors driving OA progression. However, dysregulated inflammatory cascades do not specifically signify the presence of OA. Here, we deploy a synthetic receptor platform that regulates cell behaviors in an arthritis-specific fashion to confine transgene expression to sites characterized by cartilage degeneration. Methods An scFv specific for type II collagen (CII) was used to produce a synthetic Notch (synNotch) receptor that enables "CII-synNotch" mesenchymal stromal cells (MSCs) to recognize CII fibers exposed in damaged cartilage. Engineered cell activation by both CII-treated culture surfaces and on primary tissue samples was measured via inducible reporter transgene expression. TGFβ3-expressing cells were assessed for cartilage anabolic gene expression via qRT-PCR. In a co-culture with CII-synNotch MSCs engineered to express IL-1Ra, ATDC5 chondrocytes were stimulated with IL-1α, and inflammatory responses of ATDC5s were profiled via qRT-PCR and an NF-κB reporter assay. Results CII-synNotch MSCs are highly responsive to CII, displaying activation ranges over 40-fold in response to physiologic CII inputs. CII-synNotch cells exhibit the capacity to distinguish between healthy and damaged cartilage tissue and constrain transgene expression to regions of exposed CII fibers. Receptor-regulated TGFβ3 expression resulted in upregulation of Acan and Col2a1 in MSCs, and inducible IL-1Ra expression by engineered CII-synNotch MSCs reduced pro-inflammatory gene expression in chondrocytes. Conclusion This work demonstrates proof-of-concept that the synNotch platform guides MSCs for spatially regulated, disease-dependent delivery of OA-relevant biologic drugs.
Collapse
Affiliation(s)
- Bonnie L Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | | | - Catherine A Hamann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Victoria W Tung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - David D Brand
- Research Service, Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN 38105, USA
| | - Karen A Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis VA Medical Center, Memphis, TN, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
- Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
- Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, 37212, USA
| |
Collapse
|
5
|
Rydén M, Lindblom K, Yifter-Lindgren A, Turkiewicz A, Aspberg A, Tillgren V, Englund M, Önnerfjord P. A human meniscus explant model for studying early events in osteoarthritis development by proteomics. J Orthop Res 2023; 41:2765-2778. [PMID: 37218349 DOI: 10.1002/jor.25633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 05/24/2023]
Abstract
Degenerative meniscus lesions have been associated with both osteoarthritis etiology and its progression. We, therefore, sought to establish a human meniscus ex vivo model to study the meniscal response to cytokine treatment using a proteomics approach. Lateral menisci were obtained from five knee-healthy donors. The meniscal body was cut into vertical slices and further divided into an inner (avascular) and outer region. Explants were either left untreated (controls) or stimulated with cytokines. Medium changes were conducted every 3 days up to Day 21 and liquid chromatography-mass spectrometry was performed at all the time points for the identification and quantification of proteins. Mixed-effect linear regression models were used for statistical analysis to estimate the effect of treatments versus control on protein abundance. Treatment by IL1ß increased release of cytokines such as interleukins, chemokines, and matrix metalloproteinases but a limited catabolic effect in healthy human menisci explants. Further, we observed an increased release of matrix proteins (collagens, integrins, prolargin, tenascin) in response to oncostatin M (OSM) + tumor necrosis factor (TNF) and TNF+interleukin-6 (IL6) + sIL6R treatments, and analysis of semitryptic peptides provided additional evidence of increased catabolic effects in response to these treatments. The induced activation of catabolic processes may play a role in osteoarthritis development.
Collapse
Affiliation(s)
- Martin Rydén
- Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Karin Lindblom
- Department of Clinical Sciences Lund, Section for Rheumatology and Molecular Skeletal Biology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Aida Yifter-Lindgren
- Department of Clinical Sciences Lund, Section for Rheumatology and Molecular Skeletal Biology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Aleksandra Turkiewicz
- Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Anders Aspberg
- Department of Clinical Sciences Lund, Section for Rheumatology and Molecular Skeletal Biology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Viveka Tillgren
- Department of Clinical Sciences Lund, Section for Rheumatology and Molecular Skeletal Biology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Martin Englund
- Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Patrik Önnerfjord
- Department of Clinical Sciences Lund, Section for Rheumatology and Molecular Skeletal Biology, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
6
|
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: 2.0] [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
|
7
|
Selig M, Azizi S, Walz K, Lauer JC, Rolauffs B, Hart ML. Cell morphology as a biological fingerprint of chondrocyte phenotype in control and inflammatory conditions. Front Immunol 2023; 14:1102912. [PMID: 36860844 PMCID: PMC9968733 DOI: 10.3389/fimmu.2023.1102912] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction Little is known how inflammatory processes quantitatively affect chondrocyte morphology and how single cell morphometric data could be used as a biological fingerprint of phenotype. Methods We investigated whether trainable high-throughput quantitative single cell morphology profiling combined with population-based gene expression analysis can be used to identify biological fingerprints that are discriminatory of control vs. inflammatory phenotypes. The shape of a large number of chondrocytes isolated from bovine healthy and human osteoarthritic (OA) cartilages was quantified under control and inflammatory (IL-1β) conditions using a trainable image analysis technique measuring a panel of cell shape descriptors (area, length, width, circularity, aspect ratio, roundness, solidity). The expression profiles of phenotypically relevant markers were quantified by ddPCR. Statistical analysis, multivariate data exploration, and projection-based modelling were used for identifying specific morphological fingerprints indicative of phenotype. Results Cell morphology was sensitive to both cell density and IL-1β. In both cell types, all shape descriptors correlated with expression of extracellular matrix (ECM)- and inflammatory-regulating genes. A hierarchical clustered image map revealed that individual samples sometimes responded differently in control or IL-1β conditions than the overall population. Despite these variances, discriminative projection-based modeling revealed distinct morphological fingerprints that discriminated between control and inflammatory chondrocyte phenotypes: the most essential morphological characteristics attributable to non-treated control cells was a higher cell aspect ratio in healthy bovine chondrocytes and roundness in OA human chondrocytes. In contrast, a higher circularity and width in healthy bovine chondrocytes and length and area in OA human chondrocytes indicated an inflammatory (IL-1β) phenotype. When comparing the two species/health conditions, bovine healthy and human OA chondrocytes exhibited comparable IL-1β-induced morphologies in roundness, a widely recognized marker of chondrocyte phenotype, and aspect ratio. Discussion Overall, cell morphology can be used as a biological fingerprint for describing chondrocyte phenotype. Quantitative single cell morphometry in conjunction with advanced methods for multivariate data analysis allows identifying morphological fingerprints that can discriminate between control and inflammatory chondrocyte phenotypes. This approach could be used to assess how culture conditions, inflammatory mediators, and therapeutic modulators regulate cell phenotype and function.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany.,Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Saman Azizi
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Kathrin Walz
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Jasmin C Lauer
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany.,Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Melanie L Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
8
|
Kurz B, Hart ML, Rolauffs B. Mechanical Articular Cartilage Injury Models and Their Relevance in Advancing Therapeutic Strategies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:107-124. [PMID: 37052850 DOI: 10.1007/978-3-031-25588-5_8] [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
This chapter details how Alan Grodzinsky and his team unraveled the complex electromechanobiological structure-function relationships of articular cartilage and used these insights to develop an impressively versatile shear and compression model. In this context, this chapter focuses (i) on the effects of mechanical compressive injury on multiple articular cartilage properties for (ii) better understanding the molecular concept of mechanical injury, by studying gene expression, signal transduction and the release of potential injury biomarkers. Furthermore, we detail how (iii) this was used to combine mechanical injury with cytokine exposure or co-culture systems for generating a more realistic trauma model to (iv) investigate the therapeutic modulation of the injurious response of articular cartilage. Impressively, Alan Grodzinsky's research has been and will remain to be instrumental in understanding the proinflammatory response to injury and in developing effective therapies that are based on an in-depth understanding of complex structure-function relationships that underlay articular cartilage function and degeneration.
Collapse
Affiliation(s)
- Bodo Kurz
- Department of Anatomy, Christian-Albrechts-University, Kiel, Germany.
| | - Melanie L Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
9
|
Rydén M, Önnerfjord P. In Vitro Models and Proteomics in Osteoarthritis Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:57-68. [PMID: 37052846 DOI: 10.1007/978-3-031-25588-5_4] [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
This review summarizes and exemplifies the current understanding of osteoarthritis in vitro models and describes their relevance for new insights in the future of osteoarthritis research. Our friend and highly appreciated colleague, Prof. Alan Grodzinsky has contributed greatly to the understanding of joint tissue biology and cartilage biomechanics. He frequently utilizes in vitro models and cartilage explant cultures, and recent work also includes proteomics studies. This review is dedicated to honor his 75-year birthday and will focus on recent proteomic in vitro studies related to osteoarthritis, and within this topic highlight some of his contributions to the field.
Collapse
Affiliation(s)
- Martin Rydén
- Orthopaedics, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Patrik Önnerfjord
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
10
|
Orozco GA, Eskelinen AS, Kosonen JP, Tanaka MS, Yang M, Link TM, Ma B, Li X, Grodzinsky AJ, Korhonen RK, Tanska P. Shear strain and inflammation-induced fixed charge density loss in the knee joint cartilage following ACL injury and reconstruction: A computational study. J Orthop Res 2022; 40:1505-1522. [PMID: 34533840 PMCID: PMC8926939 DOI: 10.1002/jor.25177] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 07/13/2021] [Accepted: 09/01/2021] [Indexed: 02/04/2023]
Abstract
Excessive tissue deformation near cartilage lesions and acute inflammation within the knee joint after anterior cruciate ligament (ACL) rupture and reconstruction surgery accelerate the loss of fixed charge density (FCD) and subsequent cartilage tissue degeneration. Here, we show how biomechanical and biochemical degradation pathways can predict FCD loss using a patient-specific finite element model of an ACL reconstructed knee joint exhibiting a chondral lesion. Biomechanical degradation was based on the excessive maximum shear strains that may result in cell apoptosis, while biochemical degradation was driven by the diffusion of pro-inflammatory cytokines. We found that the biomechanical model was able to predict substantial localized FCD loss near the lesion and on the medial areas of the lateral tibial cartilage. In turn, the biochemical model predicted FCD loss all around the lesion and at intact areas; the highest FCD loss was at the cartilage-synovial fluid-interface and decreased toward the deeper zones. Interestingly, simulating a downturn of an acute inflammatory response by reducing the cytokine concentration exponentially over time in synovial fluid led to a partial recovery of FCD content in the cartilage. Our novel numerical approach suggests that in vivo FCD loss can be estimated in injured cartilage following ACL injury and reconstruction. Our novel modeling platform can benefit the prediction of PTOA progression and the development of treatment interventions such as disease-modifying drug testing and rehabilitation strategies.
Collapse
Affiliation(s)
- Gustavo A. Orozco
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland Yliopistonranta 1, FI-70210 Kuopio, Finland,Department of Biomedical Engineering, Lund University, Box 188, 221 00, Lund, Sweden
| | - Atte S.A. Eskelinen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland Yliopistonranta 1, FI-70210 Kuopio, Finland
| | - Joonas P. Kosonen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland Yliopistonranta 1, FI-70210 Kuopio, Finland
| | - Matthew S. Tanaka
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 1500 Owens St, San Francisco, CA 94158, USA
| | - Mingrui Yang
- Department of Biomedical Engineering, Lerner Research Institute, Program of Advanced Musculoskeletal Imaging (PAMI), 9500 Euclid Avenue, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Thomas M. Link
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 1500 Owens St, San Francisco, CA 94158, USA
| | - Benjamin Ma
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, 1500 Owens St, San Francisco, CA 94158, USA
| | - Xiaojuan Li
- Department of Biomedical Engineering, Lerner Research Institute, Program of Advanced Musculoskeletal Imaging (PAMI), 9500 Euclid Avenue, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Alan J. Grodzinsky
- Departments of Biological Engineering, Electrical Engineering and Computer Science and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rami K. Korhonen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland Yliopistonranta 1, FI-70210 Kuopio, Finland
| | - Petri Tanska
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland Yliopistonranta 1, FI-70210 Kuopio, Finland
| |
Collapse
|
11
|
Wang Y, Guo X, Fan X, Zhang H, Xue D, Pan Z. The protective effect of mangiferin on osteoarthritis: An in vitro and in vivo study. Physiol Res 2022; 71:135-145. [PMID: 35043648 PMCID: PMC8997682 DOI: 10.33549/physiolres.934747] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/02/2021] [Indexed: 11/25/2022] Open
Abstract
Mangiferin is a kind of polyphenol chemical compound separated from these herbal medicines of Mangifera indica L., Anemarrhena asphodeloides Bge. and Belamcanda chinensis L., which has anti-inflammatory, anti-virus, and other physiological activities without toxic effects. Osteoarthritis (OA) is a chronic disease that is also a kind of arthritis disease in which articular cartilage or bones under the joint is damaged. In addition, artificial replacements are required in severe cases. At present, there are not too much researches on the potential biological activities of mangiferin that plays a protective role in the treatment of OA. In this study, we evaluated the protective effect of mangiferin on osteoarthritis (OA) in vitro and in vivo. First, the effect of different concentrations of mangiferin on rat chondrocytes was determined by MTT assay. Second, the effects of mangiferin on the expression levels of matrix metalloproteinase (MMP)-13, TNF alpha, Collagen II, Caspase-3, and cystatin-C in interleukin-1beta (IL-1beta)-induced rat chondrocytes were examined by the real-time polymerase chain reaction in vitro, meanwhile the effects of mangiferin on the nuclear factor kappa-B (NF-kappaB) signaling pathway were also investigated by Western Blot. Finally, the anti-osteoarthritic protective effect of mangiferin was evaluated in the rat model by anterior cruciate ligament transection (ACLT) combined with bilateral ovariectomy-induced OA in vivo. The results showed that the mangiferin was found to inhibit the expression of MMP-13, TNF-alpha, and Caspase-3 which also increased the expression of Collagen II and cystatin-C in IL 1beta induced rat chondrocytes. In addition, IL-1beta-induced activation of nuclear factor kappa-B (NF-kappaB) and the degradation of inhibitor of kappaB (IkappaB)-alpha were suppressed by mangiferin. For the in vivo study in a rat model of OA, 100 microl of mangiferin was administered by intra-articular injections for rats, the results showed that the cartilage degradation was suppressed by mangiferin through Micro CT and Histological Examination. According to both in vitro and in vivo results, mangiferin has a protective effect in the treatment of OA which may be a promising therapeutic agent for OA.
Collapse
Affiliation(s)
- Y Wang
- Department of Orthopaedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China. and
| | | | | | | | | | | |
Collapse
|
12
|
Arabiyat AS, Chen H, Erndt-Marino J, Burkhard K, Scola L, Fleck A, Wan LQ, Hahn MS. Hyperosmolar Ionic Solutions Modulate Inflammatory Phenotype and sGAG Loss in a Cartilage Explant Model. Cartilage 2021; 13:713S-721S. [PMID: 32975437 PMCID: PMC8804856 DOI: 10.1177/1947603520961167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The objective of this study was to compare the effects of hyperosmolar sodium (Na+), lithium (Li+) and potassium (K+) on catabolic and inflammatory osteoarthritis (OA) markers and sulfated glycosaminoglycan (sGAG) loss in TNF-α-stimulated cartilage explants. METHODS Explants from bovine stifle joints were stimulated with TNF-α for 1 day to induce cartilage degradation followed by supplementation with 50 mM potassium chloride (KCl), 50 mM lithium chloride (LiCl), 50 mM sodium chloride (NaCl), or 100 nM dexamethasone for an additional 6 days. We assessed the effect of TNF-α stimulation and hyperosmolar ionic treatment on sGAG loss and expression of OA-associated proteins: ADAMTS-5, COX-2, MMP-1, MMP-13, and VEGF. RESULTS TNF-α treatment increased sGAG loss (P < 0.001) and expression of COX-2 (P = 0.018), MMP-13 (P < 0.001), and VEGF (P = 0.017) relative to unstimulated controls. Relative to activated controls, LiCl and dexamethasone treatment attenuated sGAG loss (P = 0.008 and P = 0.042, respectively) and expression of MMP-13 (P = 0.005 and P = 0.036, respectively). In contrast, KCl treatment exacerbated sGAG loss (P = 0.032) and MMP-1 protein expression (P = 0.010). NaCl treatment, however, did not alter sGAG loss or expression of OA-related proteins. Comparing LiCl and KCl treatment shows a potent reduction (P < 0.05) in catabolic and inflammatory mediators following LiCl treatment. CONCLUSION These results suggest that these ionic species elicit varying responses in TNF-α-stimulated explants. Cumulatively, these findings support additional studies of hyperosmolar ionic solutions for potential development of novel intraarticular injections targeting OA.
Collapse
Affiliation(s)
- Ahmad S. Arabiyat
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Hongyu Chen
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Josh Erndt-Marino
- Department of Biomedical Engineering,
Tufts University, Medford, MA, USA
| | - Katie Burkhard
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
| | - Lisa Scola
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
| | - Allison Fleck
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Leo Q. Wan
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Mariah S. Hahn
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| |
Collapse
|
13
|
Hagemans FJ, Larsson S, Reijman M, Frobell RB, Struglics A, Meuffels DE. An Anterior Cruciate Ligament Rupture Increases Levels of Urine N-terminal Cross-linked Telopeptide of Type I Collagen, Urine C-terminal Cross-linked Telopeptide of Type II Collagen, Serum Aggrecan ARGS Neoepitope, and Serum Tumor Necrosis Factor-α. Am J Sports Med 2021; 49:3534-3543. [PMID: 34591687 PMCID: PMC8573615 DOI: 10.1177/03635465211042310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND An anterior cruciate ligament (ACL) rupture results in an increased risk of developing knee osteoarthritis (OA) at an early age. Before clinical signs become apparent, the OA process has already been initiated. Therefore, it is important to look at the cascade of changes, such as the activity of cytokines and proteases, which might be associated with the later development of OA. PURPOSE To compare biomarker levels in patients with a recent ACL rupture with those in controls with a healthy knee and to monitor biomarker levels over 2 years after an ACL rupture. STUDY DESIGN Descriptive laboratory study. METHODS Patients were enrolled after an ACL tear was identified. Serum and urine samples were collected at the time of enrollment in the study (3-25 weeks after the injury) and then at 14 and 27 months after the injury between January 2009 and November 2010. Reference samples were obtained from participants with healthy knees. The following biomarkers were measured with immunological assays: aggrecan ARGS neoepitope (ARGS-aggrecan), tumor necrosis factor-α (TNF-α), interferon-γ, interleukin (IL)-8, IL-10, IL-13, N-terminal cross-linked telopeptide of type I collagen (NTX-I), and C-terminal cross-linked telopeptide of type II collagen (CTX-II). RESULTS Samples were collected from 152 patients with an acute ACL rupture, who had a median age of 25 years (interquartile range [IQR], 21-32 years). There were 62 urine reference samples (median age, 25 years [IQR, 22-36 years]) and 26 serum reference samples (median age, 35 years [IQR, 24-39 years]). At a median of 11 weeks (IQR, 7-17 weeks) after trauma, serum levels of both ARGS-aggrecan and TNF-α were elevated 1.5-fold (P < .001) compared with reference samples and showed a time-dependent decrease during follow-up. Urine NTX-I and CTX-II concentrations were elevated in an early phase after trauma (1.3-fold [P < .001] and 3.7-fold [P < .001], respectively) compared with reference samples, and CTX-II levels remained elevated compared with reference samples at 2-year follow-up. Strong correlations were found between serum ARGS-aggrecan, urinary NTX-I, and urinary CTX-II (rs = 0.57-0.68). CONCLUSION In the first few months after an ACL injury, there was a measurable increase in serum levels of ARGS-aggrecan and TNF-α as well as urine levels of NTX-I and CTX-II. These markers remained high compared with those of controls with healthy knees at 2-year follow-up.
Collapse
Affiliation(s)
- Frans J.A. Hagemans
- Department of Orthopaedics and Sports Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands,Department of Orthopaedics, Center for Orthopaedic Research Alkmaar, Noordwest Ziekenhuisgroep, Alkmaar, the Netherlands
| | - Staffan Larsson
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Max Reijman
- Department of Orthopaedics and Sports Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Richard B. Frobell
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Andre Struglics
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Duncan E. Meuffels
- Department of Orthopaedics and Sports Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands,Duncan E. Meuffels, MD, PhD, Department of Orthopaedics and Sports Medicine, Erasmus University Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD, the Netherlands ()
| |
Collapse
|
14
|
Inflammatory signaling sensitizes Piezo1 mechanotransduction in articular chondrocytes as a pathogenic feed-forward mechanism in osteoarthritis. Proc Natl Acad Sci U S A 2021; 118:2001611118. [PMID: 33758095 PMCID: PMC8020656 DOI: 10.1073/pnas.2001611118] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis is a global health problem that affects load-bearing joints, causing loss of mobility and enormous healthcare costs. However, disease-modifying approaches are lacking. Here, we report a cellular mechanism of inflammatory signaling in chondrocytes, the cellular component of cartilage. We show how osteoarthritis-relevant levels of interleukin-1α reprogram articular chondrocytes so that they become more susceptible to mechanical trauma, which chondrocytes sense via Piezo1/2-mechanosensitive ion channels. We uncover that IL-1α enhances gene expression of Piezo1 in primary articular chondrocytes underlying Piezo1 enhanced function. We elucidate signaling from membrane to nucleus, including transcription factors that enhance Piezo1 expression. We also define consequences of increased expression of Piezo1, for mechanotransduction and at rest, that implicate this reprogramming mechanism in osteoarthritis pathogenesis. Osteoarthritis (OA) is a painful and debilitating condition of synovial joints without any disease-modifying therapies [A. M. Valdes, T. D. Spector, Nat. Rev. Rheumatol. 7, 23–32 (2011)]. We previously identified mechanosensitive PIEZO channels, PIEZO1 and PIEZO2, both expressed in articular cartilage, to function in chondrocyte mechanotransduction in response to injury [W. Lee et al., Proc. Natl. Acad. Sci. U.S.A. 111, E5114–E5122 (2014); W. Lee, F. Guilak, W. Liedtke, Curr. Top. Membr. 79, 263–273 (2017)]. We therefore asked whether interleukin-1–mediated inflammatory signaling, as occurs in OA, influences Piezo gene expression and channel function, thus indicative of maladaptive reprogramming that can be rationally targeted. Primary porcine chondrocyte culture and human osteoarthritic cartilage tissue were studied. We found that interleukin-1α (IL-1α) up-regulated Piezo1 in porcine chondrocytes. Piezo1 expression was significantly increased in human osteoarthritic cartilage. Increased Piezo1 expression in chondrocytes resulted in a feed-forward pathomechanism whereby increased function of Piezo1 induced excess intracellular Ca2+ at baseline and in response to mechanical deformation. Elevated resting state Ca2+ in turn rarefied the F-actin cytoskeleton and amplified mechanically induced deformation microtrauma. As intracellular substrates of this OA-related inflammatory pathomechanism, in porcine articular chondrocytes exposed to IL-1α, we discovered that enhanced Piezo1 expression depended on p38 MAP-kinase and transcription factors HNF4 and ATF2/CREBP1. CREBP1 directly bound to the proximal PIEZO1 gene promoter. Taken together, these signaling and genetic reprogramming events represent a detrimental Ca2+-driven feed-forward mechanism that can be rationally targeted to stem the progression of OA.
Collapse
|
15
|
Khella CM, Asgarian R, Horvath JM, Rolauffs B, Hart ML. An Evidence-Based Systematic Review of Human Knee Post-Traumatic Osteoarthritis (PTOA): Timeline of Clinical Presentation and Disease Markers, Comparison of Knee Joint PTOA Models and Early Disease Implications. Int J Mol Sci 2021; 22:1996. [PMID: 33671471 PMCID: PMC7922905 DOI: 10.3390/ijms22041996] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
Understanding the causality of the post-traumatic osteoarthritis (PTOA) disease process of the knee joint is important for diagnosing early disease and developing new and effective preventions or treatments. The aim of this review was to provide detailed clinical data on inflammatory and other biomarkers obtained from patients after acute knee trauma in order to (i) present a timeline of events that occur in the acute, subacute, and chronic post-traumatic phases and in PTOA, and (ii) to identify key factors present in the synovial fluid, serum/plasma and urine, leading to PTOA of the knee in 23-50% of individuals who had acute knee trauma. In this context, we additionally discuss methods of simulating knee trauma and inflammation in in vivo, ex vivo articular cartilage explant and in vitro chondrocyte models, and answer whether these models are representative of the clinical inflammatory stages following knee trauma. Moreover, we compare the pro-inflammatory cytokine concentrations used in such models and demonstrate that, compared to concentrations in the synovial fluid after knee trauma, they are exceedingly high. We then used the Bradford Hill Framework to present evidence that TNF-α and IL-6 cytokines are causal factors, while IL-1β and IL-17 are credible factors in inducing knee PTOA disease progresssion. Lastly, we discuss beneficial infrastructure for future studies to dissect the role of local vs. systemic inflammation in PTOA progression with an emphasis on early disease.
Collapse
Affiliation(s)
| | | | | | | | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (C.M.K.); (R.A.); (J.M.H.); (B.R.)
| |
Collapse
|
16
|
Anderson JR, Phelan MM, Foddy L, Clegg PD, Peffers MJ. Ex Vivo Equine Cartilage Explant Osteoarthritis Model: A Metabolomics and Proteomics Study. J Proteome Res 2020; 19:3652-3667. [PMID: 32701294 PMCID: PMC7476031 DOI: 10.1021/acs.jproteome.0c00143] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
Osteoarthritis is an age-related
degenerative musculoskeletal disease
characterized by loss of articular cartilage, synovitis, and subchondral
bone sclerosis. Osteoarthritis pathogenesis is yet to be fully elucidated
with no osteoarthritis-specific biomarkers in clinical use. Ex vivo equine cartilage explants (n =
5) were incubated in tumor necrosis factor-α (TNF-α)/interleukin-1β
(IL-1β)-supplemented culture media for 8 days, with the media
removed and replaced at 2, 5, and 8 days. Acetonitrile metabolite
extractions of 8 day cartilage explants and media samples at all time
points underwent one-dimensional (1D) 1H nuclear magnetic
resonance metabolomic analysis, with media samples also undergoing
mass spectrometry proteomic analysis. Within the cartilage, glucose
and lysine were elevated following TNF-α/IL-1β treatment,
while adenosine, alanine, betaine, creatine, myo-inositol, and uridine
decreased. Within the culture media, 4, 4, and 6 differentially abundant
metabolites and 154, 138, and 72 differentially abundant proteins
were identified at 1–2, 3–5, and 6–8 days, respectively,
including reduced alanine and increased isoleucine, enolase 1, vimentin,
and lamin A/C following treatment. Nine potential novel osteoarthritis
neopeptides were elevated in the treated media. Implicated pathways
were dominated by those involved in cellular movement. Our innovative
study has provided insightful information on early osteoarthritis
pathogenesis, enabling potential translation for clinical markers
and possible new therapeutic targets.
Collapse
Affiliation(s)
- James R Anderson
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, U.K
| | - Marie M Phelan
- NMR Metabolomics Facility, Technology Directorate & Department of Biochemistry & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, U.K
| | - Laura Foddy
- School of Veterinary Science, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool L69 3GH, U.K
| | - Peter D Clegg
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, U.K
| | - Mandy J Peffers
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, U.K
| |
Collapse
|
17
|
Eskelinen ASA, Tanska P, Florea C, Orozco GA, Julkunen P, Grodzinsky AJ, Korhonen RK. Mechanobiological model for simulation of injured cartilage degradation via pro-inflammatory cytokines and mechanical stimulus. PLoS Comput Biol 2020; 16:e1007998. [PMID: 32584809 PMCID: PMC7343184 DOI: 10.1371/journal.pcbi.1007998] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/08/2020] [Accepted: 05/28/2020] [Indexed: 01/12/2023] Open
Abstract
Post-traumatic osteoarthritis (PTOA) is associated with cartilage degradation, ultimately leading to disability and decrease of quality of life. Two key mechanisms have been suggested to occur in PTOA: tissue inflammation and abnormal biomechanical loading. Both mechanisms have been suggested to result in loss of cartilage proteoglycans, the source of tissue fixed charge density (FCD). In order to predict the simultaneous effect of these degrading mechanisms on FCD content, a computational model has been developed. We simulated spatial and temporal changes of FCD content in injured cartilage using a novel finite element model that incorporates (1) diffusion of the pro-inflammatory cytokine interleukin-1 into tissue, and (2) the effect of excessive levels of shear strain near chondral defects during physiologically relevant loading. Cytokine-induced biochemical cartilage explant degradation occurs near the sides, top, and lesion, consistent with the literature. In turn, biomechanically-driven FCD loss is predicted near the lesion, in accordance with experimental findings: regions near lesions showed significantly more FCD depletion compared to regions away from lesions (p<0.01). Combined biochemical and biomechanical degradation is found near the free surfaces and especially near the lesion, and the corresponding bulk FCD loss agrees with experiments. We suggest that the presence of lesions plays a role in cytokine diffusion-driven degradation, and also predisposes cartilage for further biomechanical degradation. Models considering both these cartilage degradation pathways concomitantly are promising in silico tools for predicting disease progression, recognizing lesions at high risk, simulating treatments, and ultimately optimizing treatments to postpone the development of PTOA.
Collapse
Affiliation(s)
| | - Petri Tanska
- Department of Applied Physics, University of Eastern Finland, Finland
| | - Cristina Florea
- Department of Applied Physics, University of Eastern Finland, Finland
- Departments of Biological Engineering, Electrical Engineering and Computer Science and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, United States of America
| | - Gustavo A. Orozco
- Department of Applied Physics, University of Eastern Finland, Finland
| | - Petro Julkunen
- Department of Applied Physics, University of Eastern Finland, Finland
- Department of Clinical Neurophysiology, Kuopio University Hospital, Finland
| | - Alan J. Grodzinsky
- Departments of Biological Engineering, Electrical Engineering and Computer Science and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, United States of America
| | - Rami K. Korhonen
- Department of Applied Physics, University of Eastern Finland, Finland
| |
Collapse
|
18
|
Haidar O, O'Neill N, Staunton CA, Bavan S, O'Brien F, Zouggari S, Sharif U, Mobasheri A, Kumagai K, Barrett-Jolley R. Pro-inflammatory Cytokines Drive Deregulation of Potassium Channel Expression in Primary Synovial Fibroblasts. Front Physiol 2020; 11:226. [PMID: 32265733 PMCID: PMC7105747 DOI: 10.3389/fphys.2020.00226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/27/2020] [Indexed: 01/15/2023] Open
Abstract
The synovium secretes synovial fluid, but is also richly innervated with nociceptors and acts as a gateway between avascular joint tissues and the circulatory system. Resident fibroblast-like synoviocytes' (FLS) calcium-activated potassium channels (K Ca) change in activity in arthritis models and this correlates with FLS activation. Objective To investigate this activation in an in vitro model of inflammatory arthritis; 72 h treatment with cytokines TNFα and IL1β. Methods FLS cells were isolated from rat synovial membranes. We analyzed global changes in FLS mRNA by RNA-sequencing, then focused on FLS ion channel genes and the corresponding FLS electrophysiological phenotype and finally modeling data with ingenuity pathway analysis (IPA) and MATLAB. Results IPA showed significant activation of inflammatory, osteoarthritic and calcium signaling canonical pathways by cytokines, and we identified ∼200 channel gene transcripts. The large K Ca (BK) channel consists of the pore forming Kcnma1 together with β-subunits. Following cytokine treatment, a significant increase in Kcnma1 RNA abundance was detected by qPCR and changes in several ion channels were detected by RNA-sequencing, including a loss of BK channel β-subunit expression Kcnmb1/2 and an increase in Kcnmb3. In electrophysiological experiments, there was a decrease in over-all current density at 20 mV without change in chord conductance at this potential. Conclusion TNFα and IL1β treatment of FLS in vitro recapitulated several common features of inflammatory arthritis at the transcriptomic level, including increase in Kcnma1 and Kcnmb3 gene expression.
Collapse
Affiliation(s)
- Omar Haidar
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Nathanael O'Neill
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Caroline A Staunton
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Selvan Bavan
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Fiona O'Brien
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Sarah Zouggari
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Umar Sharif
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, Finland.,Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.,Department of Orthopedics and Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht, Netherlands.,Versus Arthritis Centre for Sport, Exercise and Osteoarthritis Research, Queen's Medical Centre, Nottingham, United Kingdom
| | - Kosuke Kumagai
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.,Department of Orthopaedic Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Richard Barrett-Jolley
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.,Versus Arthritis Centre for Sport, Exercise and Osteoarthritis Research, Queen's Medical Centre, Nottingham, United Kingdom
| |
Collapse
|
19
|
Connizzo BK, Piet JM, Shefelbine SJ, Grodzinsky AJ. Age-associated changes in the response of tendon explants to stress deprivation is sex-dependent. Connect Tissue Res 2020; 61:48-62. [PMID: 31411079 PMCID: PMC6884684 DOI: 10.1080/03008207.2019.1648444] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose of the Study: The incidence of tendon injuries increases dramatically with age, which presents a major clinical burden. While previous studies have sought to identify age-related changes in extracellular matrix structure and function, few have been able to explain fully why aged tissues are more prone to degeneration and injury. In addition, recent studies have also demonstrated that age-related processes in humans may be sex-dependent, which could be responsible for muddled conclusions in changes with age. In this study, we investigate short-term responses through an ex vivo explant culture model of stress deprivation that specifically questions how age and sex differentially affect the ability of tendons to respond to altered mechanical stimulus.Materials and Methods: We subjected murine flexor explants from young (4 months of age) and aged (22-24 months of age) male and female mice to stress-deprived culture conditions for up to 1 week and investigated changes in viability, cell metabolism and proliferation, matrix biosynthesis and composition, gene expression, and inflammatory responses throughout the culture period.Results and Conclusions: We found that aging did have a significant influence on the response to stress deprivation, demonstrating that aged explants have a less robust response overall with reduced metabolic activity, viability, proliferation, and biosynthesis. However, age-related changes appeared to be sex-dependent. Together, this work demonstrates that the aging process and the subsequent effect of age on the ability of tendons to respond to stress-deprivation are inherently different based on sex, where male explants favor increased activity, apoptosis, and matrix remodeling while female explants favor reduced activity and tissue preservation.
Collapse
Affiliation(s)
- Brianne K. Connizzo
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Correspondence: Brianne K. Connizzo, 70 Massachusetts Avenue, NE47-377, Cambridge, MA 02139, T: 617-253-2469,
| | - Judith M. Piet
- Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Sandra J. Shefelbine
- Department of Bioengineering, Northeastern University, Boston, MA 02115, United States,Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, United States
| | - Alan J. Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| |
Collapse
|
20
|
Sudirman S, Chang HW, Chen CK, Kong ZL. A dietary polysaccharide from Eucheuma cottonii downregulates proinflammatory cytokines and ameliorates osteoarthritis-associated cartilage degradation in obese rats. Food Funct 2019; 10:5697-5706. [PMID: 31435629 DOI: 10.1039/c9fo01342c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Osteoarthritis (OA) is a common form of arthritis, which is characterized by the degeneration of articular cartilage, leading to joint dysfunction. Oral drug therapy seems to ameliorate some signs and symptoms of OA, but may be accompanied by side effects and does not appear to be effective long-term. Seaweed has received much attention for pharmacological application due to its various biomedical properties, including the anti-inflammation, antitumor, and antioxidant effects. This study investigated the ameliorative effects of a dietary polysaccharide from Eucheuma cottonii extract (ECE) on an anterior cruciate ligament transection with partial medial meniscectomy surgery (ACLT+MMx) to induce OA in high-fat diet (HFD)-induced obese rats. Male Sprague-Dawley rats were fed an HFD for 12 weeks before ACLT+MMx surgery, after which they were administered a daily oral gavage of saline (Sham, OB Sham, and OBOA) and either low-dose ECE (100 mg per kg body weight), high-dose ECE (400 mg per kg body weight), or glucosamine sulfate as a positive control (OBOAGS; 200 mg per kg body weight) for 5 weeks. Treatment with ECE decreased the body weight, triglyceride and total cholesterol (TC) levels, and the TC/high-density lipoprotein (HDL)-C ratio in the obese rats. Additionally, ECE downregulated the expression of proinflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, and leptin, and suppressed nuclear factor-kappa B and extracellular-signal-regulated kinase-1/2 expression, resulting in a decrease in the levels of matrix metalloproteinase (MMP)-1 and MMP-13 and prostaglandin-E2 and attenuated cartilage degradation. These results demonstrate that the dietary polysaccharide from ECE can suppress OA development in obese rats, suggesting its potential efficacy as a promising candidate for OA treatment.
Collapse
Affiliation(s)
- Sabri Sudirman
- Department of Food Science, National Taiwan Ocean University, Keelung City, Taiwan.
| | - Heng-Wei Chang
- Department of Food Science, National Taiwan Ocean University, Keelung City, Taiwan.
| | - Chun-Kai Chen
- Department of Food Science, National Taiwan Ocean University, Keelung City, Taiwan.
| | - Zwe-Ling Kong
- Department of Food Science, National Taiwan Ocean University, Keelung City, Taiwan.
| |
Collapse
|
21
|
Zhang T, Yao Y. Effects of inflammatory cytokines on bone/cartilage repair. J Cell Biochem 2019; 120:6841-6850. [PMID: 30335899 DOI: 10.1002/jcb.27953] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/02/2018] [Indexed: 01/24/2023]
Abstract
Many inflammatory factors can affect cell behaviors and work as a form of inter-regulatory networks through the inflammatory pathway. Inflammatory cytokines are critical for triggering bone regeneration after fracture or bone injury. Also, inflammatory cytokines play an important role in cartilage repair. The synergistic or antagonistic effects of both proinflammatory and anti-inflammatory cytokines have a great influence on fracture healing. This review discusses key inflammatory cytokines and signaling pathways involved in bone or cartilage repair.
Collapse
Affiliation(s)
- Tingshuai Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongchang Yao
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
22
|
Evasovic JM, Singer CA. Regulation of IL-17A and implications for TGF-β1 comodulation of airway smooth muscle remodeling in severe asthma. Am J Physiol Lung Cell Mol Physiol 2019; 316:L843-L868. [PMID: 30810068 PMCID: PMC6589583 DOI: 10.1152/ajplung.00416.2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/04/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
Severe asthma develops as a result of heightened, persistent symptoms that generally coincide with pronounced neutrophilic airway inflammation. In individuals with severe asthma, symptoms are poorly controlled by high-dose inhaled glucocorticoids and often lead to elevated morbidity and mortality rates that underscore the necessity for novel drug target identification that overcomes limitations in disease management. Many incidences of severe asthma are mechanistically associated with T helper 17 (TH17) cell-derived cytokines and immune factors that mediate neutrophilic influx to the airways. TH17-secreted interleukin-17A (IL-17A) is an independent risk factor for severe asthma that impacts airway smooth muscle (ASM) remodeling. TH17-derived cytokines and diverse immune mediators further interact with structural cells of the airway to induce pathophysiological processes that impact ASM functionality. Transforming growth factor-β1 (TGF-β1) is a pivotal mediator involved in airway remodeling that correlates with enhanced TH17 activity in individuals with severe asthma and is essential to TH17 differentiation and IL-17A production. IL-17A can also reciprocally enhance activation of TGF-β1 signaling pathways, whereas combined TH1/TH17 or TH2/TH17 immune responses may additively impact asthma severity. This review seeks to provide a comprehensive summary of cytokine-driven T cell fate determination and TH17-mediated airway inflammation. It will further review the evidence demonstrating the extent to which IL-17A interacts with various immune factors, specifically TGF-β1, to contribute to ASM remodeling and altered function in TH17-driven endotypes of severe asthma.
Collapse
Affiliation(s)
- Jon M Evasovic
- Department of Pharmacology, School of Medicine, University of Nevada , Reno, Nevada
| | - Cherie A Singer
- Department of Pharmacology, School of Medicine, University of Nevada , Reno, Nevada
| |
Collapse
|
23
|
Singh S, Kartha S, Bulka BA, Stiansen NS, Winkelstein BA. Physiologic facet capsule stretch can induce pain & upregulate matrix metalloproteinase-3 in the dorsal root ganglia when preceded by a physiological mechanical or nonpainful chemical exposure. Clin Biomech (Bristol, Avon) 2019; 64:122-130. [PMID: 29523370 PMCID: PMC6067996 DOI: 10.1016/j.clinbiomech.2018.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/22/2017] [Accepted: 01/15/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neck pain from cervical facet loading is common and induces inflammation and upregulation of nerve growth factor (NGF) that can sensitize the joint afferents. Yet, the mechanisms by which these occur and whether afferents can be pre-conditioned by certain nonpainful stimuli are unknown. This study tested the hypothesis that a nonpainful mechanical or chemical insult predisposes a facet joint to generate pain after a later exposure to typically nonpainful distraction. METHODS Rats were exposed to either a nonpainful distraction or an intra-articular subthreshold dose of NGF followed by a nonpainful distraction two days later. Mechanical hyperalgesia was measured daily and C6 dorsal root ganglia (DRG) tissue was assayed for NGF and matrix metalloproteinase-3 (MMP-3) expression on day 7. FINDINGS The second distraction increased joint displacement and strains compared to its first application (p = 0.0011). None of the initial exposures altered behavioral sensitivity in either of the groups being pre-conditioned or in controls; but, sensitivity was established in both groups receiving a second distraction within one day that lasted until day 7 (p < 0.024). NGF expression in the DRG was increased in both groups undergoing a pre-conditioning exposure (p < 0.0232). Similar findings were observed for MMP-3 expression, with a pre-conditioning exposure increasing levels after an otherwise nonpainful facet distraction. INTERPRETATION These findings suggest that nonpainful insults to the facet joint, when combined, can generate painful outcomes, possibly mediated by upregulation of MMP-3 and mature NGF.
Collapse
Affiliation(s)
- Sagar Singh
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Sonia Kartha
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Ben A Bulka
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Nicholas S Stiansen
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Beth A Winkelstein
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA; Department of Neurosurgery, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA.
| |
Collapse
|
24
|
Orozco GA, Tanska P, Florea C, Grodzinsky AJ, Korhonen RK. A novel mechanobiological model can predict how physiologically relevant dynamic loading causes proteoglycan loss in mechanically injured articular cartilage. Sci Rep 2018; 8:15599. [PMID: 30348953 PMCID: PMC6197240 DOI: 10.1038/s41598-018-33759-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/02/2018] [Indexed: 12/13/2022] Open
Abstract
Cartilage provides low-friction properties and plays an essential role in diarthrodial joints. A hydrated ground substance composed mainly of proteoglycans (PGs) and a fibrillar collagen network are the main constituents of cartilage. Unfortunately, traumatic joint loading can destroy this complex structure and produce lesions in tissue, leading later to changes in tissue composition and, ultimately, to post-traumatic osteoarthritis (PTOA). Consequently, the fixed charge density (FCD) of PGs may decrease near the lesion. However, the underlying mechanisms leading to these tissue changes are unknown. Here, knee cartilage disks from bovine calves were injuriously compressed, followed by a physiologically relevant dynamic compression for twelve days. FCD content at different follow-up time points was assessed using digital densitometry. A novel cartilage degeneration model was developed by implementing deviatoric and maximum shear strain, as well as fluid velocity controlled algorithms to simulate the FCD loss as a function of time. Predicted loss of FCD was quite uniform around the cartilage lesions when the degeneration algorithm was driven by the fluid velocity, while the deviatoric and shear strain driven mechanisms exhibited slightly discontinuous FCD loss around cracks. Our degeneration algorithm predictions fitted well with the FCD content measured from the experiments. The developed model could subsequently be applied for prediction of FCD depletion around different cartilage lesions and for suggesting optimal rehabilitation protocols.
Collapse
Affiliation(s)
- Gustavo A Orozco
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland.
| | - Petri Tanska
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Cristina Florea
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
- Departments of Biological Engineering, Electrical Engineering and Computer Science and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alan J Grodzinsky
- Departments of Biological Engineering, Electrical Engineering and Computer Science and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rami K Korhonen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
25
|
Connizzo BK, Grodzinsky AJ. Release of pro-inflammatory cytokines from muscle and bone causes tenocyte death in a novel rotator cuff in vitro explant culture model. Connect Tissue Res 2018; 59:423-436. [PMID: 29447021 PMCID: PMC6240787 DOI: 10.1080/03008207.2018.1439486] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Tendinopathy is a significant clinical problem thought to be associated with altered mechanical loading. Explant culture models allow researchers to alter mechanical loading in a controlled in vitro environment while maintaining tenocytes in their native matrix. However, current models do not accurately represent commonly injured tendons, ignoring contributions of associated musculature and bone, as well as regional collagen structure. This study details the characterization of amouse rotator cuff explant culture model, including bone, tendon, and muscle (BTM). MATERIALS AND METHODS Following harvest, BTM explants were maintained in stress-deprived culture for one week and tendon was then assessed for changes in cell viability, metabolism, matrix structure and content. RESULTS Matrix turnover occurred throughout culture as manifested in both gene expression and biosynthesis, but this did not translate to net changes in total collagen or sulfated glycosaminoglycan content. Furthermore, tendon structure was not significantly altered throughout culture. However, we found significant cell death in BTM tendons after 3 days in culture, which we hypothesize is cytokine-induced. Using a targeted multiplex assay, we found high levels of pro-inflammatory cytokines released to the culture medium from muscle and bone, levels that did cause cell deathin tendon-alone controls. CONCLUSIONS Overall, this model presents an innovative approach to understandingrotator cuff injury and tenocyte mechanobiology in a clinically-relevant tendon structure. Our model can be a powerful tool to investigate how mechanical and biological stimuli can alter normal tendon health and lead to tendon degeneration, and may provide a testbed for therapeutics for tendon repair.
Collapse
Affiliation(s)
- Brianne K. Connizzo
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Alan J. Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, United States,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| |
Collapse
|
26
|
Ravindra KC, Ahrens CC, Wang Y, Ramseier JY, Wishnok JS, Griffith LG, Grodzinsky AJ, Tannenbaum SR. Chemoproteomics of matrix metalloproteases in a model of cartilage degeneration suggests functional biomarkers associated with posttraumatic osteoarthritis. J Biol Chem 2018; 293:11459-11469. [PMID: 29794029 DOI: 10.1074/jbc.m117.818542] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 05/21/2018] [Indexed: 12/14/2022] Open
Abstract
Active matrix metalloproteases (MMPs) play a significant role in the pathogenesis of many diseases including osteoarthritis (OA), which involves progressive proteolytic degradation of cartilage. Clinical success of OA interventions that target MMPs has been limited by a lack of information about the presence and activity of specific disease-related proteases. We therefore developed a chemoproteomics approach based on MS to characterize the release and activity of MMPs in an in vitro model of the early inflammatory phase of posttraumatic OA (PTOA). We designed and synthesized chemical activity-based probes (ABPs) to identify active MMPs in bovine cartilage explants cultured for 30 days with the proinflammatory cytokine, interleukin-1α. Using these probes in an activity-based protein profiling-multidimensional identification technology (ABPP-MudPIT) approach, we identified active MMP-1, -2, -3, -7, -9, -12, and -13 in the medium after 10 days of culture, the time at which irreversible proteolysis of the collagen network in the explant was detected using proteolytic activation of FRET-quenched MMP substrates. Total MMP levels were quantified by shotgun proteomics, which, taken with ABPP-MudPIT data, indicated the presence of predominantly inactive MMPs in the culture medium. The selectivity of the ABPP-MudPIT approach was further validated by detection of specific endogenous MMPs activated de novo with 4-aminophenylmurcuric acetate. The utility of the new ABPP-MudPIT approach for detecting molecular biomarkers of PTOA disease initiation and potential targets for therapeutics motivates possible application in other diseases involving MMP activity.
Collapse
Affiliation(s)
- Kodihalli C Ravindra
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Caroline C Ahrens
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Yang Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Julie Y Ramseier
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - John S Wishnok
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Steven R Tannenbaum
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139.
| |
Collapse
|
27
|
Guilak F, Nims RJ, Dicks A, Wu CL, Meulenbelt I. Osteoarthritis as a disease of the cartilage pericellular matrix. Matrix Biol 2018; 71-72:40-50. [PMID: 29800616 DOI: 10.1016/j.matbio.2018.05.008] [Citation(s) in RCA: 275] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 01/16/2023]
Abstract
Osteoarthritis is a painful joint disease characterized by progressive degeneration of the articular cartilage as well as associated changes to the subchondral bone, synovium, and surrounding joint tissues. While the effects of osteoarthritis on the cartilage extracellular matrix (ECM) have been well recognized, it is now becoming apparent that in many cases, the onset of the disease may be initially reflected in the matrix region immediately surrounding the chondrocytes, termed the pericellular matrix (PCM). Growing evidence suggests that the PCM - which along with the enclosed chondrocytes are termed the "chondron" - acts as a critical transducer or "filter" of biochemical and biomechanical signals for the chondrocyte, serving to help regulate the homeostatic balance of chondrocyte metabolic activity in response to environmental signals. Indeed, it appears that alterations in PCM properties and cell-matrix interactions, secondary to genetic, epigenetic, metabolic, or biomechanical stimuli, could in fact serve as initiating or progressive factors for osteoarthritis. Here, we discuss recent advances in the understanding of the role of the PCM, with an emphasis on the reciprocity of changes that occur in this matrix region with disease, as well as how alterations in PCM properties could serve as a driver of ECM-based diseases such as osteoarthritis. Further study of the structure, function, and composition of the PCM in normal and diseased conditions may provide new insights into the understanding of the pathogenesis of osteoarthritis, and presumably new therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, Saint Louis, MO 63110, United States; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, United States; Department of Biomedical Engineering, Washington University, Saint Louis, MO 63110, United States.
| | - Robert J Nims
- Department of Orthopaedic Surgery, Washington University, Saint Louis, MO 63110, United States; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, United States
| | - Amanda Dicks
- Department of Orthopaedic Surgery, Washington University, Saint Louis, MO 63110, United States; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, United States; Department of Biomedical Engineering, Washington University, Saint Louis, MO 63110, United States
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University, Saint Louis, MO 63110, United States; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, United States
| | - Ingrid Meulenbelt
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
28
|
Pietrosimone B, Loeser RF, Blackburn JT, Padua DA, Harkey MS, Stanley LE, Luc-Harkey BA, Ulici V, Marshall SW, Jordan JM, Spang JT. Biochemical markers of cartilage metabolism are associated with walking biomechanics 6-months following anterior cruciate ligament reconstruction. J Orthop Res 2017; 35:2288-2297. [PMID: 28150869 PMCID: PMC5540809 DOI: 10.1002/jor.23534] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 01/23/2017] [Indexed: 02/06/2023]
Abstract
The purpose of our study was to determine the association between biomechanical outcomes of walking gait (peak vertical ground reaction force [vGRF], vGRF loading rate [vGRF-LR], and knee adduction moment [KAM]) 6 months following anterior cruciate ligament reconstruction (ACLR) and biochemical markers of serum type-II collagen turnover (collagen type-II cleavage product to collagen type-II C-propeptide [C2C:CPII]), plasma degenerative enzymes (matrix metalloproteinase-3 [MMP-3]), and a pro-inflammatory cytokine (interleukin-6 [IL-6]). Biochemical markers were evaluated within the first 2 weeks (6.5 ± 3.8 days) following ACL injury and again 6 months following ACLR in eighteen participants. All peak biomechanical outcomes were extracted from the first 50% of the stance phase of walking gait during a 6-month follow-up exam. Limb symmetry indices (LSI) were used to normalize the biomechanical outcomes in the ACLR limb to that of the contralateral limb (ACLR/contralateral). Bivariate correlations were used to assess associations between biomechanical and biochemical outcomes. Greater plasma MMP-3 concentrations after ACL injury and at the 6-month follow-up exam were associated with lesser KAM LSI. Lesser KAM was associated with greater plasma IL-6 at the 6-month follow-up exam. Similarly, lesser vGRF-LR LSI was associated with greater plasma MMP-3 concentrations at the 6-month follow-up exam. Lesser peak vGRF LSI was associated with higher C2C:CPII after ACL injury, yet this association was not significant after accounting for walking speed. Therefore, lesser biomechanical loading in the ACLR limb, compared to the contralateral limb, 6 months following ACLR may be related to deleterious joint tissue metabolism that could influence future cartilage breakdown. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2288-2297, 2017.
Collapse
Affiliation(s)
- Brian Pietrosimone
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States,Human Movement Science Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Richard F. Loeser
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, United States
| | - J. Troy Blackburn
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States,Human Movement Science Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Darin A. Padua
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States,Human Movement Science Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Matthew S. Harkey
- Human Movement Science Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Laura E. Stanley
- Human Movement Science Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Brittney A. Luc-Harkey
- Human Movement Science Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Veronica Ulici
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, United States
| | - Stephen W. Marshall
- Injury Prevention Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
| | - Joanne M. Jordan
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, United States
| | - Jeffery T. Spang
- Department of Orthopaedics, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, United States
| |
Collapse
|
29
|
Larsson S, Struglics A, Lohmander LS, Frobell R. Surgical reconstruction of ruptured anterior cruciate ligament prolongs trauma-induced increase of inflammatory cytokines in synovial fluid: an exploratory analysis in the KANON trial. Osteoarthritis Cartilage 2017; 25:1443-1451. [PMID: 28522220 DOI: 10.1016/j.joca.2017.05.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/18/2017] [Accepted: 05/09/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Prospectively monitor how treatment of acutely ruptured anterior cruciate ligament (ACL) affects biomarkers of inflammation and proteolytic degradation over 5 years. DESIGN We studied 119 subjects with acute ACL injury from the randomized controlled knee anterior cruciate ligament, non-surgical versus surgical treatment (KANON)-trial (Clinical trial ISRCTN 84752559) who had synovial fluid, serum and urine samples available from at least two out of six visits over 5 years after acute ACL rupture. All subjects followed a similar rehabilitation protocol where, according to randomization, 60 also had early ACL reconstruction and 59 had the option to undergo a delayed ACL reconstruction if needed. Interleukin (IL)-6, IL-8, IL-10, interferon-gamma (IFNγ), tumor necrosis factor (TNF), amino acids alanine, arginine, glycine, serine (ARGS)-aggrecan, C-terminal crosslinking telopeptide type II collagen (CTX-II) and N-terminal crosslinking telopeptide type I collagen (NTX-I) were quantified by enzyme-linked immunosorbent assays (ELISA). RESULTS Subjects randomized to early ACL reconstruction had higher cytokine concentrations in index knee synovial fluid at 4 months (IL-6, IL-8, IL-10, TNF), 8 months (IL-6 and TNF) and at 5 years (IFNγ) compared to those randomized to optional delayed reconstruction. Those that underwent delayed ACL reconstruction within 5 years (30 subjects), had higher synovial fluid concentrations of IL-6 at 5 years compared to those treated with rehabilitation alone. No differences between groups were noted for ARGS-aggrecan in synovial fluid and serum or CTX-II and NTX-I in urine over 5 years, neither as randomized nor as treated. CONCLUSIONS Surgical ACL reconstruction constitutes a second trauma to the acutely injured joint resulting in a prolonged elevation of already high synovial fluid levels of inflammatory cytokines.
Collapse
Affiliation(s)
- S Larsson
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund, Sweden.
| | - A Struglics
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund, Sweden.
| | - L S Lohmander
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund, Sweden.
| | - R Frobell
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund, Sweden.
| |
Collapse
|
30
|
Behrendt P, Häfelein K, Preusse-Prange A, Bayer A, Seekamp A, Kurz B. IL-10 ameliorates TNF-α induced meniscus degeneration in mature meniscal tissue in vitro. BMC Musculoskelet Disord 2017; 18:197. [PMID: 28511649 PMCID: PMC5434535 DOI: 10.1186/s12891-017-1561-x] [Citation(s) in RCA: 9] [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: 01/18/2017] [Accepted: 05/09/2017] [Indexed: 12/19/2022] Open
Abstract
Background Joint inflammation causes meniscus degeneration and can exacerbate post-traumatic meniscus injuries by extracellular matrix degradation, cellular de-differentiation and cell death. The aim of this study was to examine whether anti-inflammatory interleukin-10 exerts protective effects in an in vitro model of TNF-α-induced meniscus degeneration. Methods Meniscus tissue was harvested from the knees of adult cows. After 24 h of equilibrium explants were simultaneously treated with bovine TNF-α and IL-10. After an incubation time of 72 h cell death was measured histomorphometrically (nuclear blebbing, NB) and release of glycosaminoglycans (GAG, DMMB assay) and nitric oxide (NO, Griess-reagent) were analysed. Transcription levels (mRNA) of matrix degrading enzymes, collagen type X (COL10A1) and nitric oxide synthetase 2 (NOS2) were measured by quantitative real time PCR. TNF-α-dependent formation of the aggrecanase-specific aggrecan neoepitope NITEGE was visualised by immunostaining. Differences between groups were calculated using a one-way ANOVA with a Bonferroni post hoc test. Results Administration of IL-10 significantly prevented the TNF-α-related cell death (P .001), release of NO (P .003) and NOS2 expression (P .04). Release of GAG fragments (P .001), NITEGE formation and expression of MMP3 (P .007), -13 (P .02) and ADAMTS4 (P .001) were significantly reduced. The TNF-α-dependent increase in COL10A1 expression was also antagonized by IL-10 (P .02). Conclusion IL-10 prevented crucial mechanisms of meniscal degeneration induced by a key cytokine of OA, TNF-α. Administration of IL-10 might improve the biological regeneration and provide a treatment approach in degenerative meniscus injuries and in conditions of post-traumatic sports injuries.
Collapse
Affiliation(s)
- P Behrendt
- Department of Orthopaedics and Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| | - K Häfelein
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany
| | - A Preusse-Prange
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany
| | - A Bayer
- Department of Cardiovascular Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - A Seekamp
- Department of Orthopaedics and Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - B Kurz
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany
| |
Collapse
|
31
|
Grodzinsky AJ, Wang Y, Kakar S, Vrahas MS, Evans CH. Intra-articular dexamethasone to inhibit the development of post-traumatic osteoarthritis. J Orthop Res 2017; 35:406-411. [PMID: 27176565 PMCID: PMC5604325 DOI: 10.1002/jor.23295] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 05/06/2016] [Indexed: 02/04/2023]
Abstract
UNLABELLED Injury to the joint provokes a number of local pathophysiological changes, including synthesis of inflammatory cytokines, death of chondrocytes, breakdown of the extra-cellular matrix of cartilage, and reduced synthesis of matrix macromolecules. These processes combine to engender the subsequent development of post-traumatic osteoarthritis (PTOA). To prevent this from happening, it is necessary to inhibit these disparate responses to injury; given their heterogeneity, this is challenging. However, dexamethasone has the necessary pleiotropic properties required of a drug for this purpose. Using in vitro models, we have shown that low doses of dexamethasone sustain the synthesis of cartilage proteoglycans while inhibiting their breakdown after injurious compression in the presence or absence of inflammatory cytokines. Under these conditions, dexamethasone is non-toxic and maintains the viability of chondrocytes exposed chronically to such cytokines as interleukin (IL) -1, IL-6, and tumor necrosis factor-α. Moreover, the anti-inflammatory properties of dexamethasone have been appreciated for decades. In view of this information, we have initiated a pilot clinical study to determine whether a single, intra-articular injection of dexamethasone into the wrist shows promise in preventing PTOA after intra-articular fracture of the distal radius. CLINICAL SIGNIFICANCE Suppressing the various etiopathophysiological responses to injury in the joint is an attractive strategy for lowering the clinical burden of PTOA. The intra-articular administration of dexamethasone soon after injury offers a simple and inexpensive means of accomplishing this. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:406-411, 2017.
Collapse
Affiliation(s)
- Alan J. Grodzinsky
- Departments of Biological, Electrical and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Yang Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Sanjeev Kakar
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, MN
| | - Mark S. Vrahas
- Department of Orthopaedic Surgery, Harvard Medical School
| | | |
Collapse
|
32
|
Desando G, Bartolotti I, Vannini F, Cavallo C, Castagnini F, Buda R, Giannini S, Mosca M, Mariani E, Grigolo B. Repair Potential of Matrix-Induced Bone Marrow Aspirate Concentrate and Matrix-Induced Autologous Chondrocyte Implantation for Talar Osteochondral Repair: Patterns of Some Catabolic, Inflammatory, and Pain Mediators. Cartilage 2017; 8:50-60. [PMID: 27994720 PMCID: PMC5154420 DOI: 10.1177/1947603516642573] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE The low regenerative potential of cartilage contributed to the development of different cell therapies aimed to improve the clinical outcome in young patients with Osteochondral Lesions of the Talus (OLT). This study is designed to assess the regenerative potential of autologous matrix-induced Bone Marrow Aspirate Concentrate (mBMAC) and matrix-induced Autologous Chondrocyte Implantation (mACI) evaluating, on a small number of osteochondral biopsies, the expression of some catabolic, inflammatory, and pain mediators. DESIGN Twenty-two patients with OLT were analyzed in this study; 7 were treated with mACI and 15 with mBMAC. Informed consent was obtained from all the patients. Clinical assessments were performed pre-operatively and at 12, 24, and 36 months after surgery using the American Orthopedic Foot and Ankle Society (AOFAS). Histology and immunohistochemistry were used to assess cartilage repair at 24 months. Data were analyzed using non-parametric Wilcoxon-Mann-Whitney and Spearman tests. RESULTS A remarkable improvement in AOFAS score was noticed for both treatments up to 36 months; however, patients treated with mACI reported the best AOFAS score. Various degrees of tissue remodeling were observed by histological analysis for both cell strategies. However, mBMAC treatment showed a higher expression of some fibrous and hypertrophic markers compared to mACI group. A mild positivity for nerve growth factor, as pain mediator, was noticed for both treatments.M. CONCLUSIONS Our findings demonstrated the best histological and clinical results following mACI treatment since different fibrotic and hypertrophic features were evident in the mBMAC group at 24-month follow-up.
Collapse
Affiliation(s)
| | - Isabella Bartolotti
- Laboratory of Immunorheumatology and Tissue Regeneration, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Francesca Vannini
- 1Clinic of Orthopaedics and Traumatology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Carola Cavallo
- Laboratory RAMSES, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Francesco Castagnini
- 1Clinic of Orthopaedics and Traumatology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Roberto Buda
- 1Clinic of Orthopaedics and Traumatology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Sandro Giannini
- 1Clinic of Orthopaedics and Traumatology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Massimiliano Mosca
- 1Clinic of Orthopaedics and Traumatology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Erminia Mariani
- Laboratory of Immunorheumatology and Tissue Regeneration, Rizzoli Orthopedic Institute, Bologna, Italy; Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Brunella Grigolo
- Laboratory RAMSES, Rizzoli Orthopedic Institute, Bologna, Italy; Laboratory of Immunorheumatology and Tissue Regeneration, Rizzoli Orthopedic Institute, Bologna, Italy
| |
Collapse
|
33
|
Quantitative proteomics analysis of cartilage response to mechanical injury and cytokine treatment. Matrix Biol 2016; 63:11-22. [PMID: 27988350 DOI: 10.1016/j.matbio.2016.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/09/2016] [Accepted: 12/09/2016] [Indexed: 01/15/2023]
Abstract
Mechanical damage at the time of joint injury and the ensuing inflammatory response associated with elevated levels of pro-inflammatory cytokines in the synovial fluid, are reported to contribute to the progression to osteoarthritis after injury. In this exploratory study, we used a targeted proteomics approach to follow the progression of matrix degradation in response to mechanical damage and cytokine treatment of human knee cartilage explants, and thereby to study potential molecular biomarkers. This proteomics approach allowed us to unambiguously identify and quantify multiple peptides and proteins in the cartilage medium and explants upon treatment with ±injurious compression ±cytokines, treatments that mimic the earliest events in post-traumatic OA. We followed degradation of different protein domains, e.g., G1/G2/G3 of aggrecan, by measuring representative peptides of matrix proteins released into the medium at 7 time points throughout the 21-day culture period. COMP neo-epitopes, which were previously identified in the synovial fluid of knee injury/OA patients, were also released by these human cartilage explants treated with cyt and cyt+inj. The absence of collagen pro-peptides and elevated levels of specific COMP and COL3A1 neo-epitopes after human knee trauma may be relevant as potential biomarkers for post-traumatic OA. This model system thereby enables study of the kinetics of cartilage degradation and the identification of biomarkers within cartilage explants and those released to culture medium. Discovery proteomics revealed that candidate proteases were identified after specific treatment conditions, including MMP1, MMP-3, MMP-10 and MMP-13.
Collapse
|
34
|
Behrendt P, Preusse-Prange A, Klüter T, Haake M, Rolauffs B, Grodzinsky AJ, Lippross S, Kurz B. IL-10 reduces apoptosis and extracellular matrix degradation after injurious compression of mature articular cartilage. Osteoarthritis Cartilage 2016; 24:1981-1988. [PMID: 27349464 DOI: 10.1016/j.joca.2016.06.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/25/2016] [Accepted: 06/13/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim of this study was to examine whether anti-inflammatory interleukin-10 (IL-10) exerts chondroprotective effects in an in vitro model of a single mechanical injury of mature articular cartilage. METHOD Articular cartilage was harvested from the femoro-patellar groove of adult cows (Bos taurus) and cultured w/o bovine IL-10. After 24 h of equilibration explants were subjected to an axial unconfined compression (50% strain, velocity 2 mm/s, held for 10 s). After 96 h cell death was measured histomorphometrically (nuclear blebbing, NB) and the release of glycosaminoglycans (GAG, DMMB assay) and nitric oxide (NO, Griess-reagent) were analyzed. mRNA levels of matrix degrading enzymes and nitric oxide synthetase were measured by quantitative real time PCR. Differences between groups were calculated using a one-way ANOVA with a Bonferroni post hoc test. RESULTS Injurious compression significantly increased the number of cells with NB, release of GAG and nitric oxide and expression of MMP-3, -13, ADAMTS-4 and NOS2. Administration of IL-10 significantly reduced the injury related cell death and release of GAG and NO, respectively. Expression of MMP-3, -13, ADAMTS-4 and NOS2 were significantly reduced. CONCLUSION Joint injury is a complex process involving specific mechanical effects on cartilage as well as induction of an inflammatory environment. IL-10 prevented crucial mechanisms of chondrodegeneration induced by an injurious single compression. IL-10 might be a multipurpose drug candidate for the treatment of cartilage-related sports injuries or osteoarthritis (OA).
Collapse
Affiliation(s)
- P Behrendt
- Department of Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Germany.
| | - A Preusse-Prange
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany.
| | - T Klüter
- Department of Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Germany.
| | - M Haake
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany.
| | - B Rolauffs
- Siegfried Weller Institute for Trauma Research & Clinic for Trauma and Restorative Surgery, BG Trauma Clinic Tuebingen, University of Tuebingen, Tuebingen, Germany; Department of Orthopedics and Trauma Surgery, Albert Ludwigs University of Freiburg, Freiburg, Germany.
| | - A J Grodzinsky
- Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - S Lippross
- Department of Trauma Surgery, University Medical Center Schleswig-Holstein, Campus Kiel, Germany.
| | - B Kurz
- Institute of Anatomy, Christian Albrechts-University, Kiel, Germany.
| |
Collapse
|
35
|
Ciuffa R, Caron E, Leitner A, Uliana F, Gstaiger M, Aebersold R. Contribution of Mass Spectrometry-Based Proteomics to the Understanding of TNF-α Signaling. J Proteome Res 2016; 16:14-33. [PMID: 27762135 DOI: 10.1021/acs.jproteome.6b00728] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
NF-κB is a family of ubiquitous dimeric transcription factors that play a role in a myriad of cellular processes, ranging from differentiation to stress response and immunity. In inflammation, activation of NF-κB is mediated by pro-inflammatory cytokines, in particular the prototypic cytokines IL-1β and TNF-α, which trigger the activation of complex signaling cascades. In spite of decades of research, the system level understanding of TNF-α signaling is still incomplete. This is partially due to the limited knowledge at the proteome level. The objective of this review is to summarize and critically evaluate the current status of the proteomic research on TNF-α signaling. We will discuss the merits and flaws of the existing studies as well as the insights that they have generated into the proteomic landscape and architecture connected to this signaling pathway. Besides delineating past and current trends in TNF-α proteomic research, we will identify research directions and new methodologies that can further contribute to characterize the TNF-α associated proteome in space and time.
Collapse
Affiliation(s)
- Rodolfo Ciuffa
- Institute of Molecular Systems Biology, ETH Zurich , 8093 Zurich, Switzerland
| | - Etienne Caron
- Institute of Molecular Systems Biology, ETH Zurich , 8093 Zurich, Switzerland
| | - Alexander Leitner
- Institute of Molecular Systems Biology, ETH Zurich , 8093 Zurich, Switzerland
| | - Federico Uliana
- Institute of Molecular Systems Biology, ETH Zurich , 8093 Zurich, Switzerland
| | - Matthias Gstaiger
- Institute of Molecular Systems Biology, ETH Zurich , 8093 Zurich, Switzerland
| | - Ruedi Aebersold
- Institute of Molecular Systems Biology, ETH Zurich , 8093 Zurich, Switzerland.,Faculty of Science, University of Zurich , 8006 Zurich, Switzerland
| |
Collapse
|
36
|
Melin Fürst C, Åhrman E, Bratteby K, Waldemarson S, Malmström J, Blom AM. Quantitative Mass Spectrometry To Study Inflammatory Cartilage Degradation and Resulting Interactions with the Complement System. THE JOURNAL OF IMMUNOLOGY 2016; 197:3415-3424. [PMID: 27630166 DOI: 10.4049/jimmunol.1601006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/14/2016] [Indexed: 12/25/2022]
Abstract
Joint diseases are often characterized by inflammatory processes that result in pathological changes in joint tissues, including cartilage degradation and release of components into the synovial fluid. The complement system plays a central role in promoting the inflammation. Because several cartilage proteins are known to interact with complement, causing either activation or inhibition of the system, we aimed to investigate these interactions comprehensively. Bovine cartilage explants were cultured with IL-1α to induce cartilage degradation, followed by incubation with human serum. Label-free selected reaction monitoring mass spectrometry was used to specifically quantify complement proteins interacting with the cartilage explant. In parallel, the time-dependent degradation of cartilage was detected using mass spectrometry analysis (liquid chromatography-tandem mass spectrometry). Complement proteins resulting from activation of the classical, alternative, and terminal pathways were detected on IL-1α-stimulated cartilage at time points when clear alterations in extracellular matrix composition had occurred. Increased levels of the complement activation product C4d, as detected by ELISA in serum after incubation with IL-1α-stimulated cartilage, confirmed the selected reaction monitoring results indicating complement activation. Further, typical activated (cleaved) C3 fragments were detected by Western blotting in extracts of IL-1α-stimulated cartilage. No complement activation was triggered by cartilage cultured in the absence of IL-1α. Components released from IL-1α-stimulated cartilage during culture had an inhibitory effect on complement activation. These were released after a longer incubation period with IL-1α and may represent a feedback reaction to cartilage-triggered complement activation observed after a shorter incubation period.
Collapse
Affiliation(s)
- Camilla Melin Fürst
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, S-205 02 Malmö, Sweden
| | - Emma Åhrman
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, S-221 84 Lund, Sweden; and
| | - Klas Bratteby
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, S-205 02 Malmö, Sweden
| | - Sofia Waldemarson
- Department of Immunotechnology, Lund University, S-223 81 Lund, Sweden
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, S-221 84 Lund, Sweden; and
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, S-205 02 Malmö, Sweden;
| |
Collapse
|
37
|
Brand FJ, Forouzandeh M, Kaur H, Travascio F, de Rivero Vaccari JP. Acidification changes affect the inflammasome in human nucleus pulposus cells. JOURNAL OF INFLAMMATION-LONDON 2016; 13:29. [PMID: 27563282 PMCID: PMC4997758 DOI: 10.1186/s12950-016-0137-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/17/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Interleukin (IL)-1β is involved in the pathology of intervertebral disc degeneration. Under normal conditions, IL-1β is present in cells in an inactive form (pro-IL-1β). However, under pathological conditions, pro-IL-1β is turned into its active form (IL-1β) by the inflammasome, a multi-protein complex of the innate immune response that activates caspase-1. Under conditions of degeneration, the disc experiences an environment of increased acidification. However, the implications of acidification on the innate immune response remain poorly explored. METHODS Here we have studied how pH changes in human nucleus pulposus cells affect inflammasome activation by immunoblot analysis of protein lysates obtained from nucleus pulposus cells that were exposed to different pH levels in culture. RESULTS In this study, we have found that in nucleus pulposus cells, with increased acidification, there was a decrease in inflammasome activation consistent with lower levels of active IL-1β. However, this effect at a pH of 6.5, the lowest pH level tested, was abrogated when cells were treated with IL-1β. CONCLUSIONS Taken together, these findings suggest that the inflammatory response through IL-1β experienced by the human disc is not initiated in nucleus pulposus cells when the stimulus is acidification.
Collapse
Affiliation(s)
- Frank J Brand
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136 USA
| | - Mahtab Forouzandeh
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136 USA
| | - Harmanpreet Kaur
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136 USA
| | - Francesco Travascio
- Biomechanics Research Laboratory, Department of Industrial Engineering, University of Miami, Coral Gables, FL 33146 USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL 33136 USA ; Department of Neurological Surgery, Lois Pope LIFE Center, 1095 NW 14th Terrace, 3-25JJ, Miami, FL 33136-1060 USA
| |
Collapse
|
38
|
Madej W, Buma P, van der Kraan P. Inflammatory conditions partly impair the mechanically mediated activation of Smad2/3 signaling in articular cartilage. Arthritis Res Ther 2016; 18:146. [PMID: 27334538 PMCID: PMC4918093 DOI: 10.1186/s13075-016-1038-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/31/2016] [Indexed: 01/14/2023] Open
Abstract
Background Joint trauma, which is frequently related with mechanical overloading of articular cartilage, is a well-established risk for osteoarthritis (OA) development. Additionally, reports show that trauma leads to synovial joint inflammation. In consequence, after joint trauma, cartilage is influenced by deleterious excessive loading combined with the catabolic activity of proinflammatory mediators. Since the activation of TGF-β signaling by loading is considered to be a key regulatory pathway for maintaining cartilage homeostasis, we tested the effect of proinflammatory conditions on mechanically mediated activation of TGF-β/Smad2/3P signaling in cartilage. Methods Cartilage explants were subjected to dynamic mechanical compression in the presence of interleukin-1 beta (IL-1β) or osteoarthritic synovium-conditioned medium (OAS-CM). Subsequently, the activation of the Smad2/3P pathway was monitored with QPCR analysis of reporter genes and additionally the expression of receptors activating the Smad2/3P pathway was analyzed. Finally, the ability for mechanically mediated activation of Smad2/3P was tested in human OA cartilage. Results IL-1β presence during compression did not impair the upregulation of Smad2/3P reporter genes, however the results were affected by IL-1β-mediated upregulations in unloaded controls. OAS-CM significantly impaired the compression-mediated upregulation of bSmad7 and Tgbfb1. IL-1β suppressed the compression-mediated bAlk5 upregulation where 12 MPa compression applied in the presence of OAS-CM downregulated the bTgfbr2. Mechanically driven upregulation of Smad2/3P reporter genes was present in OA cartilage. Conclusions Proinflammatory conditions partly impair the mechanically mediated activation of the protective TGF-β/Smad2/3P pathway. Additionally, the excessive mechanical compression, applied in the presence of proinflammatory conditions diminishes the expression of the type II TGF-β receptor, a receptor critical for maintenance of articular cartilage.
Collapse
Affiliation(s)
- Wojciech Madej
- Orthopedic Research Laboratory, Radboud University Medical Center, Geert Grooteplein-Zuid 10, route 547, 6525, GA, Nijmegen, The Netherlands.,Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 26-28, route 272, 6525, GA, Nijmegen, The Netherlands
| | - Pieter Buma
- Orthopedic Research Laboratory, Radboud University Medical Center, Geert Grooteplein-Zuid 10, route 547, 6525, GA, Nijmegen, The Netherlands
| | - Peter van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 26-28, route 272, 6525, GA, Nijmegen, The Netherlands.
| |
Collapse
|
39
|
Wilson R, Golub SB, Rowley L, Angelucci C, Karpievitch YV, Bateman JF, Fosang AJ. Novel Elements of the Chondrocyte Stress Response Identified Using an in Vitro Model of Mouse Cartilage Degradation. J Proteome Res 2016; 15:1033-50. [PMID: 26794603 DOI: 10.1021/acs.jproteome.5b01115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The destruction of articular cartilage in osteoarthritis involves chondrocyte dysfunction and imbalanced extracellular matrix (ECM) homeostasis. Pro-inflammatory cytokines such as interleukin-1α (IL-1α) contribute to osteoarthritis pathophysiology, but the effects of IL-1α on chondrocytes within their tissue microenvironment have not been fully evaluated. To redress this we used label-free quantitative proteomics to analyze the chondrocyte response to IL-1α within a native cartilage ECM. Mouse femoral heads were cultured with and without IL-1α, and both the tissue proteome and proteins released into the media were analyzed. New elements of the chondrocyte response to IL-1α related to cellular stress included markers for protein misfolding (Armet, Creld2, and Hyou1), enzymes involved in glutathione biosynthesis and regeneration (Gstp1, Gsto1, and Gsr), and oxidative stress proteins (Prdx2, Txn, Atox1, Hmox1, and Vnn1). Other proteins previously not associated with the IL-1α response in cartilage included ECM components (Smoc2, Kera, and Crispld1) and cysteine proteases (cathepsin Z and legumain), while chondroadherin and cartilage-derived C-type lectin (Clec3a) were identified as novel products of IL-1α-induced cartilage degradation. This first proteome-level view of the cartilage IL-1α response identified candidate biomarkers of cartilage destruction and novel targets for therapeutic intervention in osteoarthritis.
Collapse
Affiliation(s)
- Richard Wilson
- Central Science Laboratory, University of Tasmania , Hobart, Tasmania 7001, Australia.,Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia
| | - Suzanne B Golub
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia.,Department of Pediatrics, University of Melbourne , Parkville, Victoria 3052, Australia
| | - Lynn Rowley
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia
| | - Constanza Angelucci
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia
| | - Yuliya V Karpievitch
- School of Physical Sciences, University of Tasmania , Hobart, Tasmania 7001, Australia.,Centre of Excellence in Plant Energy Biology, University of Western Australia and Harry Perkins Institute of Medical Research , Perth, Western Australia 6009, Australia
| | - John F Bateman
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne , Parkville, Victoria 3052, Australia
| | - Amanda J Fosang
- Murdoch Childrens Research Institute, Royal Children's Hospital , Parkville, Melbourne, Victoria 3052, Australia.,Department of Pediatrics, University of Melbourne , Parkville, Victoria 3052, Australia
| |
Collapse
|
40
|
Tan AR, VandenBerg CD, Attur M, Abramson SB, Knight MM, Bulinski JC, Ateshian GA, Cook JL, Hung CT. Cytokine preconditioning of engineered cartilage provides protection against interleukin-1 insult. Arthritis Res Ther 2015; 17:361. [PMID: 26667364 PMCID: PMC4704536 DOI: 10.1186/s13075-015-0876-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/26/2015] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND During osteoarthritis and following surgical procedures, the environment of the knee is rich in proinflammatory cytokines such as IL-1. Introduction of tissue-engineered cartilage constructs to a chemically harsh milieu may limit the functionality of the implanted tissue over long periods. A chemical preconditioning scheme (application of low doses of IL-1) was tested as a method to prepare developing engineered tissue to withstand exposure to a higher concentration of the cytokine, known to elicit proteolysis as well as rapid degeneration of cartilage. METHODS Using an established juvenile bovine model system, engineered cartilage was preconditioned with low doses of IL-1α (0.1 ng/mL, 0.5 ng/mL, and 1.0 ng/mL) for 7 days before exposure to an insult dose (10 ng/mL). The time frame over which this protection is afforded was investigated by altering the amount of time between preconditioning and insult as well as the time following insult. To explore a potential mechanism for this protection, one set of constructs was preconditioned with CoCl2, a chemical inducer of hypoxia, before exposure to the IL-1α insult. Finally, we examined the translation of this preconditioning method to extend to clinically relevant adult, passaged chondrocytes from a preclinical canine model. RESULTS Low doses of IL-1α (0.1 ng/mL and 0.5 ng/mL) protected against subsequent catabolic degradation by cytokine insult, preserving mechanical stiffness and biochemical composition. Regardless of amount of time between preconditioning scheme and insult, protection was afforded. In a similar manner, preconditioning with CoCl2 similarly allowed for mediation of catabolic damage by IL-1α. The effects of preconditioning on clinically relevant adult, passaged chondrocytes from a preclinical canine model followed the same trends with low-dose IL-1β offering variable protection against insult. CONCLUSIONS Chemical preconditioning schemes have the ability to protect engineered cartilage constructs from IL-1-induced catabolic degradation, offering potential modalities for therapeutic treatments.
Collapse
Affiliation(s)
- Andrea R Tan
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, USA.
| | - Curtis D VandenBerg
- Department of Orthopedic Surgery, St. Luke's-Roosevelt Hospital Center, 1000 10th Avenue, New York, NY, USA.
| | - Mukundan Attur
- New York University Hospital for Joint Disease, 301 E. 17th Street, New York, NY, USA.
| | - Steven B Abramson
- New York University Hospital for Joint Disease, 301 E. 17th Street, New York, NY, USA.
| | - Martin M Knight
- Institute of Bioengineering and School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, London, UK.
| | - J Chloe Bulinski
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Avenue, New York, NY, USA.
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, USA.
- Department of Mechanical Engineering, Columbia University, 500 W. 120th Street, New York, NY, USA.
| | - James L Cook
- Comparative Orthopaedic Laboratory, University of Missouri, 1100 Virginia Avenue, Columbia, MO, USA.
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, USA.
| |
Collapse
|
41
|
Joos H, Leucht F, Riegger J, Hogrefe C, Fiedler J, Dürselen L, Reichel H, Ignatius A, Brenner RE. Differential Interactive Effects of Cartilage Traumatization and Blood Exposure In Vitro and In Vivo. Am J Sports Med 2015; 43:2822-32. [PMID: 26362437 DOI: 10.1177/0363546515602248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Sport injuries of the knee often lead to posttraumatic arthritis. In addition to direct damage of the cartilage, trauma-associated intra-articular bleeding may cause hemarthrosis. Both blood exposure and trauma are known to induce cell death and inflammation and to enhance proteoglycan release in cartilage. HYPOTHESIS Blood exposure increases chondrocyte death as well as inflammatory and degenerative processes in traumatized cartilage. STUDY DESIGN Controlled laboratory study. METHODS Human macroscopically intact osteoarthritic (OA) cartilage explants were impacted by a drop-tower system (0.59 J) and cultivated with or without 10% blood. Interactive effects were studied concerning cell survival, gene expression, and the release of mediators over 24 hours and 96 hours. To evaluate the effects of trauma and hemarthrosis in vivo, a newly established blunt cartilage trauma model in the rabbit was used. Treatment of the knee joints of mature New Zealand White rabbits consisted of the following groups: control (C), arthrotomy (A), arthrotomy with cartilage trauma (AT; 1.0 J), and arthrotomy with cartilage trauma and blood injection (ATH). After 1 and 12 weeks, inflammatory mediators in the synovial fluid and histological changes of the cartilage were determined, and immunohistological staining was performed. RESULTS The in vitro studies revealed a significant additional or synergistic effect of blood exposure on trauma-induced chondrocyte death, interleukin (IL)-1β and prostaglandin-E2 (PGE2) release, and matrix metalloproteinase (MMP)/pro-MMP level. Singular arthrotomy in vivo induced a temporary inflammation. Histologically, cartilage trauma caused significant OA changes that were not aggravated by an additional hemarthrosis. Trauma led to a persistent deposition of terminal complement complex (TCC), being enhanced by hemarthrosis. However, trauma-induced formation of osteophytes and arthrotomy-induced elevation of tumor necrosis factor-α release were reduced by hemarthrosis. CONCLUSION While blood exposure clearly aggravated trauma-induced OA processes in the in vitro model, a singular blood injection revealed heterogeneous effects in vivo, enhancing TCC deposition but reducing trauma-induced osteophyte formation while the histological score of traumatized cartilage was not further impaired. CLINICAL RELEVANCE The results of this study indicate that a singular, limited bleeding event might not exacerbate early trauma-induced cartilage degeneration in joint injuries. An early removal of intra-articular blood may not prevent the final resulting cartilage damage.
Collapse
Affiliation(s)
- Helga Joos
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Frank Leucht
- Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Cathrin Hogrefe
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Jörg Fiedler
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Lutz Dürselen
- Institute of Orthopedic Research and Biomechanics, University of Ulm, Ulm, Germany
| | - Heiko Reichel
- Department of Orthopedics, University of Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University of Ulm, Ulm, Germany
| | - Rolf E Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of Ulm, Ulm, Germany
| |
Collapse
|
42
|
Genemaras AA, Reiner T, Huang CY, Kaplan L. Early intervention with Interleukin-1 Receptor Antagonist Protein modulates catabolic microRNA and mRNA expression in cartilage after impact injury. Osteoarthritis Cartilage 2015; 23:2036-44. [PMID: 26521750 DOI: 10.1016/j.joca.2015.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 04/30/2015] [Accepted: 05/20/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The purpose of this controlled laboratory study was to determine the efficacy of Interleukin-1 Receptor Antagonist Protein (IRAP) treatment as an early intervention strategy by examining the changes in microRNA and mRNA expression in cartilage in an ex-vivo porcine knee joint impact model. METHODS Custom impact device was used to create replicable injury ex-vivo to intact porcine knee joint. Injury was caused by dropping a 10 kg weight one time from 1 m directly above the knee in extension. One hour after impact 20 μg/ml IRAP solution was intra-articularly injected. At 8 h post-injury, cartilage samples were harvested for cell viability and genetic expression analysis. Genetic expression of miR-27b, miR-140, miR-125b, ADAMTS-4, ADAMTS-5, MMP-3, IL-1β, and TNF-α were analyzed by RT-PCR. Cell viability image analysis was performed using ImageJ software. Groups were compared by analysis of variance (ANOVA) followed by Tukey's post-hoc test. A P-value <0.05 was considered significant. RESULTS At 8 h after IRAP treatment, expressions of ADAMTS-4, ADAMTS-5, MMP-3, IL-1β, and TNF-α in cartilage were significantly down-regulated from injury group (all P < 0.001). MiR-140, miR-125b, and miR-27b expressions were significantly up-regulated after treatment as compared to control and injury groups (all P < 0.001). CONCLUSION This study demonstrates that IRAP treatment administered during acute phase of cartilage impact injury increases expression levels of miR-140, miR-125b, and miR-27b in cartilage, indicating increased inhibition of their respective matrix-degrading enzymes. Clinically, these findings support the potential of IRAP treatment as an early intervention strategy for the prevention of cartilage degeneration after impact injury.
Collapse
Affiliation(s)
- A A Genemaras
- Department of Biomedical Engineering, College of Engineering, University of Miami, Coral Gables, FL, USA.
| | - T Reiner
- Geriatric Research, Education, and Clinical Center and Research Service, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL, USA.
| | - C-Y Huang
- Department of Biomedical Engineering, College of Engineering, University of Miami, Coral Gables, FL, USA.
| | - L Kaplan
- Department of Biomedical Engineering, College of Engineering, University of Miami, Coral Gables, FL, USA; Department of Orthopedics, Division of Sports Medicine, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
43
|
Torrero JI, Martínez C. New developments in the treatment of osteoarthritis - focus on biologic agents. Open Access Rheumatol 2015; 7:33-43. [PMID: 27790043 PMCID: PMC5045124 DOI: 10.2147/oarrr.s50058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common diseases around the world. Medical, social, and financial consequences oblige clinicians, surgeons, and researchers to focus on finding the best treatment option, to eradicate and stop this degenerative joint disease, in order to avoid surgical options which in many instances are over-indicated. Noninvasive treatments, such as anti-inflammatory drugs, physiotherapy, orthotic devices, dietary supplements, have demonstrated lack of effectiveness. The possibility to perform intra-articular injections with hyaluronic acid, corticosteroids, or the newest but criticized treatment based on platelet-rich plasma (PRP) has changed the management of OA disease. The use of PRP has led to many differences in treatment since there is a lack of consensus about protocols, indications, number of doses, cost-effectiveness, and duration of the treatment. Many publications have suggested efficacy in tendon injuries, but when PRP has been indicated to treat cartilage injuries, things are more inconsistent. Some authors have reported their experience treating OA with PRP, and it seems that, if well indicated, it is an option as a supplementary therapy. Therefore, we need to understand that OA is a mechanical disease which not only produces changes in radiographs, but also affects the quality of life. Pathogenesis of OA has been well explained, providing us new knowledge and future possibilities to improve the clinical approach. From basic science to surgery, there is a great field we all need to contribute to, because the general population is aging and total joint replacements should not be the only solution for OA. So herein is an actual review of the developments for treating OA with biologics, intended to be useful for the population inside orthopedics who could be called bio-orthopedists, since OA is a molecular homeostasis disbalance between catabolism and anabolism triggered by mechanical stress.
Collapse
Affiliation(s)
| | - Carlos Martínez
- University of Illinois Hospital and Health Sciences System, Chicago, IL, USA
| |
Collapse
|
44
|
Struglics A, Larsson S, Kumahashi N, Frobell R, Lohmander LS. Changes in Cytokines and Aggrecan ARGS Neoepitope in Synovial Fluid and Serum and in C-Terminal Crosslinking Telopeptide of Type II Collagen and N-Terminal Crosslinking Telopeptide of Type I Collagen in Urine Over Five Years After Anterior Cruciate Ligame. Arthritis Rheumatol 2015; 67:1816-25. [DOI: 10.1002/art.39146] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 03/31/2015] [Indexed: 01/12/2023]
Affiliation(s)
| | | | - Nobuyuki Kumahashi
- Lund University, Lund, Sweden, and Shimane University School of Medicine; Izumo Japan
| | | | - L. Stefan Lohmander
- Lund University, Lund, Sweden, and University of Southern Denmark; Odense Denmark
| |
Collapse
|
45
|
Walter BA, Likhitpanichkul M, Illien-Junger S, Roughley PJ, Hecht AC, Iatridis JC. TNFα transport induced by dynamic loading alters biomechanics of intact intervertebral discs. PLoS One 2015; 10:e0118358. [PMID: 25734788 PMCID: PMC4348425 DOI: 10.1371/journal.pone.0118358] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 12/19/2014] [Indexed: 12/18/2022] Open
Abstract
Objective Intervertebral disc (IVD) degeneration is an important contributor to the development of back pain, and a key factor relating pain and degeneration are the presence of pro-inflammatory cytokines and IVD motion. There is surprisingly limited understanding of how mechanics and inflammation interact in the IVD. This study investigated interactions between mechanical loading and pro-inflammatory cytokines in a large animal organ culture model to address fundamental questions regarding (i.) how inflammatory mediators arise within the IVD, (ii.) how long inflammatory mediators persist, and (iii.) how inflammatory mediators influence IVD biomechanics. Methods Bovine caudal IVDs were cultured for 6 or 20-days under static & dynamic loading with or without exogenous TNFα in the culture medium, simulating a consequence of inflammation of the surrounding spinal tissues. TNFα transport within the IVD was assessed via immunohistochemistry. Changes in IVD structural integrity (dimensions, histology & aggrecan degradation), biomechanical behavior (Creep, Recovery & Dynamic stiffness) and pro-inflammatory cytokines in the culture medium (ELISA) were assessed. Results TNFα was able to penetrate intact IVDs when subjected to dynamic loading but not static loading. Once transported within the IVD, pro-inflammatory mediators persisted for 4–8 days after TNFα removal. TNFα exposure induced changes in IVD biomechanics (reduced diurnal displacements & increased dynamic stiffness). Discussion This study demonstrated that exposure to TNFα, as might occur from injured surrounding tissues, can penetrate healthy intact IVDs, induce expression of additional pro-inflammatory cytokines and alter IVD mechanical behavior. We conclude that exposure to pro-inflammatory cytokine may be an initiating event in the progression of IVD degeneration in addition to being a consequence of disease.
Collapse
Affiliation(s)
- Benjamin A. Walter
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Biomedical Engineering, The City College of New York, New York, NY, United States of America
| | - Morakot Likhitpanichkul
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Svenja Illien-Junger
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | | | - Andrew C. Hecht
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - James C. Iatridis
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
46
|
Willard VP, Diekman BO, Sanchez-Adams J, Christoforou N, Leong KW, Guilak F. Use of cartilage derived from murine induced pluripotent stem cells for osteoarthritis drug screening. Arthritis Rheumatol 2015; 66:3062-72. [PMID: 25047145 DOI: 10.1002/art.38780] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 07/08/2014] [Indexed: 01/10/2023]
Abstract
OBJECTIVE The discovery of novel disease-modifying drugs for osteoarthritis (OA) is limited by the lack of adequate genetically defined cartilage tissues for application in high-throughput screening systems. We addressed this need by synthesizing cartilage from induced pluripotent stem cells (iPSCs) to establish and validate an in vitro model of OA. METHODS Native or iPSC-derived mouse cartilage samples were treated with the cytokine interleukin-1α (IL-1α) for 3 days to model the inflammatory environment of OA. The biochemical content, mechanical properties, and gene expression of the resulting tissues were assayed. In addition, the inflammatory and catabolic environment of the media was assessed. To establish high-throughput capability, we used a 96-well plate format and conducted a screen of previously identified candidate OA drugs. Glycosaminoglycan (GAG) release into the medium was used as the primary output for screening. RESULTS Treatment of iPSC-derived or native cartilage with IL-1α induced characteristic features of OA in a rapid and dose-dependent manner. In addition to the loss of GAGs and tissue mechanical properties, IL-1α treatment induced the expression of matrix metalloproteinases and increased the production of the inflammatory mediators nitric oxide and prostaglandin E2 . In the high-throughput screen validation, all candidate OA therapeutic agents provided some benefit, but only the NF-κB inhibitor SC514 effectively reduced cartilage loss in response to IL-1α. CONCLUSION This work demonstrates the utility of iPSCs for studying cartilage pathology and provides a platform for identifying novel, patient-specific therapeutic agents that prevent cartilage degradation and modify the course of OA development.
Collapse
|
47
|
Lourido L, Calamia V, Mateos J, Fernández-Puente P, Fernández-Tajes J, Blanco FJ, Ruiz-Romero C. Quantitative proteomic profiling of human articular cartilage degradation in osteoarthritis. J Proteome Res 2014; 13:6096-106. [PMID: 25383958 DOI: 10.1021/pr501024p] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Osteoarthritis (OA) is the most common rheumatic pathology and is characterized primarily by articular cartilage degradation. Despite its high prevalence, there is no effective therapy to slow disease progression or regenerate the damaged tissue. Therefore, new diagnostic and monitoring tests for OA are urgently needed, which would also promote the development of alternative therapeutic strategies. In the present study, we have performed an iTRAQ-based quantitative proteomic analysis of secretomes from healthy human articular cartilage explants, comparing their protein profile to those from unwounded (early disease) and wounded (advanced disease) zones of osteoarthritic tissue. This strategy allowed us to identify a panel of 76 proteins that are distinctively released by the diseased tissue. Clustering analysis allowed the classification of proteins according to their different profile of release from cartilage. Among these proteins, the altered release of osteoprotegerin (decreased in OA) and periostin (increased in OA), both involved in bone remodelling processes, was verified in further analyses. Moreover, periostin was also increased in the synovial fluid of OA patients. Altogether, the present work provides a novel insight into the mechanisms of human cartilage degradation and a number of new cartilage-characteristic proteins with possible biomarker value for early diagnosis and prognosis of OA.
Collapse
Affiliation(s)
- Lucía Lourido
- Proteomics Group-PBR2-ProteoRed/ISCIII, Rheumatology Division, §RIER-RED de Inflamación y Enfermedades Reumáticas, ∥CIBER-BBN Instituto de Salud Carlos III, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC) , As Xubias, 84, 15006-A Coruña, Spain
| | | | | | | | | | | | | |
Collapse
|
48
|
Synergy between Piezo1 and Piezo2 channels confers high-strain mechanosensitivity to articular cartilage. Proc Natl Acad Sci U S A 2014; 111:E5114-22. [PMID: 25385580 DOI: 10.1073/pnas.1414298111] [Citation(s) in RCA: 285] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Diarthrodial joints are essential for load bearing and locomotion. Physiologically, articular cartilage sustains millions of cycles of mechanical loading. Chondrocytes, the cells in cartilage, regulate their metabolic activities in response to mechanical loading. Pathological mechanical stress can lead to maladaptive cellular responses and subsequent cartilage degeneration. We sought to deconstruct chondrocyte mechanotransduction by identifying mechanosensitive ion channels functioning at injurious levels of strain. We detected robust expression of the recently identified mechanosensitive channels, PIEZO1 and PIEZO2. Combined directed expression of Piezo1 and -2 sustained potentiated mechanically induced Ca(2+) signals and electrical currents compared with single-Piezo expression. In primary articular chondrocytes, mechanically evoked Ca(2+) transients produced by atomic force microscopy were inhibited by GsMTx4, a PIEZO-blocking peptide, and by Piezo1- or Piezo2-specific siRNA. We complemented the cellular approach with an explant-cartilage injury model. GsMTx4 reduced chondrocyte death after mechanical injury, suggesting a possible therapy for reducing cartilage injury and posttraumatic osteoarthritis by attenuating Piezo-mediated cartilage mechanotransduction of injurious strains.
Collapse
|
49
|
Neu CP. Functional imaging in OA: role of imaging in the evaluation of tissue biomechanics. Osteoarthritis Cartilage 2014; 22:1349-59. [PMID: 25278049 PMCID: PMC4185127 DOI: 10.1016/j.joca.2014.05.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 05/06/2014] [Accepted: 05/17/2014] [Indexed: 02/02/2023]
Abstract
Functional imaging refers broadly to the visualization of organ or tissue physiology using medical image modalities. In load-bearing tissues of the body, including articular cartilage lining the bony ends of joints, changes in strain, stress, and material properties occur in osteoarthritis (OA), providing an opportunity to probe tissue function through the progression of the disease. Here, biomechanical measures in cartilage and related joint tissues are discussed as key imaging biomarkers in the evaluation of OA. Emphasis will be placed on the (1) potential of radiography, ultrasound, and magnetic resonance imaging to assess early tissue pathomechanics in OA, (2) relative utility of kinematic, structural, morphological, and biomechanical measures as functional imaging biomarkers, and (3) improved diagnostic specificity through the combination of multiple imaging biomarkers with unique contrasts, including elastography and quantitative assessments of tissue biochemistry. In comparison to other modalities, magnetic resonance imaging provides an extensive range of functional measures at the tissue level, with conventional and emerging techniques available to potentially to assess the spectrum of preclinical to advance OA.
Collapse
Affiliation(s)
- C P Neu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
50
|
Joos H, Wildner A, Hogrefe C, Reichel H, Brenner RE. Interleukin-1 beta and tumor necrosis factor alpha inhibit migration activity of chondrogenic progenitor cells from non-fibrillated osteoarthritic cartilage. Arthritis Res Ther 2014; 15:R119. [PMID: 24034344 PMCID: PMC3978440 DOI: 10.1186/ar4299] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 07/11/2013] [Accepted: 09/13/2013] [Indexed: 01/06/2023] Open
Abstract
Introduction The repair capability of traumatized articular cartilage is highly limited so that joint injuries often lead to osteoarthritis. Migratory chondrogenic progenitor cells (CPC) might represent a target cell population for in situ regeneration. This study aims to clarify, whether 1) CPC are present in regions of macroscopically intact cartilage from human osteoarthritic joints, 2) CPC migration is stimulated by single growth factors and the cocktail of factors released from traumatized cartilage and 3) CPC migration is influenced by cytokines present in traumatized joints. Methods We characterized the cells growing out from macroscopically intact human osteoarthritic cartilage using a panel of positive and negative surface markers and analyzed their differentiation capacity. The migratory response to platelet-derived growth factor (PDGF)-BB, insulin-like growth factor 1 (IGF-1), supernatants obtained from in vitro traumatized cartilage and interleukin-1 beta (IL-1β) as well as tumor necrosis factor alpha (TNF-α) were tested with a modified Boyden chamber assay. The influence of IL-1β and TNF-α was additionally examined by scratch assays and outgrowth experiments. Results A comparison of 25 quadruplicate marker combinations in CPC and bone-marrow derived mesenchymal stromal cells showed a similar expression profile. CPC cultures had the potential for adipogenic, osteogenic and chondrogenic differentiation. PDGF-BB and IGF-1, such as the supernatant from traumatized cartilage, induced a significant site-directed migratory response. IL-1β and TNF-α significantly reduced basal cell migration and abrogated the stimulative effect of the growth factors and the trauma supernatant. Both cytokines also inhibited cell migration in the scratch assay and primary outgrowth of CPC from cartilage tissue. In contrast, the cytokine IL-6, which is present in trauma supernatant, did not affect growth factor induced migration of CPC. Conclusion These results indicate that traumatized cartilage releases chemoattractive factors for CPC but IL-1β and TNF-α inhibit their migratory activity which might contribute to the low regenerative potential of cartilage in vivo.
Collapse
|